1
|
Dong Y, He G, Chen K, He X, Pan M, Huang X, Yu X, Xia J. Baicalein promotes KDM4E to induce BICD1 and inhibit triple-negative breast cancer progression by blocking PAR1 signaling. Mol Carcinog 2024; 63:1288-1302. [PMID: 38607237 DOI: 10.1002/mc.23724] [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: 12/11/2023] [Revised: 02/26/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024]
Abstract
Baicalein has been implicated in the chemotherapy overcoming triple-negative breast cancer (TNBC). However, many unanswered questions remain regarding its role in treating TNBC. Here, we sought to demonstrate the molecular pathway mediated by baicalein in TNBC. Lysine-specific demethylase 4E (KDM4E), reduced in TNBC cells, was identified as a target protein of baicalein, and baicalein enhanced the protein expression and stability of KDM4E in TNBC cells. Knockdown of KDM4E attenuated the inhibitory effect of baicalein on TNBC cell activity, as demonstrated by intensified mobility, viability, and apoptosis resistance in TNBC cells. KDM4E activated protein bicaudal D homolog 1 (BICD1) expression by reducing the deposition of histone H3 lysine 9 trimethylation (H3K9me3) in its promoter, whereas BICD1 promoted protease-activated receptor-1 (PAR1) endocytosis and blocked PAR1 signaling through physical interaction with PAR1. Knockdown of KDM4E strengthened the PAR1-dependent activity of TNBC cells in response to thrombin activation, whereas TNBC progression activated by PAR1 signaling was blocked by combined overexpression of BICD1. Taken together, our data indicate that baicalein-promoted KDM4E enhanced the expression of BICD1 and activated the inhibitory effect of BICD1 on PAR1 signaling, thereby inhibiting TNBC progression.
Collapse
Affiliation(s)
- Yun Dong
- Department of Traditional Chinese Medicine, Dazhou Vocational College of Chinese Medicine, Dazhou, Sichuan, China
| | - Gaojian He
- Dean's office, Dazhou Vocational College of Chinese Medicine, Dazhou, China
| | - Kun Chen
- Department of Technology and Social Services, Dazhou Vocational College of Chinese Medicine, Dazhou, China
| | - Xuefeng He
- Department of Technology and Social Services, Dazhou Vocational College of Chinese Medicine, Dazhou, China
| | - Meitong Pan
- Department of Technology and Social Services, Dazhou Vocational College of Chinese Medicine, Dazhou, China
| | - Xuemei Huang
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiaolan Yu
- Department of Obstetrics and Gynecology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jiyi Xia
- Department of Technology and Social Services, Dazhou Vocational College of Chinese Medicine, Dazhou, China
- Dazhou Chinese Medicine Research and Development Center, Dazhou, China
| |
Collapse
|
2
|
Suades R, Padró T, Vilahur G, Badimon L. Platelet-released extracellular vesicles: the effects of thrombin activation. Cell Mol Life Sci 2022; 79:190. [PMID: 35288766 PMCID: PMC8920058 DOI: 10.1007/s00018-022-04222-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 12/13/2022]
Abstract
Platelets exert fundamental roles in thrombosis, inflammation, and angiogenesis, contributing to different pathologies from cardiovascular diseases to cancer. We previously reported that platelets release extracellular vesicles (pEVs) which contribute to thrombus formation. However, pEV composition remains poorly defined. Indeed, pEV quality and type, rather than quantity, may be relevant in intravascular cross-talk with either circulating or vascular cells. We aimed to define the phenotypic characteristics of pEVs released spontaneously and those induced by thrombin activation to better understand their role in disease dissemination. pEVs obtained from washed platelets from healthy donor blood were characterized by flow cytometry. pEVs from thrombin-activated platelets (T-pEVs) showed higher levels of P-selectin and active form of glycoprotein IIb/IIIa than baseline non-activated platelets (B-pEVs). Following mass spectrometry-based differential proteomic analysis, significant changes in the abundance of proteins secreted in T-pEVs compared to B-pEVs were found. These differential proteins were involved in coagulation, adhesion, cytoskeleton, signal transduction, metabolism, and vesicle-mediated transport. Interestingly, release of proteins relevant for cell adhesion, intrinsic pathway coagulation, and platelet activation signalling was significantly modified by thrombin stimulation. A novel pEV-associated protein (protocadherin-α4) was found to be significantly reduced in T-pEVs showing a shift towards increased expression in the membranes of activated platelets. In summary, platelet activation induced by thrombin triggers the shedding of pEVs with a complex proteomic pattern rich in procoagulant and proadhesive proteins. Crosstalk with other vascular and blood cells in a paracrine regulatory mode could extend the prothrombotic signalling as well as promote proteostasic changes in other cellular types.
Collapse
Affiliation(s)
- Rosa Suades
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain.
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain.
- Cardiovascular Research Chair, UAB, Barcelona, Spain.
| |
Collapse
|
3
|
Jiang Y, Yao B, Chen T, Mo H, Chen S, Liu Q, Sun Y. BICD1 functions as a prognostic biomarker and promotes hepatocellular carcinoma progression. Pathol Res Pract 2020; 216:152858. [PMID: 32088084 DOI: 10.1016/j.prp.2020.152858] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/13/2020] [Accepted: 02/10/2020] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most predominant type of primary liver cancer and has a high degree of malignancy as well as mortality rate. Many drivers are involved in the development and progression of HCC. A recent study has reported that high BICD cargo adaptor 1 (BICD1) expression indicates poor prognosis of glioblastoma. But, the expression and biological function of BICD1 in HCC remain unclear. In current study, we found that the expression of BICD1 was markedly up-regulated in HCC tissues compared to adjacent nontumor tissues. GEPIA dataset and GSE datasets were consistently indicated the elevated expression of BICD1 in HCC. Furthermore, BICD1 was highly expressed in HCC cell lines including Hep3B, Huh7, MHCC97H and HCCLM3 as compared with that in LO2 cells. High BICD1 expression was positively correlated with malignant clinical features, such as tumor size ≥ 5 cm, venous infiltration and advanced tumor stages. HCC patients highly expressing BICD1 showed a significant shorter overall survival compared to BICD1 low-expression cases. Moreover, TCGA-LIHC data further demonstrated that the up-regulated BICD1 expression predicted poor prognosis of HCC patients. Next, we revealed that BICD1 knockdown prominently suppressed the proliferation, migration and invasion of HCCLM3 cells. Conversely, ectopic expression of BICD1 remarkably facilitated these malignant behaviors of Hep3B cells. Interestingly, BICD1 knockdown abolished hypoxia-induced HCC cell proliferation, migration and invasion. In conclusion, we provide the first evidence to support that BICD1 functions as a predictor for the prognosis of HCC and may serve as a promising therapeutic target for further HCC treatment.
Collapse
Affiliation(s)
- Yezhen Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China; Department of General Surgery, Xi'an Beihuan Hospital, Xi'an, Shaanxi Province 710032, China
| | - Bowen Yao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Tianxiang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Huanye Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Shuangjiang Chen
- Department of General Surgery, Ankang People's Hospital, Ankang, Shaanxi Province, 725000, China
| | - Qingguang Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China.
| | - Yali Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China.
| |
Collapse
|
4
|
Do AN, Zhao W, Baldridge AS, Raffield LM, Wiggins KL, Shah SJ, Aslibekyan S, Tiwari HK, Limdi N, Zhi D, Sitlani CM, Taylor KD, Psaty BM, Sotoodehnia N, Brody JA, Rasmussen‐Torvik LJ, Lloyd‐Jones D, Lange LA, Wilson JG, Smith JA, Kardia SLR, Mosley TH, Vasan RS, Arnett DK, Irvin MR. Genome-wide meta-analysis of SNP and antihypertensive medication interactions on left ventricular traits in African Americans. Mol Genet Genomic Med 2019; 7:e00788. [PMID: 31407531 PMCID: PMC6785453 DOI: 10.1002/mgg3.788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/14/2019] [Accepted: 04/22/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Left ventricular (LV) hypertrophy affects up to 43% of African Americans (AAs). Antihypertensive treatment reduces LV mass (LVM). However, interindividual variation in LV traits in response to antihypertensive treatments exists. We hypothesized that genetic variants may modify the association of antihypertensive treatment class with LV traits measured by echocardiography. METHODS We evaluated the main effects of the three most common antihypertensive treatments for AAs as well as the single nucleotide polymorphism (SNP)-by-drug interaction on LVM and relative wall thickness (RWT) in 2,068 participants across five community-based cohorts. Treatments included thiazide diuretics (TDs), angiotensin converting enzyme inhibitors (ACE-Is), and dihydropyridine calcium channel blockers (dCCBs) and were compared in a pairwise manner. We performed fixed effects inverse variance weighted meta-analyses of main effects of drugs and 2.5 million SNP-by-drug interaction estimates. RESULTS We observed that dCCBs versus TDs were associated with higher LVM after adjusting for covariates (p = 0.001). We report three SNPs at a single locus on chromosome 20 that modified the association between RWT and treatment when comparing dCCBs to ACE-Is with consistent effects across cohorts (smallest p = 4.7 × 10-8 , minor allele frequency range 0.09-0.12). This locus has been linked to LV hypertrophy in a previous study. A marginally significant locus in BICD1 (rs326641) was validated in an external population. CONCLUSIONS Our study identified one locus having genome-wide significant SNP-by-drug interaction effect on RWT among dCCB users in comparison to ACE-I users. Upon additional validation in future studies, our findings can enhance the precision of medical approaches in hypertension treatment.
Collapse
Affiliation(s)
- Anh N. Do
- Department of EpidemiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Wei Zhao
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - Laura M. Raffield
- Department of GeneticsUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Kerri L. Wiggins
- Cardiovascular Health Research Unit, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Sanjiv J. Shah
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Stella Aslibekyan
- Department of EpidemiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Hemant K. Tiwari
- Department of BiostatisticsUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Nita Limdi
- Department of NeurologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Degui Zhi
- School of Biomedical InformaticsUniversity of Texas Health Sciences Center at HoustonHoustonTexasUSA
| | - Colleen M. Sitlani
- Cardiovascular Health Research Unit, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Kent D. Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population SciencesLABioMed at Harbor‐UCLA Medical CenterSeattleWashingtonUSA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health ServicesUniversity of WashingtonSeattleWashingtonUSA
- Kaiser Permanente Washington Health Research InstituteSeattleWashingtonUSA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Departments of Medicine and EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Laura J. Rasmussen‐Torvik
- Department of Preventive Medicine Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | | | - Leslie A. Lange
- Department of MedicineUniversity of Colorado DenverAuroraColoradoUSA
| | - James G. Wilson
- Department of Physiology and BiophysicsUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Jennifer A. Smith
- Department of EpidemiologyUniversity of MichiganAnn ArborMichiganUSA
| | | | - Thomas H. Mosley
- Department of MedicineUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Ramachandran S. Vasan
- Departments of Medicine and Preventive MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Donna K. Arnett
- College of Public HealthUniversity of KentuckyLexingtonKentuckyUSA
| | - Marguerite R. Irvin
- Department of EpidemiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
5
|
Abstract
This overview article for the Comprehensive Physiology collection is focused on detailing platelets, how platelets respond to various stimuli, how platelets interact with their external biochemical environment, and the role of platelets in physiological and pathological processes. Specifically, we will discuss the four major functions of platelets: activation, adhesion, aggregation, and inflammation. We will extend this discussion to include various mechanisms that can induce these functional changes and a discussion of some of the salient receptors that are responsible for platelets interacting with their external environment. We will finish with a discussion of how platelets interact with their vascular environment, with a special focus on interactions with the extracellular matrix and endothelial cells, and finally how platelets can aid and possibly initiate the progression of various vascular diseases. Throughout this overview, we will highlight both the historical investigations into the role of platelets in health and disease as well as some of the more current work. Overall, the authors aim for the readers to gain an appreciation for the complexity of platelet functions and the multifaceted role of platelets in the vascular system. © 2017 American Physiological Society. Compr Physiol 8:1117-1156, 2018.
Collapse
Affiliation(s)
- David A Rubenstein
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Wei Yin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
6
|
Sinha RK, Wang Y, Zhao Z, Xu X, Burnier L, Gupta N, Fernández JA, Martin G, Kupriyanov S, Mosnier LO, Zlokovic BV, Griffin JH. PAR1 biased signaling is required for activated protein C in vivo benefits in sepsis and stroke. Blood 2018; 131:1163-1171. [PMID: 29343482 PMCID: PMC5855020 DOI: 10.1182/blood-2017-10-810895] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/28/2017] [Indexed: 12/12/2022] Open
Abstract
Activated protein C (APC) cleaves protease-activated receptor 1 (PAR1) in vitro at R46 to initiate beneficial cell signaling; however, thrombin and APC can cleave at R41. To elucidate PAR1-dependent aspects of the pharmacologic in vivo mechanisms of APC, we generated C57BL/6 mouse strains carrying QQ41 or QQ46 point mutations in PAR1 (F2r gene). Using these strains, we determined whether or not recombinant murine signaling-selective APC mutants would reduce septic death or provide neuroprotection against ischemic stroke when mice carried PAR1-homozygous mutations that prevent cleavage at either R41 or R46. Intercrossing PAR1+/R46Q mice generated expected numbers of PAR1+/+, PAR1+/R46Q, and R46Q/R46Q offspring whereas intercrossing PAR1+/R41Q mice gave decreased R41Q/R41Q homozygotes (resembling intercrossing PAR1+/PAR1-knockout mice). QQ41-PAR1 and QQ46-PAR1 brain endothelial cells showed the predicted retention or loss of cellular responses to thrombin receptor-activating peptide, thrombin, or APC for each PAR1 mutation. In sepsis studies, exogenous APC reduced mortality from 50% to 10% in Escherichia coli-induced pneumonia for wild-type (Wt) PAR1 and QQ41-PAR1 mice (P < .01) but had no benefit for QQ46-PAR1 mice. In transient distal middle cerebral artery occlusion stroke studies, exogenous APC significantly reduced infarct size, edema, and neuronal apoptosis for Wt mice and QQ41-PAR1 mice but had no detectable benefits for mice carrying QQ46-PAR1. In functional studies of forelimb-asymmetry and foot-fault tests at 24 hours after stroke induction, signaling-selective APC was beneficial for Wt and QQ41-PAR1 mice but not QQ46-PAR1 mice. These results support the concept that APC-induced, PAR1-dependent biased signaling following R46 cleavage is central to the in vivo benefits of APC.
Collapse
Affiliation(s)
| | - Yaoming Wang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA; and
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA; and
| | - Xiao Xu
- The Scripps Research Institute, La Jolla, CA
| | | | - Naveen Gupta
- The Scripps Research Institute, La Jolla, CA
- Department of Medicine, University of California San Diego, San Diego, CA
| | | | - Greg Martin
- The Scripps Research Institute, La Jolla, CA
| | | | | | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA; and
| | - John H Griffin
- The Scripps Research Institute, La Jolla, CA
- Department of Medicine, University of California San Diego, San Diego, CA
| |
Collapse
|
7
|
Huang SP, Chang YC, Low QH, Wu ATH, Chen CL, Lin YF, Hsiao M. BICD1 expression, as a potential biomarker for prognosis and predicting response to therapy in patients with glioblastomas. Oncotarget 2017; 8:113766-113791. [PMID: 29371945 PMCID: PMC5768362 DOI: 10.18632/oncotarget.22667] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/19/2017] [Indexed: 12/22/2022] Open
Abstract
There is variation in the survival and therapeutic outcome of patients with glioblastomas (GBMs). Therapy resistance is an important challenge in the treatment of GBM patients. The aim of this study was to identify Temozolomide (TMZ) related genes and confirm their clinical relevance. The TMZ-related genes were discovered by analysis of the gene-expression profiling in our cell-based microarray. Their clinical relevance was verified by in silico meta-analysis of the Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) datasets. Our results demonstrated that BICD1 expression could predict both prognosis and response to therapy in GBM patients. First, high BICD1 expression was correlated with poor prognosis in the TCGA GBM cohort (n=523) and in the CGGA glioma cohort (n=220). Second, high BICD1 expression predicted poor outcome in patients with TMZ treatment (n=301) and radiation therapy (n=405). Third, multivariable Cox regression analysis confirmed BICD1 expression as an independent factor affecting the prognosis and therapeutic response of TMZ and radiation in GBM patients. Additionally, age, MGMT and BICD1 expression were combinedly utilized to stratify GBM patients into more distinct risk groups, which may provide better outcome assessment. Finally, we observed a strong correlation between BICD1 expression and epithelial-mesenchymal transition (EMT) in GBMs, and proposed a possible mechanism of BICD1-associated survival or therapeutic resistance in GBMs accordingly. In conclusion, our study suggests that high BICD1 expression may result in worse prognosis and could be a predictor of poor response to TMZ and radiation therapies in GBM patients.
Collapse
Affiliation(s)
- Shang-Pen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurology, PoJen General Hospital, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chan Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Qie Hua Low
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Alexander T H Wu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Chi-Long Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yuan-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan.,Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
8
|
Thrombin-Induced Calpain Activation Promotes Protease-Activated Receptor 1 Internalization. Int J Cell Biol 2017; 2017:1908310. [PMID: 29250115 PMCID: PMC5700505 DOI: 10.1155/2017/1908310] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/02/2017] [Accepted: 10/15/2017] [Indexed: 12/19/2022] Open
Abstract
The serine protease thrombin activates Protease-Activated Receptors (PARs), a family of G-protein-coupled receptors (GPCRs) activated by the proteolytic cleavage of their extracellular N-terminal domain. Four members of this family have been identified: PAR1–4. The activation of Protease-Activated Receptor 1(PAR1), the prototype of this receptor family, leads to an increase in intracellular Ca+2 concentration ([Ca+2]i) mediated by Gq11α coupling and phospholipase C (PLC) activation. We have previously shown that the stimulation of PAR1 by thrombin promotes intracellular signaling leading to RPE cell transformation, proliferation, and migration which characterize fibroproliferative eye diseases leading to blindness. Within this context, the elucidation of the mechanisms involved in PAR1 inactivation is of utmost importance. Due to the irreversible nature of PAR1 activation, its inactivation must be efficiently regulated in order to terminate signaling. Using ARPE-19 human RPE cell line, we characterized thrombin-induced [Ca+2]i increase and demonstrated the calcium-dependent activation of μ-calpain mediated by PAR1. Calpains are a family of calcium-activated cysteine proteases involved in multiple cellular processes including the internalization of membrane proteins through clathrin-coated vesicles. We demonstrated that PAR1-induced calpain activation results in the degradation of α-spectrin by calpain, essential for receptor endocytosis, and the consequent decrease in PAR1 membrane expression. Collectively, the present results identify a novel μ-calpain-dependent mechanism for PAR1 inactivation following exposure to thrombin.
Collapse
|
9
|
Budzinska M, Wicher KB, Terenzio M. Neuronal Roles of the Bicaudal D Family of Motor Adaptors. VITAMINS AND HORMONES 2016; 104:133-152. [PMID: 28215293 DOI: 10.1016/bs.vh.2016.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
All cell types rely on active intracellular cargo transport to shuttle essential cellular components such as proteins, lipids, RNA, and even organelles from the center to the periphery and vice versa. Additionally, several signaling pathways take advantage of intracellular transport to propagate their signals by moving activated receptors and protein effectors to specific locations inside the cell. Neurons particularly, being a very polarized cell type, are highly dependent on molecular motors for the anterograde and retrograde delivery of essential cellular components and signaling molecules. For these reasons, motor adaptor proteins have been extensively investigated in regard to their role in physiology and pathology of the nervous system. In this chapter, we will concentrate on a family of motor adaptor proteins, Bicaudal D (BICD), and their function in the context of the nervous system. BicD was originally described as essential for the correct localization of maternal mRNAs in Drosophila's oocyte and a regulator of the Golgi to ER retrograde transport in mammalian cells. Both mammalian BICD1 and BICD2 are highly expressed in the nervous system during development, and their importance in neuronal homeostasis has been recently under scrutiny. Several mutations in BICD2 have been linked to the development of neuromuscular diseases, and BICD2 knockout (KO) mice display migration defects of the radial cerebellar granule cells. More in line with the overall topic of this book, BICD1 was identified as a novel regulator of neurotrophin (NT) signaling as its deletion leads to defective sorting of ligand-activated NT receptors with dramatic consequences on the NT-mediated signaling pathway.
Collapse
Affiliation(s)
- M Budzinska
- Molecular NeuroPathobiology Laboratory, UCL Institute of Neurology, University College London, London, United Kingdom
| | - K B Wicher
- Ossianix, Stevenage Bioscience Catalyst, Stevenage, United Kingdom
| | - M Terenzio
- Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
10
|
Ramachandran R, Altier C, Oikonomopoulou K, Hollenberg MD. Proteinases, Their Extracellular Targets, and Inflammatory Signaling. Pharmacol Rev 2016; 68:1110-1142. [PMID: 27677721 DOI: 10.1124/pr.115.010991] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Given that over 2% of the human genome codes for proteolytic enzymes and their inhibitors, it is not surprising that proteinases serve many physiologic-pathophysiological roles. In this context, we provide an overview of proteolytic mechanisms regulating inflammation, with a focus on cell signaling stimulated by the generation of inflammatory peptides; activation of the proteinase-activated receptor (PAR) family of G protein-coupled receptors (GPCR), with a mechanism in common with adhesion-triggered GPCRs (ADGRs); and by proteolytic ion channel regulation. These mechanisms are considered in the much wider context that proteolytic mechanisms serve, including the processing of growth factors and their receptors, the regulation of matrix-integrin signaling, and the generation and release of membrane-tethered receptor ligands. These signaling mechanisms are relevant for inflammatory, neurodegenerative, and cardiovascular diseases as well as for cancer. We propose that the inflammation-triggering proteinases and their proteolytically generated substrates represent attractive therapeutic targets and we discuss appropriate targeting strategies.
Collapse
Affiliation(s)
- Rithwik Ramachandran
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Christophe Altier
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Katerina Oikonomopoulou
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| | - Morley D Hollenberg
- Inflammation Research Network-Snyder Institute for Chronic Disease, Department of Physiology & Pharmacology (R.R., C.A., M.D.H.) and Department of Medicine (M.D.H.),University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pathology and Laboratory Medicine, Toronto Western Hospital, Toronto, Ontario, Canada (K.O.); and Department of Physiology and Pharmacology, Western University, London, Ontario, Canada (R.R.)
| |
Collapse
|
11
|
Braoudaki M, Lambrou GI, Vougas K, Karamolegou K, Tsangaris GT, Tzortzatou-Stathopoulou F. Protein biomarkers distinguish between high- and low-risk pediatric acute lymphoblastic leukemia in a tissue specific manner. J Hematol Oncol 2013; 6:52. [PMID: 23849470 PMCID: PMC3717072 DOI: 10.1186/1756-8722-6-52] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/04/2013] [Indexed: 12/13/2022] Open
Abstract
The current study evaluated the differential expression detected in the proteomic profiles of low risk- and high risk- ALL pediatric patients to characterize candidate biomarkers related to diagnosis, prognosis and patient targeted therapy. Bone marrow and peripheral blood plasma and cell lysates samples were obtained from pediatric patients with low- (LR) and high-risk (HR) ALL at diagnosis. As controls, non-leukemic pediatric patients were studied. Cytogenetic analysis was carried out by G- banding and interphase fluorescent in situ hybridization. Differential proteomic analysis was performed using two-dimensional gel electrophoresis and protein identification by matrix-assisted laser desorption ionization time-of-flight mass spectrometry. The differential expression of certain proteins was confirmed by Western blot analysis. The obtained data revealed that CLUS, CERU, APOE, APOA4, APOA1, GELS, S10A9, AMBP, ACTB, CATA and AFAM proteins play a significant role in leukemia prognosis, potentially serving as distinctive biomarkers for leukemia aggressiveness, or as suppressor proteins in HR-ALL cases. In addition, vitronectin and plasminogen probably contributed to leukemogenesis, whilst bicaudal D-related protein 1 could afford a significant biomarker for pediatric ALL therapeutics.
Collapse
Affiliation(s)
- Maria Braoudaki
- First Department of Pediatrics, University of Athens Medical School, Choremeio Research Laboratory, Thivon & Levadias 11527 Goudi-Athens, Greece
| | | | | | | | | | | |
Collapse
|
12
|
Oates E, Rossor A, Hafezparast M, Gonzalez M, Speziani F, MacArthur D, Lek M, Cottenie E, Scoto M, Foley A, Hurles M, Houlden H, Greensmith L, Auer-Grumbach M, Pieber T, Strom T, Schule R, Herrmann D, Sowden J, Acsadi G, Menezes M, Clarke N, Züchner S, Muntoni F, North K, Reilly M, Reilly MM. Mutations in BICD2 cause dominant congenital spinal muscular atrophy and hereditary spastic paraplegia. Am J Hum Genet 2013; 92:965-73. [PMID: 23664120 DOI: 10.1016/j.ajhg.2013.04.018] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 04/22/2013] [Accepted: 04/23/2013] [Indexed: 10/26/2022] Open
Abstract
Dominant congenital spinal muscular atrophy (DCSMA) is a disorder of developing anterior horn cells and shows lower-limb predominance and clinical overlap with hereditary spastic paraplegia (HSP), a lower-limb-predominant disorder of corticospinal motor neurons. We have identified four mutations in bicaudal D homolog 2 (Drosophila) (BICD2) in six kindreds affected by DCSMA, DCSMA with upper motor neuron features, or HSP. BICD2 encodes BICD2, a key adaptor protein that interacts with the dynein-dynactin motor complex, which facilitates trafficking of cellular cargos that are critical to motor neuron development and maintenance. We demonstrate that mutations resulting in amino acid substitutions in two binding regions of BICD2 increase its binding affinity for the cytoplasmic dynein-dynactin complex, which might result in the perturbation of BICD2-dynein-dynactin-mediated trafficking, and impair neurite outgrowth. These findings provide insight into the mechanism underlying both the static and the slowly progressive clinical features and the motor neuron pathology that characterize BICD2-associated diseases, and underscore the importance of the dynein-dynactin transport pathway in the development and survival of both lower and upper motor neurons.
Collapse
|
13
|
Protease-Activated Receptors. Platelets 2013. [DOI: 10.1016/b978-0-12-387837-3.00013-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
14
|
Weiler H. Multiple receptor-mediated functions of activated protein C. Hamostaseologie 2012; 31:185-95. [PMID: 21826371 DOI: 10.5482/ha-1166] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 06/17/2011] [Indexed: 01/08/2023] Open
Abstract
The central effector protease of the protein C pathway, activated protein C (APC), interacts with the endothelial cell protein C receptor, with protease activated receptors (PAR), the apolipoprotein E2 receptor, and integrins to exert multiple effects on haemostasis and immune cell function. Such receptor interactions modify the activation of PC and determine the biological response to endogenous and therapeutically administered APC. This review summarizes the current knowledge about interactions of APC with cell surface-associated receptors, novel substrates such as histones and tissue factor pathway inhibitor, and their implications for the biologic function of APC in the control of coagulation and inflammation.
Collapse
Affiliation(s)
- H Weiler
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee WI 53226, USA.
| |
Collapse
|
15
|
Targeting proteinase-activated receptors: therapeutic potential and challenges. Nat Rev Drug Discov 2012; 11:69-86. [PMID: 22212680 DOI: 10.1038/nrd3615] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Proteinase-activated receptors (PARs), a family of four seven-transmembrane G protein-coupled receptors, act as targets for signalling by various proteolytic enzymes. PARs are characterized by a unique activation mechanism involving the proteolytic unmasking of a tethered ligand that stimulates the receptor. Given the emerging roles of these receptors in cancer as well as in disorders of the cardiovascular, musculoskeletal, gastrointestinal, respiratory and central nervous system, PARs have become attractive targets for the development of novel therapeutics. In this Review we summarize the mechanisms by which PARs modulate cell function and the roles they can have in physiology and diseases. Furthermore, we provide an overview of possible strategies for developing PAR antagonists.
Collapse
|
16
|
Della-Morte D, Beecham A, Rundek T, Wang L, McClendon MS, Slifer S, Blanton SH, Di Tullio MR, Sacco RL. A follow-up study for left ventricular mass on chromosome 12p11 identifies potential candidate genes. BMC MEDICAL GENETICS 2011; 12:100. [PMID: 21791083 PMCID: PMC3199748 DOI: 10.1186/1471-2350-12-100] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 07/26/2011] [Indexed: 11/14/2022]
Abstract
Background Left ventricular mass (LVM) is an important risk factor for cardiovascular disease. Previously we found evidence for linkage to chromosome 12p11 in Dominican families, with a significant increase in a subset of families with high average waist circumference (WC). In the present study, we use association analysis to further study the genetic effect on LVM. Methods Association analysis with LVM was done in the one LOD critical region of the linkage peak in an independent sample of 897 Caribbean Hispanics. Genotype data were available on 7085 SNPs from 23 to 53 MB on chromosome 12p11. Adjustment was made for vascular risk factors and population substructure using an additive genetic model. Subset analysis by WC was performed to test for a difference in genetic effects between the high and low WC subsets. Results In the overall analysis, the most significant association was found to rs10743465, downstream of the SOX5 gene (p = 1.27E-05). Also, 19 additional SNPs had nominal p < 0.001. In the subset analysis, the most significant difference in genetic effect between those with high and low WC occurred with rs1157480 (p = 1.37E-04 for the difference in β coefficients), located upstream of TMTC1. Twelve additional SNPs in or near 6 genes had p < 0.001. Conclusions The current study supports previously identified evidence by linkage for a genetic effect on LVM on chromosome 12p11 using association analysis in population-based Caribbean Hispanic cohort. SOX5 may play an important role in the regulation of LVM. An interaction of TMTC1 with abdominal obesity may contribute to phenotypic variation of LVM.
Collapse
Affiliation(s)
- David Della-Morte
- Department of Neurology, Evelyn F. McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Vazquez-Pianzola P, Urlaub H, Suter B. Pabp binds to the osk 3'UTR and specifically contributes to osk mRNA stability and oocyte accumulation. Dev Biol 2011; 357:404-18. [PMID: 21782810 DOI: 10.1016/j.ydbio.2011.07.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 07/05/2011] [Accepted: 07/07/2011] [Indexed: 12/16/2022]
Abstract
RNA localization is tightly coordinated with RNA stability and translation control. Bicaudal-D (Bic-D), Egalitarian (Egl), microtubules and their motors are part of a Drosophila transport machinery that localizes mRNAs to specific cellular regions during oogenesis and embryogenesis. We identified the Poly(A)-binding protein (Pabp) as a protein that forms an RNA-dependent complex with Bic-D in embryos and ovaries. pabp also interacts genetically with Bic-D and, similar to Bic-D, pabp is essential in the germline for oocyte growth and accumulation of osk mRNA in the oocyte. In the absence of pabp, reduced stability of osk mRNA and possibly also defects in osk mRNA transport prevent normal oocyte localization of osk mRNA. pabp also interacts genetically with osk and lack of one copy of pabp(+) causes osk to become haploinsufficient. Moreover, pointing to a poly(A)-independent role, Pabp binds to A-rich sequences (ARS) in the osk 3'UTR and these turned out to be required in vivo for osk function during early oogenesis. This effect of pabp on osk mRNA is specific for this RNA and other tested mRNAs localizing to the oocyte are less and more indirectly affected by the lack of pabp.
Collapse
|
18
|
Saba LM, Bennett B, Hoffman PL, Barcomb K, Ishii T, Kechris K, Tabakoff B. A systems genetic analysis of alcohol drinking by mice, rats and men: influence of brain GABAergic transmission. Neuropharmacology 2011; 60:1269-80. [PMID: 21185315 PMCID: PMC3079014 DOI: 10.1016/j.neuropharm.2010.12.019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 12/01/2010] [Accepted: 12/15/2010] [Indexed: 11/19/2022]
Abstract
Genetic influences on the predisposition to complex behavioral or physiological traits can reflect genetic polymorphisms that lead to altered gene product function, and/or variations in gene expression levels. We have explored quantitative variations in an animal's alcohol consumption, using a genetical genomic/phenomic approach. In our studies, gene expression is correlated with amount of alcohol consumed, and genomic regions that regulate the alcohol consumption behavior and the quantitative levels of gene expression (behavioral and expression quantitative trait loci [QTL]) are determined and used as a filter to identify candidate genes predisposing the behavior. We determined QTLs for alcohol consumption using the LXS panel of recombinant inbred mice. We then identified genes that were: 1) differentially expressed between five high and five low alcohol-consuming lines or strains of mice; and 2) were physically located in, or had an expression QTL (eQTL) within the alcohol consumption QTLs. Comparison of mRNA and protein levels in brains of high and low alcohol consuming mice led us to a bioinformatic examination of potential regulation by microRNAs of an identified candidate transcript, Gnb1 (G protein beta subunit 1). We combined our current analysis with our earlier work identifying candidate genes for the alcohol consumption trait in mice, rats and humans. Our overall analysis leads us to postulate that the activity of the GABAergic system, and in particular GABA release and GABA receptor trafficking and signaling, which involves G protein function, contributes significantly to genetic variation in the predisposition to varying levels of alcohol consumption. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.
Collapse
Affiliation(s)
- Laura M. Saba
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Beth Bennett
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Paula L. Hoffman
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Kelsey Barcomb
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Takao Ishii
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| | - Katerina Kechris
- Colorado School of Public Health, Campus Box B119, Aurora, CO 80045 USA,
| | - Boris Tabakoff
- University of Colorado Denver School of Medicine, PO Box 6511, MS 8303, Aurora, CO 80045 USA; , , , , ,
| |
Collapse
|
19
|
Koukos G, Sevigny L, Zhang P, Covic L, Kuliopulos A. Serine and metalloprotease signaling through PAR1 in arterial thrombosis and vascular injury. IUBMB Life 2011; 63:412-8. [PMID: 21557445 DOI: 10.1002/iub.465] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 03/10/2011] [Indexed: 12/22/2022]
Abstract
Thrombin-dependent platelet activation has been shown to be important in the setting of angioplasty and stenting, which may cause ischemic complications including acute myocardial infarction and death. Inhibitors of the high-affinity thrombin receptor, protease-activated receptor 1 (PAR1), are now being evaluated in clinical trials for safety and efficacy in patients with atherothrombotic disease. However, it is unknown whether chronic inhibition of PAR1 in these large patient populations will have beneficial or possibly adverse effects on other biologic processes involved in blood vessel homeostasis and the response to vascular injury. Most recently, PAR1 was found to be cleaved at a distinct site by matrix metalloprotease-1 (MMP-1) to create a longer tethered ligand, which activates a distinct spectrum of G protein pathways in platelets. The differential activation by serine proteases such as thrombin and the metalloprotease MMP-1, places the protease receptor PAR1 at the junction of two major protease classes critically involved in thrombosis, matrix remodeling, and the response to vascular injury.
Collapse
Affiliation(s)
- Georgios Koukos
- Hemostasis and Thrombosis Laboratory, Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | | | | | | | | |
Collapse
|
20
|
Interdicting protease-activated receptor-2-driven inflammation with cell-penetrating pepducins. Proc Natl Acad Sci U S A 2011; 108:8491-6. [PMID: 21536878 DOI: 10.1073/pnas.1017091108] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Protease-activated receptor-2 (PAR2), a cell surface receptor for trypsin-like proteases, plays a key role in a number of acute and chronic inflammatory diseases of the joints, lungs, brain, gastrointestinal tract, and vascular systems. Despite considerable effort by the pharmaceutical industry, PAR2 has proven recalcitrant to targeting by small molecule inhibitors, which have been unable to effectively prevent the interaction of the protease-generated tethered ligand with the body of the receptor. Here, we report the development of first-in-class cell-penetrating lipopeptide "pepducin" antagonists of PAR2. The design of the third intracellular (i3) loop pepducins were based on a structural model of a PAR2 dimer and by mutating key pharmacophores in the receptor intracellular loops and analogous pepducins. Individual pharmacophores were identified, which controlled constitutive, agonist, and antagonist activities. This approach culminated in the identification of the P2pal-18S pepducin which completely suppressed trypsin and mast cell tryptase signaling through PAR2 in neutrophils and colon cancer cells. The PAR2 pepducin was highly efficacious in blocking PAR2-dependent inflammatory responses in mouse models. These effects were lost in PAR2-deficient and mast-cell-deficient mice, thereby validating the specificity of the pepducin in vivo. These data provide proof of concept that PAR2 pepducin antagonists may afford effective treatments of potentially debilitating inflammatory diseases and serve as a blueprint for developing highly potent and specific i3-loop-based pepducins for other G protein-coupled receptors (GPCRs).
Collapse
|
21
|
Adams MN, Ramachandran R, Yau MK, Suen JY, Fairlie DP, Hollenberg MD, Hooper JD. Structure, function and pathophysiology of protease activated receptors. Pharmacol Ther 2011; 130:248-82. [PMID: 21277892 DOI: 10.1016/j.pharmthera.2011.01.003] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 01/03/2011] [Indexed: 12/18/2022]
Abstract
Discovered in the 1990s, protease activated receptors(1) (PARs) are membrane-spanning cell surface proteins that belong to the G protein coupled receptor (GPCR) family. A defining feature of these receptors is their irreversible activation by proteases; mainly serine. Proteolytic agonists remove the PAR extracellular amino terminal pro-domain to expose a new amino terminus, or tethered ligand, that binds intramolecularly to induce intracellular signal transduction via a number of molecular pathways that regulate a variety of cellular responses. By these mechanisms PARs function as cell surface sensors of extracellular and cell surface associated proteases, contributing extensively to regulation of homeostasis, as well as to dysfunctional responses required for progression of a number of diseases. This review examines common and distinguishing structural features of PARs, mechanisms of receptor activation, trafficking and signal termination, and discusses the physiological and pathological roles of these receptors and emerging approaches for modulating PAR-mediated signaling in disease.
Collapse
Affiliation(s)
- Mark N Adams
- Mater Medical Research Institute, Aubigny Place, Raymond Terrace, South Brisbane Qld 4101, Australia
| | | | | | | | | | | | | |
Collapse
|
22
|
Leonardi S, Tricoci P, Becker RC. Thrombin Receptor Antagonists for the Treatment of Atherothrombosis. Drugs 2010; 70:1771-83. [DOI: 10.2165/11538060-000000000-00000] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
23
|
Affiliation(s)
- Marco Terenzio
- Molecular NeuroPathobiology, Cancer Research UK London Research Institute, London, UK
| | | |
Collapse
|