Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 300
Filter
1.
J Thromb Haemost ; 22(7): 2039-2051, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38670314

ABSTRACT

BACKGROUND: Activated protein C (APC) has anticoagulant and cytoprotective cell-signaling activities, which often require protease-activated receptor (PAR) 1 and PAR3 and PAR cleavages at noncanonical sites (R46-N47 and R41-G42, respectively). Some PAR1-derived (P1) peptides and PAR3-derived (P3) peptides, eg, P1-47-66 and P3-42-65, mimic APC's cell signaling. In anti-inflammatory assays, these 2 peptides at low concentrations synergistically attenuate cellular inflammation. OBJECTIVES: To determine whether a P1 peptide covalently linked to a P3 peptide mimics APC's anti-inflammatory and endothelial barrier stabilization activities. METHODS: Anti-inflammatory assays employed stimulated THP-1 cells and caspase-1 measurements. Cultured human EA.hy926 or murine aortic endothelial cells (ECs) exposed to thrombin were monitored for transendothelial electrical resistance. Bivalent covalently linked P1:P3 peptides were studied for APC-like activities. RESULTS: In anti-inflammatory assays, P1-47-55 was as active as P1-47-66 and some P3 peptides (eg, P3-44-54 and P3-51-65) were as active as P3-42-65. The bivalent P1:P3 peptide comprising P1-47-55-(Gly[10 residues])-P3-51-65 (designated "G10 peptide") was more potently anti-inflammatory than the P1 or P3 peptide alone. In transendothelial electrical resistance studies of thrombin-challenged ECs, P1-47-55 and the G10 peptide mimicked APC's protective actions. In dose-response studies, the G10 peptide was more potent than the P1-47-55 peptide. In murine EC studies, the murine PAR-sequence-derived G10 peptide mimicked murine APC's activity. Anti-PAR1 and anti-PAR3 antibodies, but not anti-endothelial protein C receptor antibodies, abated G10's cytoprotection, showing that G10's actions involve PAR1:PAR3. G10 significantly increased survival in murine endotoxemia. CONCLUSION: The PAR-sequence-derived G10 peptide is a bivalent agonist that mimics APC's cytoprotective, anti-inflammatory, and endothelial barrier-stabilizing actions and APC's protection against endotoxemic mortality.


Subject(s)
Endothelial Cells , Protein C , Receptor, PAR-1 , Protein C/metabolism , Protein C/chemistry , Humans , Animals , Receptor, PAR-1/agonists , Receptor, PAR-1/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Mice, Inbred C57BL , THP-1 Cells , Thrombin/metabolism , Endothelial Protein C Receptor/metabolism , Receptors, Thrombin/agonists , Receptors, Thrombin/metabolism , Signal Transduction , Receptors, Proteinase-Activated/agonists , Receptors, Proteinase-Activated/metabolism , Peptides/pharmacology , Peptides/chemistry , Endotoxemia/drug therapy , Endotoxemia/metabolism , Peptide Fragments/pharmacology , Male , Disease Models, Animal
2.
J Invest Dermatol ; 144(5): 950-953, 2024 May.
Article in English | MEDLINE | ID: mdl-38430083

ABSTRACT

Pruritus or itch is a defining symptom of atopic dermatitis (AD). The origins of itch are complex, and it is considered both a defense mechanism and a cause of disease that leads to inflammation and psychological stress. Considerable progress has been made in understanding the processes that trigger itch, particularly the pruritoceptive origins that are generated in the skin. This perspective review discusses the implications of a recent observation that the V8 protease expressed by Staphylococcus aureus can directly trigger sensory neurons in the skin through activation of protease-activated receptor 1. This may be a key to understanding why itch is so common in AD because S. aureus commonly overgrows in this disease owing to deficient antimicrobial defense from both the epidermis and the cutaneous microbiome. Increased understanding of the role of microbes in AD provides increased opportunities for safely improving the treatment of this disorder.


Subject(s)
Dermatitis, Atopic , Pruritus , Staphylococcus aureus , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/complications , Dermatitis, Atopic/immunology , Humans , Pruritus/microbiology , Pruritus/immunology , Animals , Skin/microbiology , Skin/pathology , Sensory Receptor Cells/physiology , Sensory Receptor Cells/metabolism , Receptors, Proteinase-Activated/metabolism , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology
3.
J Thromb Haemost ; 22(6): 1550-1557, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38460839

ABSTRACT

Platelets are well-known players in several cardiovascular diseases such as atherosclerosis and venous thrombosis. There is increasing evidence demonstrating that reactive oxygen species (ROS) are generated within activated platelets. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is a major source of ROS generation in platelets. Ligand binding to platelet receptor glycoprotein (GP) VI stimulates intracellular ROS generation consisting of a spleen tyrosine kinase-independent production involving NOX activation and a following spleen tyrosine kinase-dependent generation. In addition to GPVI, stimulation of platelet thrombin receptors (protease-activated receptors [PARs]) can also trigger NOX-derived ROS production. Our recent study found that mitochondria-derived ROS production can be induced by engagement of thrombin receptors but not by GPVI, indicating that mitochondria are another source of PAR-dependent ROS generation apart from NOX. However, mitochondria are not involved in GPVI-dependent ROS generation. Once generated, the intracellular ROS are also involved in modulating platelet function and thrombus formation; therefore, the site-specific targeting of ROS production or clearance of excess ROS within platelets is a potential intervention and treatment option for thrombotic events. In this review, we will summarize the signaling pathways involving regulation of platelet ROS production and their role in platelet function and thrombosis, with a focus on GPVI- and PAR-dependent platelet responses.


Subject(s)
Blood Platelets , Oxidation-Reduction , Reactive Oxygen Species , Signal Transduction , Thrombosis , Humans , Blood Platelets/metabolism , Reactive Oxygen Species/metabolism , Thrombosis/blood , Platelet Membrane Glycoproteins/metabolism , Animals , Platelet Activation , Mitochondria/metabolism , NADPH Oxidases/metabolism , Receptors, Thrombin/metabolism , Receptors, Proteinase-Activated/metabolism
4.
J Biol Chem ; 300(2): 105614, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38159863

ABSTRACT

The activation and mobilization of immune cells play a crucial role in immunotherapy. Existing therapeutic interventions, such as cytokines administration, aim to enhance immune cell activity. However, these approaches usually result in modest effectiveness and toxic side effects, thereby restricting their clinical application. Protease-activated receptors (PARs), a subfamily of G protein-coupled receptors, actively participate in the immune system by directly activating immune cells. The activation of PARs by proteases or synthetic ligands can modulate immune cell behavior, signaling, and responses to treat immune-related diseases, suggesting the significance of PARs agonism in immunotherapy. However, the agonism of PARs in therapeutical applications remains rarely discussed, since it has been traditionally considered that PARs activation facilitates disease progressions. This review aims to comprehensively summarize the activation, rather than inhibition, of PARs in immune-related physiological responses and diseases. Additionally, we will discuss the emerging immunotherapeutic potential of PARs agonism, providing a new strategic direction for PARs-mediated immunotherapy.


Subject(s)
Immunotherapy , Receptors, Proteinase-Activated , Peptide Hydrolases/metabolism , Receptors, G-Protein-Coupled , Receptors, Proteinase-Activated/agonists , Receptors, Proteinase-Activated/metabolism , Signal Transduction , Neoplasms/immunology , Neoplasms/therapy , Immune System Diseases/immunology , Immune System Diseases/therapy , Humans , Animals
5.
Int J Mol Sci ; 24(9)2023 May 05.
Article in English | MEDLINE | ID: mdl-37176005

ABSTRACT

Protease-activated receptors (PARs) are a class of integral membrane proteins that are cleaved by a variety of proteases, most notably thrombin, to reveal a tethered ligand and promote activation. PARs are critical mediators of platelet function in hemostasis and thrombosis, and therefore are attractive targets for anti-platelet therapies. Animal models studying platelet PAR physiology have relied heavily on genetically modified mouse strains, which have provided ample insight but have some inherent limitations. The current review aims to summarize the notable PAR expression and functional differences between the mouse and human, in addition to highlighting some recently developed tools to further study human physiology in mouse models.


Subject(s)
Receptors, Proteinase-Activated , Receptors, Thrombin , Humans , Mice , Animals , Receptors, Proteinase-Activated/metabolism , Receptors, Thrombin/metabolism , Species Specificity , Blood Platelets/metabolism , Thrombin/metabolism
6.
Clin. transl. oncol. (Print) ; 25(5): 1242-1251, mayo 2023. ilus
Article in English | IBECS | ID: ibc-219509

ABSTRACT

Colorectal cancer (CRC) is one of the common malignancies with a global trend of increasing incidence and mortality. There is an urgent need to identify new predictive markers and therapeutic targets for the treatment of CRC. Protease-activated receptors (PARs) are a class of G-protein-coupled receptors, with currently identified subtypes including PAR1, PAR2, PAR3 and PAR4. Increasingly, studies suggest that PARs play an important role in the growth and metastasis of CRC. By targeting multiple signaling pathways may contribute to the pathogenesis of CRC. In this review, we first describe recent studies on the role of PARs in CRC inflammation-cancer transformation, focusing on the important role of PARs in signaling pathways associated with inflammation-cancer transformation, and summarize the progress of research on PARs-targeted drugs (AU)


Subject(s)
Humans , Colorectal Neoplasms/metabolism , Receptors, Proteinase-Activated/metabolism , Receptors, Thrombin/metabolism , Signal Transduction , Inflammation
7.
J Thromb Haemost ; 21(8): 2236-2247, 2023 08.
Article in English | MEDLINE | ID: mdl-37068592

ABSTRACT

BACKGROUND: Thromboelastography (TEG) is used for real-time determination of hemostatic status in patients with acute risk of bleeding. Thrombin is thought to drive clotting in TEG through generation of polymerized fibrin and activation of platelets through protease-activated receptors (PARs). However, the specific role of platelet agonist receptors and signaling in TEG has not been reported. OBJECTIVES: Here, we investigated the specific receptors and signaling pathways required for platelet function in TEG using genetic and pharmacologic inhibition of platelet proteins in mouse and human blood samples. METHODS: Clotting parameters (R time, α-angle [α], and maximum amplitude [MA]), were determined in recalcified, kaolin-triggered citrated blood samples using a TEG 5000 analyzer. RESULTS: We confirmed the requirement of platelets, platelet contraction, and αIIbß3 integrin function for normal α and MA. Loss of the integrin adaptor Talin1 in megakaryocytes/platelets (Talin1mKO) also reduced α and MA, but only minimal defects were observed in samples from mice lacking Rap1 GTPase signaling. PAR4mKO samples showed impaired α but normal MA. However, impaired TEG traces similar to those in platelet-depleted samples were observed with samples from PAR4mKO mice depleted of glycoprotein VI on platelets or with addition of a Syk inhibitor. We reproduced these results in human blood with combined inhibition of PAR1, PAR4, and Syk. CONCLUSION: Our results demonstrate that standard TEG is not sensitive to platelet signaling pathways critical for integrin inside-out activation and platelet hemostatic function. Furthermore, we provide the first evidence that PARs and glycoprotein VI play redundant roles in platelet-mediated clot contraction in TEG.


Subject(s)
Blood Platelets , Hemostatics , Animals , Humans , Mice , Blood Platelets/metabolism , Glycoproteins/metabolism , Integrins/metabolism , Receptors, Proteinase-Activated/metabolism , Receptors, Thrombin/genetics , Receptors, Thrombin/metabolism , Thrombelastography/methods
8.
Clin Transl Oncol ; 25(5): 1242-1251, 2023 May.
Article in English | MEDLINE | ID: mdl-36547764

ABSTRACT

Colorectal cancer (CRC) is one of the common malignancies with a global trend of increasing incidence and mortality. There is an urgent need to identify new predictive markers and therapeutic targets for the treatment of CRC. Protease-activated receptors (PARs) are a class of G-protein-coupled receptors, with currently identified subtypes including PAR1, PAR2, PAR3 and PAR4. Increasingly, studies suggest that PARs play an important role in the growth and metastasis of CRC. By targeting multiple signaling pathways may contribute to the pathogenesis of CRC. In this review, we first describe recent studies on the role of PARs in CRC inflammation-cancer transformation, focusing on the important role of PARs in signaling pathways associated with inflammation-cancer transformation, and summarize the progress of research on PARs-targeted drugs.


Subject(s)
Neoplasms , Receptors, Proteinase-Activated , Humans , Receptors, Proteinase-Activated/metabolism , Receptors, Thrombin/metabolism , Signal Transduction , Inflammation
9.
Biomed Res Int ; 2022: 3865844, 2022.
Article in English | MEDLINE | ID: mdl-36246974

ABSTRACT

Matriptases are cell surface proteolytic enzymes belonging to the type II transmembrane serine protease family that mediate inflammatory skin disorders and cancer progression. Matriptases may affect the development of periodontitis via protease-activated receptor-2 activity. However, the cellular mechanism by which matriptases are involved in periodontitis is unknown. In this study, we examined the antiperiodontitis effects of matriptase on Porphyromonas gingivalis-derived lipopolysaccharide (PG-LPS)-stimulated human gingival fibroblasts (HGFs). Matriptase small interfering RNA-transfected HGFs were treated with PG-LPS. The mRNA and protein levels of proinflammatory cytokines and matrix metalloproteinase 1 (MMP-1) were evaluated using the quantitative real-time polymerase chain reaction (qRT-PCR) and an enzyme-linked immunosorbent assay (ELISA), respectively. Western blot analyses were performed to measure the levels of Toll-like receptor 4 (TLR4)/interleukin-1 (IL-1) receptor-associated kinase (IRAK)/transforming growth factor ß-activated kinase 1 (TAK1), p65, and p50 in PG-LPS-stimulated HGFs. Matriptase downregulation inhibited LPS-induced proinflammatory cytokine expression, including the expression of IL-6, IL-8, tumor necrosis factor-α (TNF-α), and IL-Iß. Moreover, matriptase downregulation inhibited PG-LPS-stimulated MMP-1 expression. Additionally, we confirmed that the mechanism underlying the effects of matriptase downregulation involves the suppression of PG-LPS-induced IRAK1/TAK1 and NF-κB. These results suggest that downregulation of matriptase PG-LPS-induced MMP-1 and proinflammatory cytokine expression via TLR4-mediated IRAK1/TAK1 and NF-κB signaling pathways in HGFs.


Subject(s)
Fibroblasts , Matrix Metalloproteinase 1 , Periodontitis , Serine Endopeptidases , Cytokines/metabolism , Down-Regulation , Fibroblasts/metabolism , Humans , Interleukin-1/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Lipopolysaccharides/toxicity , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , NF-kappa B/metabolism , Periodontitis/genetics , Periodontitis/metabolism , Porphyromonas gingivalis , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Proteinase-Activated/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Cell Mol Biol Lett ; 27(1): 77, 2022 Sep 10.
Article in English | MEDLINE | ID: mdl-36088291

ABSTRACT

Protease activated receptors (PARs) are among the first receptors shown to transactivate other receptors: noticeably, these interactions are not limited to members of the same family, but involve receptors as diverse as receptor kinases, prostanoid receptors, purinergic receptors and ionic channels among others. In this review, we will focus on the evidence for PAR interactions with members of their own family, as well as with other types of receptors. We will discuss recent evidence as well as what we consider as emerging areas to explore; from the signalling pathways triggered, to the physiological and pathological relevance of these interactions, since this additional level of molecular cross-talk between receptors and signaling pathways is only beginning to be explored and represents a novel mechanism providing diversity to receptor function and play important roles in physiology and disease.


Subject(s)
Receptors, Proteinase-Activated , Signal Transduction , Receptors, Proteinase-Activated/metabolism , Signal Transduction/physiology
11.
Front Immunol ; 13: 912748, 2022.
Article in English | MEDLINE | ID: mdl-35844627

ABSTRACT

Candida albicans Sap6, a secreted aspartyl protease (Sap), contributes to fungal virulence in oral candidiasis. Beside its protease activity, Sap6 contains RGD (RGDRGD) motif required for its binding to host integrins. Sap6 activates immune cells to induce proinflammatory cytokines, although its ability to interact and activate human oral epithelial cells (OECs) remain unknown. Addition of purified recombinant Sap6 (rSap6) to OECs resulted in production of IL-1ß and IL-8 cytokines similar to live hyphal C. albicans. OECs exposed to rSap6 showed phosphorylation of p38 and MKP1 and expression of c-Fos not found with C. albicans Δsap6, heat-inactivated Sap6, or rSap6ΔRGD . Heat inactivated rSap6 was able to induce IL-1ß but not IL-8 in OECs, while rSap6ΔRGD induced IL-8 but not IL-1ß suggesting parallel signaling pathways. C. albicans hyphae increased surface expression of Protease Activated Receptors PAR1, PAR2 and PAR3, while rSap6 increased PAR2 expression exclusively. Pretreatment of OECs with a PAR2 antagonist blocked rSap6-induced p38 MAPK signaling and IL-8 release, while rSap6ΔRGD had reduced MKP1 signaling and IL-1ß release independent from PAR2. OECs exposed to rSap6 exhibited loss of barrier function as measured by TEER and reduction in levels of E-cadherin and occludin junctional proteins that was prevented by pretreating OECs with a PAR2 antagonist. OECs treated with PAR2 antagonist also showed reduced rSap6-mediated invasion by C. albicans cells. Thus, Sap6 may initiate OEC responses mediated both through protease activation of PAR2 and by its RGD domain. This novel role of PAR2 suggests new drug targets to block C. albicans oral infection.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Candida albicans , Fungal Proteins/metabolism , Receptor, PAR-2/metabolism , Stomatitis/microbiology , Cytokines/metabolism , Host-Pathogen Interactions , Humans , Inflammation , Receptors, Proteinase-Activated/metabolism
12.
FEBS J ; 289(14): 4000-4020, 2022 07.
Article in English | MEDLINE | ID: mdl-35175692

ABSTRACT

Inflammatory diseases have become increasingly prevalent throughout the world. Coronavirus disease 2019 (COVID-19), which has recently become pandemic, also exhibits hyperinflammation and cytokine release syndrome. To address inflammation-related diseases, numerous molecular targets have been explored in preclinical studies and clinical trials. Among them, the protease-activated receptors (PARs) that belong to G protein-coupled receptors are one of the most popular classes of drug targets, participating in inflammatory signalling and diseases. PARs activation can trigger downstream intracellular signalling to modulate a variety of inflammatory responses in multiple systems, including nervous, respiratory, digestive, circulatory, urinary and immune systems. Importantly, there are the Yin-Yang effects, comprising anti- and pro-inflammatory roles, of PARs activation in different types of inflammations, and these are context-dependent. Alternatively, it was recently revealed that PARs not only modulate inflammatory-related tumour progression, but also participate in inflammatory cytokine release related to COVID-19 via direct interaction with severe acute respiratory syndrome coronavirus 2 protein, suggesting that PARs also participate in other diseases via inflammatory responses. In this review, we renew and discuss the findings of PARs as molecular targets for treating inflammatory diseases, highlighting the novel roles of PARs and facilitating a better understanding of their designated values in the specific inflammatory environment.


Subject(s)
COVID-19 Drug Treatment , Receptors, Proteinase-Activated , Humans , Inflammation , Receptors, Proteinase-Activated/metabolism , Signal Transduction/physiology
13.
Prostate ; 82(6): 723-739, 2022 05.
Article in English | MEDLINE | ID: mdl-35167724

ABSTRACT

BACKGROUND: Metastatic prostate cancer lesions in the skeleton are frequently characterized by excessive formation of bone. Prostate cancer cells secrete factors, including serine proteases, that are capable of influencing the behavior of surrounding cells. Some of these proteases activate protease-activated receptor-2 (PAR2 ), which is expressed by osteoblasts (bone-forming cells) and precursors of osteoclasts (bone-resorbing cells). The aim of the current study was to investigate a possible role for PAR2 in regulating the behavior of bone cells exposed to metastatic prostate cancer cells. METHODS: The effect of medium conditioned by the PC3, DU145, and MDA-PCa-2b prostate cancer cell lines was investigated in assays of bone cell function using cells isolated from wildtype and PAR2 -null mice. Osteoclast differentiation was assessed by counting tartrate-resistant acid phosphatase-positive multinucleate cells in bone marrow cultured in osteoclastogenic medium. Osteoblasts were isolated from calvariae of neonatal mice, and BrdU incorporation was used to assess their proliferation. Assays of alkaline phosphatase activity and quantitative PCR analysis of osteoblastic gene expression were used to assess osteoblast differentiation. Responses of osteoblasts to medium conditioned by MDA-PCa-2b cells were analyzed by RNAseq. RESULTS: Conditioned medium (CM) from all three cell lines inhibited osteoclast differentiation independently of PAR2 . Media from PC3 and DU145 cells had no effect on assays of osteoblast function. Medium conditioned by MDA-PCa-2b cells stimulated BrdU incorporation in both wildtype and PAR2 -null osteoblasts but increased alkaline phosphatase activity and Runx2 and Col1a1 expression in wildtype but not PAR2 -null cells. Functional enrichment analysis of RNAseq data identified enrichment of multiple gene ontology terms associated with lysosomal function in both wildtype and PAR2 -null cells in response to MDA-PCa-2b-CM. Analysis of individual genes identified osteogenesis-associated genes that were either upregulated by MDA-PCa-2b-CM selectively in wildtype cells or downregulated selectively in PAR2 -null cells. CONCLUSIONS: Factors secreted by prostate cancer cells influence bone cell behavior through both PAR2 -dependent and -independent mechanisms. Both PAR2 -independent suppression of osteoclast differentiation and PAR2 -dependent stimulation of osteogenesis are likely to determine the nature of prostate cancer metastases in bone.


Subject(s)
Bone Neoplasms , Prostatic Neoplasms , Receptor, PAR-2/metabolism , Alkaline Phosphatase/metabolism , Alkaline Phosphatase/pharmacology , Animals , Bone Neoplasms/secondary , Bromodeoxyuridine/metabolism , Bromodeoxyuridine/pharmacology , Cell Differentiation , Cell Line, Tumor , Humans , Male , Mice , Osteoblasts/metabolism , Osteoclasts/metabolism , Prostatic Neoplasms/pathology , Receptors, Proteinase-Activated/metabolism
14.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163205

ABSTRACT

Acute Respiratory Distress Syndrome is the most common cause of respiratory failure among critically ill patients, and its importance has been heightened during the COVID-19 pandemic. Even with the best supportive care, the mortality rate in the most severe cases is 40-50%, and the only pharmacological agent shown to be of possible benefit has been steroids. Mesenchymal stromal cells (MSCs) have been tested in several pre-clinical models of lung injury and been found to have significant therapeutic benefit related to: (a) potent immunomodulation; (b) secretion of epithelial and endothelial growth factors; and (c) augmentation of host defense to infection. Initial translational efforts have shown signs of promise, but the results have not yielded the anticipated outcomes. One potential reason is the relatively low survival of MSCs in inflammatory conditions as shown in several studies. Therefore, strategies to boost the survival of MSCs are needed to enhance their therapeutic effect. Protease-activated receptors (PARs) may represent one such possibility as they are G-protein coupled receptors expressed by MSCs and control several facets of cell behavior. This review summarizes some of the existing literature about PARs and MSCs and presents possible future areas of investigation in order to develop potential, PAR-modified MSCs with enhanced therapeutic efficiency.


Subject(s)
Graft Survival/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Receptors, Proteinase-Activated/physiology , Respiratory Distress Syndrome/therapy , Animals , COVID-19/genetics , COVID-19/pathology , COVID-19/therapy , Cell Survival/genetics , Critical Illness/therapy , Humans , Mesenchymal Stem Cells/physiology , Receptors, Proteinase-Activated/genetics , Receptors, Proteinase-Activated/metabolism , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/virology , SARS-CoV-2/physiology , Signal Transduction/physiology , Transfection , Treatment Outcome
15.
Am J Pathol ; 192(2): 361-378, 2022 02.
Article in English | MEDLINE | ID: mdl-35144762

ABSTRACT

As per the classical view of the coagulation system, it functions solely in plasma to maintain hemostasis. An experimental approach modeling vascular reconstitution was used to show that vascular endothelial cells (ECs) endogenously synthesize coagulation factors during angiogenesis. Intracellular thrombin generated from this synthesis promotes the mitotic function of vascular endothelial cell growth factor A (VEGF-A). The thrombin concurrently cleaves C5a from EC-synthesized complement component C5 and unmasks the tethered ligand for EC-expressed protease-activated receptor 4 (PAR4). The two ligands jointly trigger EC C5a receptor-1 (C5ar1) and PAR4 signaling, which together promote VEGF receptor 2 growth signaling. C5ar1 is functionally associated with PAR4, enabling C5a or thrombin to elicit Gαi and/or Gαq signaling. EC coagulation factor and EC complement component synthesis concurrently down-regulate with contact inhibition. The connection of these processes with VEGF receptor 2 signaling provides new insights into mechanisms underlying angiogenesis. Knowledge of endogenous coagulation factor/complement component synthesis and joint PAR4/C5ar1 signaling could be applied to other cell types.


Subject(s)
Blood Coagulation Factors/biosynthesis , Endothelial Cells/metabolism , Neovascularization, Physiologic , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Proteinase-Activated/metabolism , Signal Transduction , Animals , Blood Coagulation Factors/genetics , Female , Male , Mice , Mice, Knockout , Receptor, Anaphylatoxin C5a/genetics , Receptors, Proteinase-Activated/genetics
16.
Thromb Haemost ; 122(6): 961-973, 2022 06.
Article in English | MEDLINE | ID: mdl-34619794

ABSTRACT

The Ig-ITIM bearing receptors, PECAM-1 and CEACAM1, have been shown net negative regulators of platelet-collagen interactions and hemiITAM signaling pathways. In this study, a double knockout (DKO) mouse was developed with deleted PECAM-1 and CEACAM1 to study their combined contribution in platelet activation by glycoprotein VI, C-type lectin-like receptor 2, protease activated receptor (PAR4), ADP purinergic receptors, and thromboxane receptor (TP) A2 pathways. In addition, their collective contribution was examined in thrombus formation under high shear and microvascular thrombosis using in vivo models. DKO platelets responded normally to ADP purinergic receptors and the TP A2 pathway. However, DKO platelets released significantly higher amounts of P-selectin compared with hyper-responsive Pecam-1-/- or Ceacam1-/- versus wild-type (WT) upon stimulation with collagen-related peptide or rhodocytin. In contrast, DKO platelets showed increased amounts of P-selectin exposure upon stimulation with PAR4 agonist peptide or thrombin but not Pecam-1-/- , Ceacam1-/- , or WT platelets. Blockade of phospholipase C (PLC) or Rho A kinase revealed that DKO platelets enhanced α-granule release via PAR4/Gαq/PLC signaling without crosstalk with Src/Syk or G12/13 signaling pathways. Severely delayed clot retraction in vitro was observed in DKO phenotype. The DKO model revealed a significant increase in thrombus formation compared with the hyper-responsive Ceacam1-/- or Pecam-1-/- versus WT phenotype. DKO platelets have similar glycoprotein surface expression compared with Pecam-1-/- , Ceacam1-/- , and WT platelets. This study demonstrates that PECAM-1 and CEACAM1 work in concert to negatively regulate hemiITAM signaling, platelet-collagen interactions, and PAR4 Gαq protein- coupled signaling pathways. Both PECAM-1 and CEACAM1 are required for negative regulation of platelet activation and microvascular thrombosis in vivo.


Subject(s)
P-Selectin , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Thrombosis , Adenosine Diphosphate/metabolism , Animals , Antigens, CD , Blood Platelets/metabolism , Carcinoembryonic Antigen/metabolism , Cell Adhesion Molecules , Collagen/metabolism , Mice , P-Selectin/metabolism , Platelet Activation , Platelet Aggregation , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Receptors, Proteinase-Activated/metabolism , Receptors, Purinergic/metabolism , Thrombosis/genetics , Thrombosis/metabolism
17.
Int J Mol Sci ; 22(19)2021 Oct 02.
Article in English | MEDLINE | ID: mdl-34639054

ABSTRACT

The protease activity in inflammatory bowel disease (IBD) and irritable bowel syndrome has been studied extensively using synthetic fluorogenic substrates targeting specific sets of proteases. We explored activities in colonic tissue from a 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis rat model by investigating the cleavage of bioactive peptides. Pure trypsin- and elastase-like proteases on the one hand and colonic tissue from rats with TNBS-induced colitis in the acute or post-inflammatory phase on the other, were incubated with relevant peptides to identify their cleavage pattern by mass spectrometry. An increased cleavage of several peptides was observed in the colon from acute colitis rats. The tethered ligand (TL) sequences of peptides mimicking the N-terminus of protease-activated receptors (PAR) 1 and 4 were significantly unmasked by acute colitis samples and these cleavages were positively correlated with thrombin activity. Increased cleavage of ß-endorphin and disarming of the TL-sequence of the PAR3-based peptide were observed in acute colitis and linked to chymotrypsin-like activity. Increased processing of the enkephalins points to the involvement of proteases with specificities different from trypsin- or chymotrypsin-like enzymes. In conclusion, our results suggest thrombin, chymotrypsin-like proteases and a set of proteases with different specificities as potential therapeutic targets in IBD.


Subject(s)
Colitis/metabolism , Peptides/metabolism , Receptors, Proteinase-Activated/metabolism , Amino Acid Sequence , Animals , Biomarkers , Colitis/etiology , Colitis/pathology , Disease Models, Animal , Disease Susceptibility , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Male , Peptides/chemistry , Proteolysis , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Tohoku J Exp Med ; 255(1): 1-8, 2021 09.
Article in English | MEDLINE | ID: mdl-34511578

ABSTRACT

Endothelial nitric oxide synthase (eNOS) dysfunction is known to exacerbate the progression and prognosis of diabetic kidney disease (DKD). One of the mechanisms through which this is achieved is that low eNOS levels are associated with hypercoagulability, which promotes kidney injury. In the extrinsic coagulation cascade, the tissue factor (factor III) and downstream coagulation factors, such as active factor X (FXa), exacerbate inflammation through activation of the protease-activated receptors (PARs). Recently, it has been shown that the lack of or reduced eNOS expression in diabetic mice, as a model of advanced DKD, increases renal tissue factor levels and PAR1 and 2 expression in their kidneys. Furthermore, pharmaceutical inhibition or genetic deletion of coagulation factors or PARs ameliorated inflammation in DKD in mice lacking eNOS. In this review, we summarize the relationship between eNOS, coagulation, and PARs and propose a novel therapeutic option for the management of patients with DKD.


Subject(s)
Diabetic Nephropathies/etiology , Nitric Oxide Synthase Type III/deficiency , Receptors, Proteinase-Activated/metabolism , Animals , Antibodies, Neutralizing/administration & dosage , Blood Coagulation , Diabetic Nephropathies/blood , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Factor Xa Inhibitors/pharmacology , Humans , Kidney/drug effects , Kidney/metabolism , Mice , Mice, Knockout , Nitric Oxide Synthase Type III/genetics , Receptors, Proteinase-Activated/deficiency , Receptors, Proteinase-Activated/genetics , Signal Transduction , Thromboplastin/antagonists & inhibitors , Thromboplastin/metabolism
19.
Cells ; 10(7)2021 07 12.
Article in English | MEDLINE | ID: mdl-34359930

ABSTRACT

Chronic spontaneous urticaria (CSU) is a common skin disorder characterized by an almost daily recurrence of wheal and flare with itch for more than 6 weeks, in association with the release of stored inflammatory mediators, such as histamine, from skin mast cells and/or peripheral basophils. The involvement of the extrinsic coagulation cascade triggered by tissue factor (TF) and complement factors, such as C3a and C5a, has been implied in the pathogenesis of CSU. However, it has been unclear how the TF-triggered coagulation pathway and complement factors induce the activation of skin mast cells and peripheral basophils in patients with CSU. In this review, we focus on the role of vascular endothelial cells, leukocytes, extrinsic coagulation factors and complement components on TF-induced activation of skin mast cells and peripheral basophils followed by the edema formation clinically recognized as urticaria. These findings suggest that medications targeting activated coagulation factors and/or complement components may represent new and effective treatments for patients with severe and refractory CSU.


Subject(s)
Blood Coagulation , Chronic Urticaria/blood , Chronic Urticaria/pathology , Complement System Proteins/metabolism , Mast Cells/pathology , Animals , Humans , Leukocytes/metabolism , Receptors, Proteinase-Activated/metabolism
20.
Int J Mol Sci ; 22(16)2021 Aug 20.
Article in English | MEDLINE | ID: mdl-34445691

ABSTRACT

The essential role of G-protein coupled receptors (GPCRs) in tumor growth is recognized, yet a GPCR based drug in cancer is rare. Understanding the molecular path of a tumor driver gene may lead to the design and development of an effective drug. For example, in members of protease-activated receptor (PAR) family (e.g., PAR1 and PAR2), a novel PH-binding motif is allocated as critical for tumor growth. Animal models have indicated the generation of large tumors in the presence of PAR1 or PAR2 oncogenes. These tumors showed effective inhibition when the PH-binding motif was either modified or were inhibited by a specific inhibitor targeted to the PH-binding motif. In the second part of the review we discuss several aspects of some cardinal GPCRs in tumor angiogenesis.


Subject(s)
Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Proteinase-Activated/metabolism , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Neoplasms/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Pleckstrin Homology Domains/genetics , Pleckstrin Homology Domains/physiology , Protein Domains/genetics , Protein Domains/physiology , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Receptors, G-Protein-Coupled/physiology , Receptors, Proteinase-Activated/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...