1
|
Das K, Rao LVM. Coagulation protease-induced extracellular vesicles: their potential effects on coagulation and inflammation. J Thromb Haemost 2024; 22:2976-2990. [PMID: 39127325 DOI: 10.1016/j.jtha.2024.07.022] [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: 04/26/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 08/12/2024]
Abstract
Coagulation proteases, in addition to playing an essential role in blood coagulation, often influence diverse cellular functions by inducing specific signaling pathways via the activation of protease-activated receptors (PARs). PAR activation-induced cellular effects are known to be cell-specific as PARs are expressed selectively in specific cell types. However, a growing body of evidence indicates that coagulation protease-induced PAR activation in a specific cell type could affect cellular responses in other cell types via communicating through extracellular vesicles (EVs) as coagulation protease-induced PAR signaling could promote the release of EVs in various cell types. EVs are membrane-enclosed nanosized vesicles that facilitate intercellular communication by transferring bioactive molecules, such as proteins, lipids, messenger RNAs, and microRNAs, etc., from donor cells to recipient cells. Our recent findings established that factor (F)VIIa promotes the release of EVs from vascular endothelium via endothelial cell protein C receptor-dependent activation of PAR1-mediated biased signaling. FVIIa-released EVs exhibit procoagulant activity and cytoprotective responses in both in vitro and in vivo model systems. This review discusses how FVIIa and other coagulation proteases trigger the release of EVs. The review specifically discusses how FVIIa-released EVs are enriched with phosphatidylserine and anti-inflammatory microRNAs and the impact of FVIIa-released EVs on hemostasis in therapeutic settings. The review also briefly highlights the therapeutic potential of FVIIa-released EVs in treating bleeding and inflammatory disorders, such as hemophilic arthropathy.
Collapse
Affiliation(s)
- Kaushik Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas at Tyler School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, Texas, USA.
| |
Collapse
|
2
|
Ziegler AR, Anderson BM, Latorre R, McQuade RM, Dufour A, Schmidt BL, Bunnett NW, Scott NE, Edgington-Mitchell LE. N-terminomics profiling of naïve and inflamed murine colon reveals proteolytic signatures of legumain. J Cell Physiol 2024:e31466. [PMID: 39392222 DOI: 10.1002/jcp.31466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/27/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Legumain is a cysteine protease broadly associated with inflammation. It has been reported to cleave and activate protease-activated receptor 2 to provoke pain associated with oral cancer. Outside of gastric and colon cancer, little has been reported on the roles of legumain within the gastrointestinal tract. Using a legumain-selective activity-based probe, LE28, we report that legumain is activated within colonocytes and macrophages of the murine colon, and that it is upregulated in models of acute experimental colitis. We demonstrated that loss of legumain activity in colonocytes, either through pharmacological inhibition or gene deletion, had no impact on epithelial permeability in vitro. Moreover, legumain inhibition or deletion had no obvious impacts on symptoms or histological features associated with dextran sulfate sodium-induced colitis, suggesting its proteolytic activity is dispensable for colitis initiation. To gain insight into potential functions of legumain within the colon, we performed field asymmetric waveform ion mobility spectrometry-facilitated quantitative proteomics and N-terminomics analyses on naïve and inflamed colon tissue from wild-type and legumain-deficient mice. We identified 16 altered cleavage sites with an asparaginyl endopeptidase signature that may be direct substrates of legumain and a further 16 cleavage sites that may be indirectly mediated by legumain. We also analyzed changes in protein abundance and proteolytic events broadly associated with colitis in the gut, which permitted comparison to recent analyses on mucosal biopsies from patients with inflammatory bowel disease. Collectively, these results shed light on potential functions of legumain and highlight its potential roles in the transition from inflammation to colorectal cancer.
Collapse
Affiliation(s)
- Alexander R Ziegler
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Bethany M Anderson
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Rocco Latorre
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York, USA
| | - Rachel M McQuade
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria, Australia
| | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Brian L Schmidt
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, Bluestone Center for Clinical Research, New York, New York, USA
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York, USA
| | - Nichollas E Scott
- Department of Microbiology and Immunology, Peter Doherty Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
3
|
Xu K, Wang L, Lin M, He G. Update on protease-activated receptor 2 in inflammatory and autoimmune dermatological diseases. Front Immunol 2024; 15:1449126. [PMID: 39364397 PMCID: PMC11446762 DOI: 10.3389/fimmu.2024.1449126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024] Open
Abstract
Protease-activated receptor 2 (PAR2) is a cell-surface receptor expressed in various cell types, including keratinocytes, neurons, immune and inflammatory cells. Activation of PAR2, whether via its canonical or biased pathways, triggers a series of signaling cascades that mediate numerous functions. This review aims to highlight the emerging roles and interactions of PAR2 in different skin cells. It specifically summarizes the latest insights into the roles of PAR2 in skin conditions such as atopic dermatitis (AD), psoriasis, vitiligo and melasma. It also considers these roles from the perspective of the cutaneous microenvironment in relation to other inflammatory and autoimmune dermatological disorders. Additionally, the review explores PAR2's involvement in associated comorbidities from both cutaneous and extracutaneous diseases. Therefore, PAR2 may serve as a key target for interactions among various cells within the local skin environment.
Collapse
Affiliation(s)
- Kejia Xu
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Mao Lin
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Gu He
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Wang X, Liu Q, Zhuang Z, Cheng J, Zhang W, Jiang Q, Guo Y, Li R, Lu X, Cui L, Weng J, Tang Y, Yue J, Gao S, Hong K, Qiao J, Jiang H, Guo J, Zhang Z. Decoding the pathogenesis of spermatogenic failure in cryptorchidism through single-cell transcriptomic profiling. Cell Rep Med 2024; 5:101709. [PMID: 39226895 PMCID: PMC11528238 DOI: 10.1016/j.xcrm.2024.101709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/20/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024]
Abstract
Cryptorchidism, commonly known as undescended testis, affects 1%-9% of male newborns, posing infertility and testis tumor risks. Despite its prevalence, the detailed pathophysiology underlying male infertility within cryptorchidism remains unclear. Here, we profile and analyze 46,644 single-cell transcriptomes from individual testicular cells obtained from adult males diagnosed with cryptorchidism and healthy controls. Spermatogenesis compromise in cryptorchidism links primarily to spermatogonium self-renewal and differentiation dysfunctions. We illuminate the involvement of testicular somatic cells, including immune cells, thereby unveiling the activation and degranulation of mast cells in cryptorchidism. Mast cells are identified as contributors to interstitial fibrosis via transforming growth factor β1 (TGF-β1) and cathepsin G secretion. Furthermore, significantly increased levels of secretory proteins indicate mast cell activation and testicular fibrosis in the seminal plasma of individuals with cryptorchidism compared to controls. These insights serve as valuable translational references, enriching our comprehension of testicular pathogenesis and informing more precise diagnosis and targeted therapeutic strategies for cryptorchidism.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Qiang Liu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Ziyan Zhuang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Jianxing Cheng
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Wenxiu Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Qiaoling Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yifei Guo
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Ran Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Xiaojian Lu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Lina Cui
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China
| | - Jiaming Weng
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Yanlin Tang
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Jingwei Yue
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Songzhan Gao
- Department of Andrology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kai Hong
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing, China; Department of Urology, Institute of Urology, Peking University First Hospital, Beijing, China.
| | - Jingtao Guo
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China; University of the Chinese Academy of Sciences, Beijing, China.
| | - Zhe Zhang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Department of Urology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
5
|
Slotabec L, Seale B, Wang H, Wen C, Filho F, Rouhi N, Adenawoola MI, Li J. Platelets at the intersection of inflammation and coagulation in the APC-mediated response to myocardial ischemia/reperfusion injury. FASEB J 2024; 38:e23890. [PMID: 39143722 PMCID: PMC11373610 DOI: 10.1096/fj.202401128r] [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: 05/21/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Thromboinflammation is a complex pathology associated with inflammation and coagulation. In cases of cardiovascular disease, in particular ischemia-reperfusion injury, thromboinflammation is a common complication. Increased understanding of thromboinflammation depends on an improved concept of the mechanisms of cells and proteins at the axis of coagulation and inflammation. Among these elements are activated protein C and platelets. This review summarizes the complex interactions of activated protein C and platelets regulating thromboinflammation in cardiovascular disease. By unraveling the pathways of platelets and APC in the inflammatory and coagulation cascades, this review summarizes the role of these vital mediators in the development and perpetuation of heart disease and the thromboinflammation-driven complications of cardiovascular disease. Furthermore, this review emphasizes the significance of the counteracting effects of platelets and APC and their combined role in disease states.
Collapse
Affiliation(s)
- Lily Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Blaise Seale
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Changhong Wen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Fernanda Filho
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael I Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
6
|
Guignard S, Saifeddine M, Mihara K, Motahhary M, Savignac M, Guiraud L, Sagnat D, Sebbag M, Khou S, Rolland C, Edir A, Bournet B, Buscail L, Buscail E, Alric L, Camare C, Ambli M, Vergnolle N, Hollenberg MD, Deraison C, Bonnart C. Chymotrypsin activity signals to intestinal epithelium by protease-activated receptor-dependent mechanisms. Br J Pharmacol 2024; 181:2725-2749. [PMID: 38637276 DOI: 10.1111/bph.16341] [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: 06/29/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND AND PURPOSE Chymotrypsin is a pancreatic protease secreted into the lumen of the small intestine to digest food proteins. We hypothesized that chymotrypsin activity may be found close to epithelial cells and that chymotrypsin signals to them via protease-activated receptors (PARs). We deciphered molecular pharmacological mechanisms and gene expression regulation for chymotrypsin signalling in intestinal epithelial cells. EXPERIMENTAL APPROACH The presence and activity of chymotrypsin were evaluated by Western blot and enzymatic activity tests in the luminal and mucosal compartments of murine and human gut samples. The ability of chymotrypsin to cleave the extracellular domain of PAR1 or PAR2 was assessed using cell lines expressing N-terminally tagged receptors. The cleavage site of chymotrypsin on PAR1 and PAR2 was determined by HPLC-MS analysis. The chymotrypsin signalling mechanism was investigated in CMT93 intestinal epithelial cells by calcium mobilization assays and Western blot analyses of (ERK1/2) phosphorylation. The transcriptional consequences of chymotrypsin signalling were analysed on colonic organoids. KEY RESULTS We found that chymotrypsin was present and active in the vicinity of the colonic epithelium. Molecular pharmacological studies have shown that chymotrypsin cleaves both PAR1 and PAR2 receptors. Chymotrypsin activated calcium and ERK1/2 signalling pathways through PAR2, and this pathway promoted interleukin-10 (IL-10) up-regulation in colonic organoids. In contrast, chymotrypsin disarmed PAR1, preventing further activation by its canonical agonist, thrombin. CONCLUSION AND IMPLICATIONS Our results highlight the ability of chymotrypsin to signal to intestinal epithelial cells via PARs, which may have important physiological consequences in gut homeostasis.
Collapse
Affiliation(s)
- Simon Guignard
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Mahmoud Saifeddine
- Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Koichiro Mihara
- Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Majid Motahhary
- Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Magali Savignac
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291-Centre National de la Recherche Scientifique UMR5051, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Laura Guiraud
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - David Sagnat
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Mireille Sebbag
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Sokchea Khou
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Corinne Rolland
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Anissa Edir
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Barbara Bournet
- Department of Gastroenterology, Toulouse University Hospital, Toulouse, France
| | - Louis Buscail
- Department of Gastroenterology, Toulouse University Hospital, Toulouse, France
| | - Etienne Buscail
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Department of Digestive Surgery, Toulouse University Hospital, Toulouse, France
| | - Laurent Alric
- Department of Internal Medicine and Digestive Diseases, Rangueil, Toulouse III University Hospital, University of Toulouse, Toulouse, France
| | - Caroline Camare
- Department of Clinical Biochemistry, Toulouse University Hospital, Toulouse, France
- University of Toulouse, UMR1297, INSERM/Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Mouna Ambli
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Nathalie Vergnolle
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Morley D Hollenberg
- Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
- Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Céline Deraison
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Chrystelle Bonnart
- IRSD, University of Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| |
Collapse
|
7
|
Healy LD, Fernández JA, Aiolfi R, Mosnier LO, Griffin JH. An orthosteric/allosteric bivalent peptide agonist comprising covalently linked protease-activated receptor-derived peptides mimics in vitro and in vivo activities of activated protein C. J Thromb Haemost 2024; 22:2039-2051. [PMID: 38670314 DOI: 10.1016/j.jtha.2024.04.007] [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: 10/12/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024]
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.
Collapse
Affiliation(s)
- Laura D Healy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - José A Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Roberto Aiolfi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Laurent O Mosnier
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA.
| |
Collapse
|
8
|
Torosian TA, Barsukova AI, Chichkova NV, Vartapetian AB. Phytaspase Does Not Require Proteolytic Activity for Its Stress-Induced Internalization. Int J Mol Sci 2024; 25:6729. [PMID: 38928451 PMCID: PMC11203471 DOI: 10.3390/ijms25126729] [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: 05/04/2024] [Revised: 05/31/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Phytaspases differ from other members of the plant subtilisin-like protease family by having rare aspartate cleavage specificity and unusual localization dynamics. Phytaspases are secreted from healthy plant cells but are re-internalized upon perception of death-inducing stresses. Although proteolytic activity is required for the secretion of plant subtilases, its requirement for the retrograde transportation of phytaspases is currently unknown. To address this issue, we employed an approach to complement in trans the externalization of a prodomain-less form of Nicotiana tabacum phytaspase (NtPhyt) with the free prodomain in Nicotiana benthamiana leaf cells. Using this approach, the generation of the proteolytically active NtPhyt and its transport to the extracellular space at a level comparable to that of the native NtPhyt (synthesized as a canonical prodomain-containing precursor protein) were achieved. The application of this methodology to NtPhyt with a mutated catalytic Ser537 residue resulted in the secretion of the inactive, although processed (prodomain-free), protein as well. Notably, the externalized NtPhyt Ser537Ala mutant was still capable of retrograde transportation into plant cells upon the induction of oxidative stress. Our data thus indicate that the proteolytic activity of NtPhyt is dispensable for stress-induced retrograde transport of the enzyme.
Collapse
Affiliation(s)
- Tatevik A. Torosian
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 199991, Russia; (T.A.T.); (A.I.B.)
| | - Anastasia I. Barsukova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 199991, Russia; (T.A.T.); (A.I.B.)
| | - Nina V. Chichkova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 199991, Russia;
| | - Andrey B. Vartapetian
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 199991, Russia;
| |
Collapse
|
9
|
Radisky ES. Extracellular proteolysis in cancer: Proteases, substrates, and mechanisms in tumor progression and metastasis. J Biol Chem 2024; 300:107347. [PMID: 38718867 PMCID: PMC11170211 DOI: 10.1016/j.jbc.2024.107347] [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: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024] Open
Abstract
A vast ensemble of extracellular proteins influences the development and progression of cancer, shaped and reshaped by a complex network of extracellular proteases. These proteases, belonging to the distinct classes of metalloproteases, serine proteases, cysteine proteases, and aspartic proteases, play a critical role in cancer. They often become dysregulated in cancer, with increases in pathological protease activity frequently driven by the loss of normal latency controls, diminished regulation by endogenous protease inhibitors, and changes in localization. Dysregulated proteases accelerate tumor progression and metastasis by degrading protein barriers within the extracellular matrix (ECM), stimulating tumor growth, reactivating dormant tumor cells, facilitating tumor cell escape from immune surveillance, and shifting stromal cells toward cancer-promoting behaviors through the precise proteolysis of specific substrates to alter their functions. These crucial substrates include ECM proteins and proteoglycans, soluble proteins secreted by tumor and stromal cells, and extracellular domains of cell surface proteins, including membrane receptors and adhesion proteins. The complexity of the extracellular protease web presents a significant challenge to untangle. Nevertheless, technological strides in proteomics, chemical biology, and the development of new probes and reagents are enabling progress and advancing our understanding of the pivotal importance of extracellular proteolysis in cancer.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| |
Collapse
|
10
|
Ye S, Agalave NM, Ma F, Mahmood DFD, Al-Grety A, Khoonsari PE, Leng L, Svensson CI, Bucala R, Kultima K, Vera PL. MIF-Modulated Spinal Proteins Associated with Persistent Bladder Pain: A Proteomics Study. Int J Mol Sci 2024; 25:4484. [PMID: 38674069 PMCID: PMC11050327 DOI: 10.3390/ijms25084484] [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: 03/04/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Bladder pain is a prominent symptom in Interstitial Cystitis/Bladder Pain Syndrome (IC/BPS). We studied spinal mechanisms of bladder pain in mice using a model where repeated activation of intravesical Protease Activated Receptor-4 (PAR4) results in persistent bladder hyperalgesia (BHA) with little or no bladder inflammation. Persistent BHA is mediated by spinal macrophage migration inhibitory factor (MIF), and is associated with changes in lumbosacral proteomics. We investigated the contribution of individual spinal MIF receptors to persistent bladder pain as well as the spinal proteomics changes associated with relief of persistent BHA by spinal MIF antagonism. Female mice with persistent BHA received either intrathecal (i.t.) MIF monoclonal antibodies (mAb) or mouse IgG1 (isotype control antibody). MIF antagonism temporarily reversed persistent BHA (peak effect: 2 h), while control IgG1 had no effect. Moreover, i.t. antagonism of the MIF receptors CD74 and C-X-C chemokine receptor type 4 (CXCR4) partially reversed persistent BHA. For proteomics experiments, four separate groups of mice received either repeated intravesical scrambled peptide and sham i.t. injection (control, no pain group) or repeated intravesical PAR4 and: sham i.t.; isotype IgG1 i.t. (15 μg); or MIF mAb (15 μg). L6-S1 spinal segments were excised 2 h post-injection and examined for proteomics changes using LC-MS/MS. Unbiased proteomics analysis identified and relatively quantified 6739 proteins. We selected proteins that showed significant changes compared to control (no pain group) after intravesical PAR4 (sham or IgG i.t. treatment) and showed no significant change after i.t. MIF antagonism. Six proteins decreased during persistent BHA (V-set transmembrane domain-containing protein 2-like confirmed by immunohistochemistry), while two proteins increased. Spinal MIF antagonism reversed protein changes. Therefore, spinal MIF and MIF receptors mediate persistent BHA and changes in specific spinal proteins. These novel MIF-modulated spinal proteins represent possible new targets to disrupt spinal mechanisms that mediate persistent bladder pain.
Collapse
Affiliation(s)
- Shaojing Ye
- Research & Development, Lexington VA Health Care System, Lexington, KY 40502, USA; (S.Y.); (F.M.); (D.F.D.M.)
| | - Nilesh M. Agalave
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, SE-751 85 Uppsala, Sweden; (N.M.A.); (A.A.-G.); (P.E.K.); (K.K.)
| | - Fei Ma
- Research & Development, Lexington VA Health Care System, Lexington, KY 40502, USA; (S.Y.); (F.M.); (D.F.D.M.)
| | - Dlovan F. D. Mahmood
- Research & Development, Lexington VA Health Care System, Lexington, KY 40502, USA; (S.Y.); (F.M.); (D.F.D.M.)
| | - Asma Al-Grety
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, SE-751 85 Uppsala, Sweden; (N.M.A.); (A.A.-G.); (P.E.K.); (K.K.)
| | - Payam E. Khoonsari
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, SE-751 85 Uppsala, Sweden; (N.M.A.); (A.A.-G.); (P.E.K.); (K.K.)
| | - Lin Leng
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA; (L.L.); (R.B.)
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Karolinska Institutet (KI), SE-171 65 Solna, Sweden;
| | - Richard Bucala
- Department of Internal Medicine, Yale University, New Haven, CT 06510, USA; (L.L.); (R.B.)
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, SE-751 85 Uppsala, Sweden; (N.M.A.); (A.A.-G.); (P.E.K.); (K.K.)
| | - Pedro L. Vera
- Research & Development, Lexington VA Health Care System, Lexington, KY 40502, USA; (S.Y.); (F.M.); (D.F.D.M.)
- Department of Physiology, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
11
|
Li X, Kurahara LH, Zhao Z, Zhao F, Ishikawa R, Ohmichi K, Li G, Yamashita T, Hashimoto T, Hirano M, Sun Z, Hirano K. Therapeutic Effect of Proteinase-Activated Receptor-1 Antagonist on Colitis-Associated Carcinogenesis. Cell Mol Gastroenterol Hepatol 2024; 18:105-131. [PMID: 38614455 PMCID: PMC11127032 DOI: 10.1016/j.jcmgh.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/30/2024] [Accepted: 04/03/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND & AIMS Inflammatory bowel disease is associated with carcinogenesis, which limits the prognosis of the patients. The local expression of proteinases and proteinase-activated receptor 1 (PAR1) increases in inflammatory bowel disease. The present study investigated the therapeutic effects of PAR1 antagonism on colitis-associated carcinogenesis. METHODS A colitis-associated carcinogenesis model was prepared in mice by treatment with azoxymethane (AOM) and dextran sulfate sodium (DSS). PAR1 antagonist E5555 was administered in long- and short-term protocol, starting on the day of AOM injection and 1 week after completing AOM/DSS treatment, respectively. The fecal samples were collected for metagenome analysis of gut microbiota. The intestinal myofibroblasts of the Crohn's disease patients were used to elucidate underlying cellular mechanisms. Caco-2 cells were used to investigate a possible source of PAR1 agonist proteinases. RESULTS AOM/DSS model showed weight loss, diarrhea, tumor development, inflammation, fibrosis, and increased production of inflammatory cytokines. The β-diversity, but not α-diversity, of microbiota significantly differed between AOM/DSS and control mice. E5555 alleviated these pathological changes and altered the microbiota β-diversity in AOM/DSS mice. The thrombin expression was up-regulated in tumor and non-tumor areas, whereas PAR1 mRNA expression was higher in tumor areas compared with non-tumor areas. E5555 inhibited thrombin-triggered elevation of cytosolic Ca2+ concentration and ERK1/2 phosphorylation, as well as IL6-induced signal transducer and activator of transcription 3 (STAT3) phosphorylation in intestinal myofibroblasts. Caco-2 cell-conditioned medium contained immunoreactive thrombin, which cleaved the recombinant protein containing the extracellular domain of PAR1 at the thrombin cleavage site. CONCLUSIONS PAR1 antagonism is proposed to be a novel therapeutic strategy for treatment of inflammatory bowel disease and its associated carcinogenesis.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Lin-Hai Kurahara
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan.
| | - Zhixin Zhao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Feiyan Zhao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Ryo Ishikawa
- Department of Diagnostic Pathology, Kagawa University Hospital, Kagawa University, Kagawa, Japan
| | - Kiyomi Ohmichi
- Department of Diagnostic Pathology, Kagawa University Hospital, Kagawa University, Kagawa, Japan
| | - Gaopeng Li
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Tetsuo Yamashita
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takeshi Hashimoto
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Mayumi Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| |
Collapse
|
12
|
Hou JJ, Ding L, Yang T, Yang YF, Jin YP, Zhang XP, Ma AH, Qin YH. The proteolytic activity in inflammatory bowel disease: insight from gut microbiota. Microb Pathog 2024; 188:106560. [PMID: 38272327 DOI: 10.1016/j.micpath.2024.106560] [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: 08/02/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic, recurrent inflammatory disease caused by the destruction of the intestinal mucosal epithelium that affects a growing number of people worldwide. Although the etiology of IBD is complex and still elucidated, the role of dysbiosis and dysregulated proteolysis is well recognized. Various studies observed altered composition and diversity of gut microbiota, as well as increased proteolytic activity (PA) in serum, plasma, colonic mucosa, and fecal supernatant of IBD compared to healthy individuals. The imbalance of intestinal microecology and intestinal protein hydrolysis were gradually considered to be closely related to IBD. Notably, the pivotal role of intestinal microbiota in maintaining proteolytic balance received increasing attention. In summary, we have speculated a mesmerizing story, regarding the hidden role of PA and microbiota-derived PA hidden in IBD. Most importantly, we provided the diagnosis and therapeutic targets for IBD as well as the formulation of new treatment strategies for other digestive diseases and protease-related diseases.
Collapse
Affiliation(s)
- Jun-Jie Hou
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Liang Ding
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Tao Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yan-Fei Yang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Ping Jin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Xiao-Ping Zhang
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - A-Huo Ma
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China
| | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People's Hospital, Shaoxing, PR China.
| |
Collapse
|
13
|
Hirano K. New Mechanistic Insight Into Biased Signaling of Proteinase-Activated Receptor 1. Arterioscler Thromb Vasc Biol 2024; 44:617-619. [PMID: 38269587 DOI: 10.1161/atvbaha.123.320616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Affiliation(s)
- Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Japan
| |
Collapse
|
14
|
Chen J, Ren T, Xie L, Hu H, Li X, Maitusong M, Zhou X, Hu W, Xu D, Qian Y, Cheng S, Yu K, Wang JA, Liu X. Enhancing aortic valve drug delivery with PAR2-targeting magnetic nano-cargoes for calcification alleviation. Nat Commun 2024; 15:557. [PMID: 38228638 PMCID: PMC10792006 DOI: 10.1038/s41467-024-44726-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
Calcific aortic valve disease is a prevalent cardiovascular disease with no available drugs capable of effectively preventing its progression. Hence, an efficient drug delivery system could serve as a valuable tool in drug screening and potentially enhance therapeutic efficacy. However, due to the rapid blood flow rate associated with aortic valve stenosis and the lack of specific markers, achieving targeted drug delivery for calcific aortic valve disease has proved to be challenging. Here we find that protease-activated-receptor 2 (PAR2) expression is up-regulated on the plasma membrane of osteogenically differentiated valvular interstitial cells. Accordingly, we develop a magnetic nanocarrier functionalized with PAR2-targeting hexapeptide for dual-active targeting drug delivery. We show that the nanocarriers effectively deliver XCT790-an anti-calcification drug-to the calcified aortic valve under extra magnetic field navigation. We demonstrate that the nano-cargoes consequently inhibit the osteogenic differentiation of valvular interstitial cells, and alleviate aortic valve calcification and stenosis in a high-fat diet-fed low-density lipoprotein receptor-deficient (Ldlr-/-) mouse model. This work combining PAR2- and magnetic-targeting presents an effective targeted drug delivery system for treating calcific aortic valve disease in a murine model, promising future clinical translation.
Collapse
Affiliation(s)
- Jinyong Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Tanchen Ren
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China.
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China.
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China.
| | - Lan Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Haochang Hu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Xu Li
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, 200030, Shanghai, P.R. China
| | - Miribani Maitusong
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Xuhao Zhou
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Wangxing Hu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Dilin Xu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Yi Qian
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Si Cheng
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Kaixiang Yu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China
| | - Jian An Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China.
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China.
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, P.R. China.
| | - Xianbao Liu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, P.R. China.
- State Key Laboratory of Transvascular Implantation Devices, 310009, Hangzhou, P.R. China.
- Cardiovascular Key Laboratory of Zhejiang Province, 310009, Hangzhou, P.R. China.
| |
Collapse
|
15
|
Eftekhari R, Ewanchuk BW, Rawji KS, Yates RM, Noorbakhsh F, Kuipers HF, Hollenberg MD. Blockade of Proteinase-Activated Receptor 2 (PAR2) Attenuates Neuroinflammation in Experimental Autoimmune Encephalomyelitis. J Pharmacol Exp Ther 2024; 388:12-22. [PMID: 37699708 DOI: 10.1124/jpet.123.001685] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/17/2023] [Accepted: 08/08/2023] [Indexed: 09/14/2023] Open
Abstract
Proteinase-activated receptor-2 (PAR2), which modulates inflammatory responses, is elevated in the central nervous system in multiple sclerosis (MS) and in its murine model, experimental autoimmune encephalomyelitis (EAE). In PAR2-null mice, disease severity of EAE is markedly diminished. We therefore tested whether inhibiting PAR2 activation in vivo might be a viable strategy for the treatment of MS. Using the EAE model, we show that a PAR2 antagonist, the pepducin palmitoyl-RSSAMDENSEKKRKSAIK-amide (P2pal-18S), attenuates EAE progression by affecting immune cell function. P2pal-18S treatment markedly diminishes disease severity and reduces demyelination, as well as the infiltration of T-cells and macrophages into the central nervous system. Moreover, P2pal-18S decreases granulocyte-macrophage colony-stimulating factor (GM-CSF) production and T-cell activation in cultured splenocytes and prevents macrophage polarization in vitro. We conclude that PAR2 plays a key role in regulating neuroinflammation in EAE and that PAR2 antagonists represent promising therapeutic agents for treating MS and other neuroinflammatory diseases. SIGNIFICANCE STATEMENT: Proteinase-activated receptor-2 modulates inflammatory responses and is increased in multiple sclerosis lesions. We show that the proteinase-activated receptor-2 antagonist palmitoyl-RSSAMDENSEKKRKSAIK-amide reduces disease in the murine experimental autoimmune encephalomyelitis model of multiple sclerosis by inhibiting T-cell and macrophage activation and infiltration into the central nervous system, making it a potential treatment for multiple sclerosis.
Collapse
Affiliation(s)
- Rahil Eftekhari
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Benjamin W Ewanchuk
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Khalil S Rawji
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Robin M Yates
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Farshid Noorbakhsh
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Hedwich F Kuipers
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| |
Collapse
|
16
|
Scarisbrick IA. PARting Neuroinflammation with Protease-Activated Receptor 2 Pepducins. J Pharmacol Exp Ther 2024; 388:8-11. [PMID: 38169447 DOI: 10.1124/jpet.123.001893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/26/2023] [Indexed: 01/05/2024] Open
Affiliation(s)
- Isobel A Scarisbrick
- Center for Regenerative Biotherapeutics, Department of Physical Medicine and Rehabilitation, and Department of Physiology and Biomedical Engineering, Mayo Clinic Graduate School of Biomedical Sciences, Regenerative Sciences, Rochester, Minnesota
| |
Collapse
|
17
|
Ma J, Liu Y, Yuan J, Ma Y, Zhao X, Chen K, Zhang X, Zhang F, Wang H. Bcl-xL mediates interferon-beta secretion by protease-activated receptor 2 deficiency through the mitochondrial permeability transition pore in colorectal cancer metastasis. Cancer Lett 2024; 580:216483. [PMID: 37972702 DOI: 10.1016/j.canlet.2023.216483] [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: 07/05/2023] [Revised: 10/27/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Cellular plasticity and immune escape are synergistic drivers of tumor colonization in metastatic organs. Activation of protease-activated receptor 2 (PAR2) signaling promotes metastasis of colorectal carcinoma (CRC). The role of PAR2 in regulating the immune microenvironment and cancer progression remains unclear. We demonstrated that the regulation of liver metastasis by PAR2 requires a competent immune system. PAR2 knockdown enhanced liver infiltration of activated CD8+ T cells prior to metastatic foci formation in an interferon receptor-dependent manner. PAR2 depletion increased interferon (IFN)-β production via the cGAS-STING and RIG-1 pathways. PAR2 inhibition increased mitochondrial permeability and cytosolic accumulation of mitochondrial DNA, which was reversed by Bcl-xL expression. Strikingly, shRNA against PAR2 with an immune checkpoint blocker (ICB) acted synergistically to suppress liver metastasis. Analysis of single-cell sequence data and 24 paired samples confirmed the regulatory effect of PAR2 on the metastatic immune environment in human CRC. Therefore, PAR2 signaling is involved in stabilizing the mitochondrial membrane and regulating the immune microenvironment through IFN-β during liver metastasis in CRC. The synergistic effect of the PAR2 inhibitor and ICB provides a potential therapeutic strategy for metastatic CRC treatment.
Collapse
Affiliation(s)
- Jianhui Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yu Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Junhu Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xinhua Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Kun Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiaoli Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Department of Injury and Repair, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Fanyu Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| |
Collapse
|
18
|
Julovi SM, McKelvey K, Minhas N, Chan YKA, Xue M, Jackson CJ. Involvement of PAR-2 in the Induction of Cell-Specific Matrix Metalloproteinase-2 by Activated Protein C in Cutaneous Wound Healing. Int J Mol Sci 2023; 25:370. [PMID: 38203540 PMCID: PMC10779272 DOI: 10.3390/ijms25010370] [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: 11/25/2023] [Revised: 12/23/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
We previously reported that human keratinocytes express protease-activated receptor (PAR)-2 and play an important role in activated protein C (APC)-induced cutaneous wound healing. This study investigated the involvement of PAR-2 in the production of gelatinolytic matrix metalloproteinases (MMP)-2 and -9 by APC during cutaneous wound healing. Full-thickness excisional wounds were made on the dorsum of male C57BL/6 mice. Wounds were treated with APC on days 1, 2, and 3 post-wounding. Cultured neonatal foreskin keratinocytes were treated with APC with or without intact PAR-2 signalling to examine the effects on MMP-2 and MMP-9 production. Murine dermal fibroblasts from PAR-2 knock-out (KO) mice were also assessed. MMP-2 and -9 were measured via gelatin zymography, fluorometric assay, and immunohistochemistry. APC accelerated wound healing in WT mice, but had a negligible effect in PAR-2 KO mice. APC-stimulated murine cutaneous wound healing was associated with the differential and temporal production of MMP-2 and MMP-9, with the latter peaking on day 1 and the former on day 6. Inhibition of PAR-2 in human keratinocytes reduced APC-induced MMP-2 activity by 25~50%, but had little effect on MMP-9. Similarly, APC-induced MMP-2 activation was reduced by 40% in cultured dermal fibroblasts derived from PAR-2 KO mice. This study shows for the first time that PAR-2 is essential for APC-induced MMP-2 production. Considering the important role of MMP-2 in wound healing, this work helps explain the underlying mechanisms of action of APC to promote wound healing through PAR-2.
Collapse
Affiliation(s)
- Sohel M. Julovi
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Pacific Highway, St. Leonards, NSW 2065, Australia; (K.M.); (N.M.); (Y.-K.A.C.); (M.X.); (C.J.J.)
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, NSW 2045, Australia
| | - Kelly McKelvey
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Pacific Highway, St. Leonards, NSW 2065, Australia; (K.M.); (N.M.); (Y.-K.A.C.); (M.X.); (C.J.J.)
| | - Nikita Minhas
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Pacific Highway, St. Leonards, NSW 2065, Australia; (K.M.); (N.M.); (Y.-K.A.C.); (M.X.); (C.J.J.)
| | - Yee-Ka Agnes Chan
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Pacific Highway, St. Leonards, NSW 2065, Australia; (K.M.); (N.M.); (Y.-K.A.C.); (M.X.); (C.J.J.)
| | - Meilang Xue
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Pacific Highway, St. Leonards, NSW 2065, Australia; (K.M.); (N.M.); (Y.-K.A.C.); (M.X.); (C.J.J.)
| | - Christopher J. Jackson
- Sutton Arthritis Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Pacific Highway, St. Leonards, NSW 2065, Australia; (K.M.); (N.M.); (Y.-K.A.C.); (M.X.); (C.J.J.)
| |
Collapse
|
19
|
Parekh AB. House dust mite allergens, store-operated Ca 2+ channels and asthma. J Physiol 2023. [PMID: 38054814 DOI: 10.1113/jp284931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
The house dust mite is the principal source of aero-allergen worldwide. Exposure to mite-derived allergens is associated with the development of asthma in susceptible individuals, and the majority of asthmatics are allergic to the mite. Mite-derived allergens are functionally diverse and activate multiple cell types within the lung that result in chronic inflammation. Allergens activate store-operated Ca2+ release-activated Ca2+ (CRAC) channels, which are widely expressed in multiple cell types within the lung that are associated with the pathogenesis of asthma. Opening of CRAC channels stimulates Ca2+ -dependent transcription factors, including nuclear factor of activated T cells and nuclear factor-κB, which drive expression of a plethora of pro-inflammatory cytokines and chemokines that help to sustain chronic inflammation. Here, I describe drivers of asthma, properties of mite-derived allergens, how the allergens are recognized by cells, the signalling pathways used by the receptors and how these are transduced into functional effects, with a focus on CRAC channels. In vivo experiments that demonstrate the effectiveness of targeting CRAC channels as a potential new therapy for treating mite-induced asthma are also discussed, in tandem with other possible approaches.
Collapse
Affiliation(s)
- Anant B Parekh
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| |
Collapse
|
20
|
Lee SK, Malik RA, Zhou J, Wang W, Gross PL, Weitz JI, Ramachandran R, Trigatti BL. PAR4 Inhibition Reduces Coronary Artery Atherosclerosis and Myocardial Fibrosis in SR-B1/LDLR Double Knockout Mice. Arterioscler Thromb Vasc Biol 2023; 43:2165-2178. [PMID: 37675637 PMCID: PMC10597419 DOI: 10.1161/atvbaha.123.319767] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND SR-B1 (scavenger receptor class B type 1)/LDLR (low-density lipoprotein receptor) double knockout mice fed a high-fat, high-cholesterol diet containing cholate exhibit coronary artery disease characterized by occlusive coronary artery atherosclerosis, platelet accumulation in coronary arteries, and myocardial fibrosis. Platelets are involved in atherosclerosis development, and PAR (protease-activated receptor) 4 has a prominent role in platelet function in mice. However, the role of PAR4 on coronary artery disease in mice has not been tested. METHODS We tested the effects of a PAR4 inhibitory pepducin (RAG8) on diet-induced aortic sinus and coronary artery atherosclerosis, platelet accumulation in atherosclerotic coronary arteries, and myocardial fibrosis in SR-B1/LDLR double knockout mice. SR-B1/LDLR double knockout mice were fed a high-fat, high-cholesterol diet containing cholate and injected daily with 20 mg/kg of either the RAG8 pepducin or a control reverse-sequence pepducin (SRQ8) for 20 days. RESULTS Platelets from the RAG8-treated mice exhibited reduced thrombin and PAR4 agonist peptide-mediated activation compared with those from control SRQ8-treated mice when tested ex vivo. Although aortic sinus atherosclerosis levels did not differ, RAG8-treated mice exhibited reduced coronary artery atherosclerosis, reduced platelet accumulation in atherosclerotic coronary arteries, and reduced myocardial fibrosis. These protective effects were not accompanied by changes in circulating lipids, inflammatory cytokines, or immune cells. However, RAG8-treated mice exhibited reduced VCAM-1 (vascular cell adhesion molecule 1) protein levels in nonatherosclerotic coronary artery cross sections and reduced leukocyte accumulation in atherosclerotic coronary artery cross sections compared with those from SRQ8-treated mice. CONCLUSIONS The PAR4 inhibitory RAG8 pepducin reduced coronary artery atherosclerosis and myocardial fibrosis in SR-B1/LDLR double knockout mice fed a high-fat, high-cholesterol diet containing cholate. Furthermore, RAG8 reduced VCAM-1 in nonatherosclerotic coronary arteries and reduced leukocyte and platelet accumulation in atherosclerotic coronary arteries. These findings identify PAR4 as an attractive target in reducing coronary artery disease development, and the use of RAG8 may potentially be beneficial in cardiovascular disease.
Collapse
Affiliation(s)
- Samuel K. Lee
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| | - Rida A. Malik
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
| | - Ji Zhou
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
| | - Wei Wang
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| | - Peter L. Gross
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
| | - Jeffrey I. Weitz
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.R.)
| | - Bernardo L. Trigatti
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| |
Collapse
|
21
|
Li F, Wang C, Guo X, Akutsu T, Webb GI, Coin LJM, Kurgan L, Song J. ProsperousPlus: a one-stop and comprehensive platform for accurate protease-specific substrate cleavage prediction and machine-learning model construction. Brief Bioinform 2023; 24:bbad372. [PMID: 37874948 DOI: 10.1093/bib/bbad372] [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: 07/30/2023] [Revised: 08/30/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023] Open
Abstract
Proteases contribute to a broad spectrum of cellular functions. Given a relatively limited amount of experimental data, developing accurate sequence-based predictors of substrate cleavage sites facilitates a better understanding of protease functions and substrate specificity. While many protease-specific predictors of substrate cleavage sites were developed, these efforts are outpaced by the growth of the protease substrate cleavage data. In particular, since data for 100+ protease types are available and this number continues to grow, it becomes impractical to publish predictors for new protease types, and instead it might be better to provide a computational platform that helps users to quickly and efficiently build predictors that address their specific needs. To this end, we conceptualized, developed, tested and released a versatile bioinformatics platform, ProsperousPlus, that empowers users, even those with no programming or little bioinformatics background, to build fast and accurate predictors of substrate cleavage sites. ProsperousPlus facilitates the use of the rapidly accumulating substrate cleavage data to train, empirically assess and deploy predictive models for user-selected substrate types. Benchmarking tests on test datasets show that our platform produces predictors that on average exceed the predictive performance of current state-of-the-art approaches. ProsperousPlus is available as a webserver and a stand-alone software package at http://prosperousplus.unimelb-biotools.cloud.edu.au/.
Collapse
Affiliation(s)
- Fuyi Li
- College of Information Engineering, Northwest A&F University, Shaanxi 712100, China
- South Australian immunoGENomics Cancer Institute (SAiGENCI), Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC 3000, Australia
| | - Cong Wang
- College of Information Engineering, Northwest A&F University, Shaanxi 712100, China
| | - Xudong Guo
- College of Information Engineering, Northwest A&F University, Shaanxi 712100, China
| | - Tatsuya Akutsu
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Kyoto 611-0011, Japan
| | - Geoffrey I Webb
- Monash Data Futures Institute, Monash University, VIC 3800, Australia
| | - Lachlan J M Coin
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, VIC 3000, Australia
| | - Lukasz Kurgan
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Jiangning Song
- Monash Data Futures Institute, Monash University, VIC 3800, Australia
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia
| |
Collapse
|
22
|
Kuzmina IV, Ovchinnikova NV, Tolpygo SM. Serum Activity of Proteolytic Enzyme Trypsin in Rats under Conditions of Water and Food Deprivation. Bull Exp Biol Med 2023; 175:608-611. [PMID: 37861909 DOI: 10.1007/s10517-023-05910-2] [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: 02/20/2023] [Indexed: 10/21/2023]
Abstract
Trypsin is mainly regarded as a digestive enzyme, but there is evidence that activation of protease-activated receptor-2 (PAR-2) leads to behavioral changes. There are no data on trypsin activity in the serum of animals under conditions of thirst and starvation in the available literature. In our experiments, water deprivation led to a significant (p⩽0.05) increase in trypsin activity in rats, and food deprivation led to its decrease in comparison with controls (free access to water and food). After deprived rats received water and food, a decrease in trypsin activity was observed in both experimental groups. Changes in trypsin activity under conditions of water or food deprivation and after satiation were accompanied by shifts in some biochemical parameters of the bloods. Under conditions of metabolic stress (starvation and thirst), opposite changes in trypsin activity seem to indicate its participation in the mechanisms of adequate restructuring of metabolism and maintenance of vital processes in the body.
Collapse
Affiliation(s)
- I V Kuzmina
- P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia.
| | - N V Ovchinnikova
- P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia
| | - S M Tolpygo
- P. K. Anokhin Research Institute of Normal Physiology, Moscow, Russia
| |
Collapse
|
23
|
Lee I, Tantisirivat P, Edgington-Mitchell LE. Chemical Tools to Image the Activity of PAR-Cleaving Proteases. ACS BIO & MED CHEM AU 2023; 3:295-304. [PMID: 37599791 PMCID: PMC10436261 DOI: 10.1021/acsbiomedchemau.3c00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 08/22/2023]
Abstract
Protease-activated receptors (PARs) comprise a family of four G protein-coupled receptors (GPCRs) that have broad functions in health and disease. Unlike most GPCRs, PARs are uniquely activated by proteolytic cleavage of their extracellular N termini. To fully understand PAR activation and function in vivo, it is critical to also study the proteases that activate them. As proteases are heavily regulated at the post-translational level, measures of total protease abundance have limited utility. Measures of protease activity are instead required to inform their function. This review will introduce several classes of chemical probes that have been developed to measure the activation of PAR-cleaving proteases. Their strengths, weaknesses, and applications will be discussed, especially as applied to image protease activity at the whole organism, tissue, and cellular level.
Collapse
Affiliation(s)
- Irene
Y. Lee
- Department
of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology
Institute, The University of Melbourne, Parkville, Victoria 3052 Australia
| | - Piyapa Tantisirivat
- Department
of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology
Institute, The University of Melbourne, Parkville, Victoria 3052 Australia
| | - Laura E. Edgington-Mitchell
- Department
of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology
Institute, The University of Melbourne, Parkville, Victoria 3052 Australia
| |
Collapse
|
24
|
Airola C, Pallozzi M, Cerrito L, Santopaolo F, Stella L, Gasbarrini A, Ponziani FR. Microvascular Thrombosis and Liver Fibrosis Progression: Mechanisms and Clinical Applications. Cells 2023; 12:1712. [PMID: 37443746 PMCID: PMC10341358 DOI: 10.3390/cells12131712] [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: 05/09/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Fibrosis is an unavoidable consequence of chronic inflammation. Extracellular matrix deposition by fibroblasts, stimulated by multiple pathways, is the first step in the onset of chronic liver disease, and its propagation promotes liver dysfunction. At the same time, chronic liver disease is characterized by alterations in primary and secondary hemostasis but unlike previously thought, these changes are not associated with an increased risk of bleeding complications. In recent years, the role of coagulation imbalance has been postulated as one of the main mechanisms promoting hepatic fibrogenesis. In this review, we aim to investigate the function of microvascular thrombosis in the progression of liver disease and highlight the molecular and cellular networks linking hemostasis to fibrosis in this context. We analyze the predictive and prognostic role of coagulation products as biomarkers of liver decompensation (ascites, variceal hemorrhage, and hepatic encephalopathy) and liver-related mortality. Finally, we evaluate the current evidence on the application of antiplatelet and anticoagulant therapies for prophylaxis of hepatic decompensation or prevention of the progression of liver fibrosis.
Collapse
Affiliation(s)
- Carlo Airola
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Maria Pallozzi
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Lucia Cerrito
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Francesco Santopaolo
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Leonardo Stella
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
| | - Antonio Gasbarrini
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Hepatology Unit, CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (M.P.); (L.C.); (F.S.); (L.S.); (A.G.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
25
|
Huang S, Cao G, Dai D, Xu Q, Ruiz S, Shindo S, Nakamura S, Kawai T, Lin J, Han X. Porphyromonas gingivalis outer membrane vesicles exacerbate retinal microvascular endothelial cell dysfunction in diabetic retinopathy. Front Microbiol 2023; 14:1167160. [PMID: 37250057 PMCID: PMC10213754 DOI: 10.3389/fmicb.2023.1167160] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/14/2023] [Indexed: 05/31/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the leading causes of blindness. Periodontitis is one of the highest oral incidences and has been closely related to various systemic conditions through Porphyromonas gingivalis (P. gingivalis). P. gingivalis OMVs, derived from P. gingivalis, can cause endothelial dysfunction and potentially affect microvascular diseases. Current epidemiological studies provide limited evidence suggesting that periodontitis is associated with DR. However, there is a lack of basic research elucidating how periodontitis affects the severity of DR. This study aimed to explore the potential of P. gingivalis OMVs to contribute to the pathogenesis of DR and explore how it affect the retinal microvascular endothelium. The results demonstrated that P. gingivalis OMVs accelerated the blood-retinal barrier damage in DR mice. In vitro studies showed that the expression of inflammatory factors in human retinal microvascular endothelial cells (HRMECs) was increased after P. gingivalis OMVs stimulation, and the increased reactive oxygen species production, mitochondrial dysfunction, apoptosis, and altered endothelial permeability were observed in HRMECs under P. gingivalis OMVs stimulation. In addition, we found that protease-activated receptor-2 (PAR-2) regulated OMVs-induced TNF-α, MMP-9 mRNA expression, cell death, and endothelial permeability. Overall, we suggested that P. gingivalis OMVs induced mitochondria-related cell death of HRMECs and accelerated endothelial dysfunction, thus aggravating DR, in which PAR-2 plays a potential role. This study is the first research report to delineate the potential molecular mechanism of P. gingivalis OMVs on DR pathogenesis, which uniquely focused on elucidating the possible impact of periodontal pathogen derivatives on DR progression.
Collapse
Affiliation(s)
- Shengyuan Huang
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Guoqin Cao
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Dong Dai
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qiuping Xu
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Sunniva Ruiz
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Satoru Shindo
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Shin Nakamura
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Toshihisa Kawai
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Jiang Lin
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaozhe Han
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| |
Collapse
|
26
|
Polito MP, Marini G, Palamenghi M, Enzo E. Decoding the Human Epidermal Complexity at Single-Cell Resolution. Int J Mol Sci 2023; 24:ijms24108544. [PMID: 37239891 DOI: 10.3390/ijms24108544] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
The epidermis is one of the largest tissues in the human body, serving as a protective barrier. The basal layer of the epidermis, which consists of epithelial stem cells and transient amplifying progenitors, represents its proliferative compartment. As keratinocytes migrate from the basal layer to the skin surface, they exit the cell cycle and initiate terminal differentiation, ultimately generating the suprabasal epidermal layers. A deeper understanding of the molecular mechanisms and pathways driving keratinocytes' organization and regeneration is essential for successful therapeutic approaches. Single-cell techniques are valuable tools for studying molecular heterogeneity. The high-resolution characterization obtained with these technologies has identified disease-specific drivers and new therapeutic targets, further promoting the advancement of personalized therapies. This review summarizes the latest findings on the transcriptomic and epigenetic profiling of human epidermal cells, analyzed from human biopsy or after in vitro cultivation, focusing on physiological, wound healing, and inflammatory skin conditions.
Collapse
Affiliation(s)
- Maria Pia Polito
- Centre for Regenerative Medicine ''Stefano Ferrari'', University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Grazia Marini
- Centre for Regenerative Medicine ''Stefano Ferrari'', University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Michele Palamenghi
- Centre for Regenerative Medicine ''Stefano Ferrari'', University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Elena Enzo
- Centre for Regenerative Medicine ''Stefano Ferrari'', University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
27
|
Shpakov AO. Allosteric Regulation of G-Protein-Coupled Receptors: From Diversity of Molecular Mechanisms to Multiple Allosteric Sites and Their Ligands. Int J Mol Sci 2023; 24:6187. [PMID: 37047169 PMCID: PMC10094638 DOI: 10.3390/ijms24076187] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Allosteric regulation is critical for the functioning of G protein-coupled receptors (GPCRs) and their signaling pathways. Endogenous allosteric regulators of GPCRs are simple ions, various biomolecules, and protein components of GPCR signaling (G proteins and β-arrestins). The stability and functional activity of GPCR complexes is also due to multicenter allosteric interactions between protomers. The complexity of allosteric effects caused by numerous regulators differing in structure, availability, and mechanisms of action predetermines the multiplicity and different topology of allosteric sites in GPCRs. These sites can be localized in extracellular loops; inside the transmembrane tunnel and in its upper and lower vestibules; in cytoplasmic loops; and on the outer, membrane-contacting surface of the transmembrane domain. They are involved in the regulation of basal and orthosteric agonist-stimulated receptor activity, biased agonism, GPCR-complex formation, and endocytosis. They are targets for a large number of synthetic allosteric regulators and modulators, including those constructed using molecular docking. The review is devoted to the principles and mechanisms of GPCRs allosteric regulation, the multiplicity of allosteric sites and their topology, and the endogenous and synthetic allosteric regulators, including autoantibodies and pepducins. The allosteric regulation of chemokine receptors, proteinase-activated receptors, thyroid-stimulating and luteinizing hormone receptors, and beta-adrenergic receptors are described in more detail.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
28
|
Fernández-Carrión R, Sorlí JV, Asensio EM, Pascual EC, Portolés O, Alvarez-Sala A, Francès F, Ramírez-Sabio JB, Pérez-Fidalgo A, Villamil LV, Tinahones FJ, Estruch R, Ordovas JM, Coltell O, Corella D. DNA-Methylation Signatures of Tobacco Smoking in a High Cardiovascular Risk Population: Modulation by the Mediterranean Diet. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3635. [PMID: 36834337 PMCID: PMC9964856 DOI: 10.3390/ijerph20043635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 06/18/2023]
Abstract
Biomarkers based on DNA methylation are relevant in the field of environmental health for precision health. Although tobacco smoking is one of the factors with a strong and consistent impact on DNA methylation, there are very few studies analyzing its methylation signature in southern European populations and none examining its modulation by the Mediterranean diet at the epigenome-wide level. We examined blood methylation smoking signatures on the EPIC 850 K array in this population (n = 414 high cardiovascular risk subjects). Epigenome-wide methylation studies (EWASs) were performed analyzing differential methylation CpG sites by smoking status (never, former, and current smokers) and the modulation by adherence to a Mediterranean diet score was explored. Gene-set enrichment analysis was performed for biological and functional interpretation. The predictive value of the top differentially methylated CpGs was analyzed using receiver operative curves. We characterized the DNA methylation signature of smoking in this Mediterranean population by identifying 46 differentially methylated CpGs at the EWAS level in the whole population. The strongest association was observed at the cg21566642 (p = 2.2 × 10-32) in the 2q37.1 region. We also detected other CpGs that have been consistently reported in prior research and discovered some novel differentially methylated CpG sites in subgroup analyses. In addition, we found distinct methylation profiles based on the adherence to the Mediterranean diet. Particularly, we obtained a significant interaction between smoking and diet modulating the cg5575921 methylation in the AHRR gene. In conclusion, we have characterized biomarkers of the methylation signature of tobacco smoking in this population, and suggest that the Mediterranean diet can increase methylation of certain hypomethylated sites.
Collapse
Affiliation(s)
- Rebeca Fernández-Carrión
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José V. Sorlí
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eva M. Asensio
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eva C. Pascual
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Olga Portolés
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Andrea Alvarez-Sala
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Francesc Francès
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Alejandro Pérez-Fidalgo
- Department of Medical Oncology, University Clinic Hospital of Valencia, 46010 Valencia, Spain
- Biomedical Research Networking Centre on Cancer (CIBERONC), Health Institute Carlos III, 28029 Madrid, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Laura V. Villamil
- Department of Physiology, School of Medicine, University Antonio Nariño, Bogotá 111511, Colombia
| | - Francisco J. Tinahones
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), University of Málaga, 29590 Málaga, Spain
| | - Ramon Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Jose M. Ordovas
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
- Nutritional Control of the Epigenome Group, Precision Nutrition and Obesity Program, IMDEA Food, UAM + CSIC, 28049 Madrid, Spain
| | - Oscar Coltell
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Computer Languages and Systems, Universitat Jaume I, 12071 Castellón, Spain
| | - Dolores Corella
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
29
|
Yakupu A, Zhang D, Guan H, Jiang M, Dong J, Niu Y, Tang J, Liu Y, Ma X, Lu S. Single-cell analysis reveals melanocytes may promote inflammation in chronic wounds through cathepsin G. Front Genet 2023; 14:1072995. [PMID: 36755572 PMCID: PMC9900029 DOI: 10.3389/fgene.2023.1072995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
During acute wound (AW) healing, a series of proper communications will occur between different epidermal cells at precise temporal stages to restore the integrity of the skin. However, it is still unclear what variation happened in epidermal cell interaction in the chronic wound environment. To provide new insights into chronic wound healing, we reconstructed the variations in the epidermal cell-cell communication network that occur in chronic wound healing via single-cell RNA-seq (scRNA-seq) data analysis. We found that the intricate cellular and molecular interactions increased in pressure ulcer (PU) compared to AW, especially the PARs signaling pathways were significantly upregulated. It shows that the PARs signaling pathways' main source was melanocytes and the CTSG-F2RL1 ligand-receptor pairs were its main contributor. Cathepsin G (CatG or CTSG) is a serine protease mainly with trypsin- and chymotrypsin-like specificity. It is synthesized and secreted by some immune or non-immune cells. Whereas, it has not been reported that melanocytes can synthesize and secrete the CTSG. F2R Like Trypsin Receptor 1 (F2RL1) is a member of proteinase-activated receptors (PARs) that are irreversibly activated by proteolytic cleavage and its stimulation can promote inflammation and inflammatory cell infiltration. In this study, we found that melanocytes increased in pressure ulcers, melanocytes can synthesize and secrete the CTSG and may promote inflammation in chronic wounds through CTSG-F2RL1 pairs, which may be a novel potential target and a therapeutic strategy in the treatment of chronic wounds.
Collapse
Affiliation(s)
- Aobuliaximu Yakupu
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Di Zhang
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haonan Guan
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minfei Jiang
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiaoyun Dong
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiwen Niu
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiajun Tang
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yingkai Liu
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xian Ma
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,*Correspondence: Xian Ma, ; Shuliang Lu,
| | - Shuliang Lu
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,*Correspondence: Xian Ma, ; Shuliang Lu,
| |
Collapse
|
30
|
Abstract
Proteases are an evolutionarily conserved family of enzymes that degrade peptide bonds and have been implicated in several common gastrointestinal (GI) diseases. Although luminal proteolytic activity is important for maintenance of homeostasis and health, the current review describes recent advances in our understanding of how overactivity of luminal proteases contributes to the pathophysiology of celiac disease, irritable bowel syndrome, inflammatory bowel disease and GI infections. Luminal proteases, many of which are produced by the microbiota, can modulate the immunogenicity of dietary antigens, reduce mucosal barrier function and activate pro-inflammatory and pro-nociceptive host signaling. Increased proteolytic activity has been ascribed to both increases in protease production and decreases in inhibitors of luminal proteases. With the identification of strains of bacteria that are important sources of proteases and their inhibitors, the stage is set to develop drug or microbial therapies to restore protease balance and alleviate disease.
Collapse
Affiliation(s)
- Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Mabel Guzman
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen’s University, Kingston, Ontario, Canada
| | - Josie Libertucci
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Alan E. Lomax
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen’s University, Kingston, Ontario, Canada,CONTACT Alan E. Lomax Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, K7L 2V7, Canada
| |
Collapse
|
31
|
Lv J, Liu J, Chao G, Zhang S. PARs in the inflammation-cancer transformation of CRC. Clin Transl Oncol 2022; 25:1242-1251. [PMID: 36547764 DOI: 10.1007/s12094-022-03052-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
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.
Collapse
Affiliation(s)
- Jianyu Lv
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China
| | - Jinguo Liu
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China
| | - Guanqun Chao
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University, Hanghou, China.
| | - Shuo Zhang
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|