1
|
Cazzola M, Page C, Rogliani P, Calzetta L, Matera MG. PI3K Inhibitors as Potential Therapeutic Agents for the Treatment of COPD with Associated Atherosclerosis. Drugs 2025; 85:741-753. [PMID: 40214902 PMCID: PMC12098434 DOI: 10.1007/s40265-025-02179-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 05/24/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) and cardiovascular disease (CVD) share a complex and multifactorial relationship characterized by overlapping risk factors, systemic inflammation, and intertwined pathophysiological mechanisms, with atherosclerosis emerging as a central inflammatory process connecting COPD and CVD, driven by systemic inflammation, oxidative stress, and endothelial dysfunction. While systemic inflammation is recognized as a critical link between these conditions, the precise pathways through which inflammation arises remain under investigation. There is therefore a need for therapeutic strategies to mitigate cardiovascular risks in patients with COPD. Among the pathways contributing to this interplay, the phosphoinositide 3-kinase (PI3K) signaling pathway has gained significant attention. Dysregulated PI3K signaling contributes to inflammation, oxidative stress, and endothelial dysfunction, which are key drivers of both COPD and CVD. Consequently, PI3K inhibitors have emerged as a promising therapeutic approach to mitigate inflammation and oxidative damage, offering a targeted strategy to address the shared pathological mechanisms underlying these diseases. A comprehensive understanding of the role of PI3K signaling and its inhibitors could facilitate the development of novel interventions to reduce cardiovascular risk in patients with COPD.
Collapse
Affiliation(s)
- Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Via Montpellier 1, 00131, Rome, Italy.
| | - Clive Page
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Via Montpellier 1, 00131, Rome, Italy
| | - Luigino Calzetta
- Unit of Respiratory Diseases and Lung Function, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| |
Collapse
|
2
|
Guo Z, Li P, Shi Z, Li X, Bao S, Zhong B, Zhang M, Wu Q. FKBP5 inhibitor suppresses platelet activation and thrombosis by inhibiting IKBKE/PI3K/Rap1 pathway. Int J Biol Macromol 2025; 311:144068. [PMID: 40345291 DOI: 10.1016/j.ijbiomac.2025.144068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/17/2025] [Accepted: 05/07/2025] [Indexed: 05/11/2025]
Abstract
Platelet activation and thrombosis play key roles in cardiovascular disease, and the function of FKBP5 in these processes is receiving increasing attention. The study evaluated the potential role of FKBP5 inhibitors in inhibiting platelet activation and thrombosis, and explored the regulation of the IKBKE/PI3K/Rap1 signaling pathway. Bioinformatics analysis, platelet aggregation assay, lactate dehydrogenase (LDH) and adenosine triphosphate (ATP) release assay, flow cytometry, Western blot technique, and mesenteric thrombosis model were used to evaluate the effect of FKBP5 inhibitors. Data were analyzed by statistical methods to ensure the reliability of the results. The results showed that FKBP5 inhibitors significantly inhibited platelet aggregation caused by multiple activators and reduced platelet production. Further analysis proved that FKBP5 inhibitors effectively blocked the activation of IKBKE/PI3K/Rap1 signaling pathway. In vivo experiments, FKBP5 inhibitors successfully inhibited mesenteric artery thrombosis, demonstrating its importance in preventing thrombosis, a finding that provides new ideas for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Ziwei Guo
- Graduate School of Dalian Medical University, Dalian 116044, Liaoning Province, China; Department of Cardiothoracic Surgery, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Peijin Li
- Department of Cardiothoracic Surgery, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China; Graduate School of Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Zehui Shi
- Graduate School of Dalian Medical University, Dalian 116044, Liaoning Province, China; Department of Cardiothoracic Surgery, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Xuejiao Li
- Graduate School of Dalian Medical University, Dalian 116044, Liaoning Province, China; Department of Cardiothoracic Surgery, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Sixu Bao
- Department of Cardiothoracic Surgery, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China; Graduate School of Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Bin Zhong
- Department of Cardiothoracic Surgery, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Ming Zhang
- Department of Cardiothoracic Surgery, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China
| | - Qiyong Wu
- Department of Cardiothoracic Surgery, Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, China.
| |
Collapse
|
3
|
Buitrago L, Menezes MR, Larson C, Li J, Kartika T, Banerjee P, Glickman F, Coller B. Unbiased high-throughput screening of drug-repurposing libraries identifies small-molecule inhibitors of clot retraction. Blood Adv 2025; 9:1049-1068. [PMID: 39374578 PMCID: PMC11909436 DOI: 10.1182/bloodadvances.2024013810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024] Open
Abstract
ABSTRACT Platelet clot retraction, the ultimate phase of platelet thrombus formation, is critical for clot stabilization. It requires functional αIIbβ3 receptors, fibrin, and the integrated actions of the actin-myosin contractile and cytoskeletal systems. Disturbances in clot retraction have been associated with both bleeding and thrombosis. We recently demonstrated that platelets treated with the αIIbβ3 antagonist peptide Arg-Gly-Asp-Trp, which eliminates fibrinogen-mediated platelet aggregation, are still able to retract clots. We have exploited this observation to develop an unbiased, functional high-throughput assay to identify small-molecule inhibitors of fibrin-mediated clot retraction adapted for a 384-well plate format. We tested 9710 compounds from drug-repurposing libraries (DRLs). These libraries contain compounds that are either US Food and Drug Administration approved or have undergone preclinical/clinical development. We identified 27 compounds from the Library of Pharmacologically Active Compounds library as inhibitors of clot retraction, of which 14 are known inhibitors of platelet function. From the DRLs, we identified 135 compounds (1.6% hit rate). After extensive curation, these compounds were categorized based on the activity of their reported target. Multiple kinase and phosphodiesterase inhibitors with known antiplatelet effects were identified, along with multiple deubiquitination and receptor inhibitors, as well as compounds that have not previously been reported to have antiplatelet activity. Studies of 1 of the deubiquitination inhibitors (degrasyn) suggest that its effects are downstream of thrombin-induced platelet-fibrinogen interactions and thus may permit the separation of platelet thrombin-induced aggregation-mediated events from clot retraction. Additional studies of the identified compounds may lead to novel mechanisms of inhibiting thrombosis.
Collapse
Affiliation(s)
- Lorena Buitrago
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY
| | - Miriam-Rose Menezes
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY
| | - Chloe Larson
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY
| | - Jihong Li
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY
| | - Thomas Kartika
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY
| | - Priyam Banerjee
- Bio-Imaging Resource Center, The Rockefeller University, New York, NY
| | - Fraser Glickman
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY
| | - Barry Coller
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, NY
| |
Collapse
|
4
|
Tang Q, Tang K, Markby GR, Parys M, Phadwal K, MacRae VE, Corcoran BM. Autophagy regulates cellular senescence by mediating the degradation of CDKN1A/p21 and CDKN2A/p16 through SQSTM1/p62-mediated selective autophagy in myxomatous mitral valve degeneration. Autophagy 2025:1-23. [PMID: 39988732 DOI: 10.1080/15548627.2025.2469315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
Myxomatous mitral valve degeneration (MMVD) is one of the most important age-dependent degenerative heart valve disorders in both humans and dogs. It is characterized by the aberrant remodeling of extracellular matrix (ECM), regulated by senescent myofibroblasts (aVICs) transitioning from quiescent valve interstitial cells (qVICs), primarily under TGFB1/TGF-β1 control. In the present study, we found senescent aVICs exhibited impaired macroautophagy/autophagy as evidenced by compromised autophagy flux and immature autophagosomes. MTOR-dependent autophagy induced by rapamycin and torin-1 attenuated cell senescence and decreased the expression of cyclin-dependent kinase inhibitors (CDKIs) CDKN2A/p16INK4A and CDKN1A/p21CIP1. Furthermore, induction of autophagy in aVICs by ATG (autophagy related) gene overexpression restored autophagy flux, with a concomitant reduction in CDKN1A and CDKN2A expression and senescence-associated secretory phenotype (SASP). Conversely, autophagy deficiency induced CDKN1A and CDKN2A accumulation and SASP, whereas ATG re-expression alleviated senescent phenotypic transformation. Notably, CDKN1A and CDKN2A localized to autophagosomes and lysosomes following MTOR antagonism or MG132 treatment. SQSTM1/p62 was identified as the autophagy receptor to selectively sequester CDKN1A and CDKN2A cargoes for autophagic degradation. Our findings are the first demonstration that CDKN1A and CDKN2A are degraded through SQSTM1-mediated selective autophagy, independent of the ubiquitin-proteasome pathway. These data will inform development of therapeutic strategies for the treatment of canine and human MMVD, and for the treatment of Alzheimer disease, Parkinson disease and other age-related degenerative disorders.Abbreviations: ACTA2/α-SMA: actin alpha 2, smooth muscle; AKT: AKT serine/threonine kinase; aVICs: activated valve interstitial cells; ATG: autophagy related; baf-A1: bafilomycin A1; BrdU, bromodeoxyuridine; BSA: bovine serum albumin; CDKIs, cyclin-dependent kinase inhibitors; CDKN1A/p21: cyclin dependent kinase inhibitor 1A; CDKN2A/p16: cyclin dependent kinase inhibitor 2A; co-IP: co-immunoprecipitation; DMSO: dimethylsulfoxide; ECM, extracellular matrix; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; eGFP: green fluorescent protein; ELISA: enzyme-linked immunosorbent assay; HEK-293T, human embryonic kidney 293T; HRP: horseradish peroxidase; KO: knockout; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; LIR: MAP1LC3/LC3-interacting region; MFS: Marfan syndrome; MKI67/Ki-67: marker of proliferation Ki-67; MMVD: myxomatous mitral valve degeneration; MTOR: mechanistic target of rapamycin kinase; MTORC: MTOR complex; OE: overexpression; PBST, phosphate-buffered saline with 0.1% Tween-20; PCNA: proliferating cell nuclear antigen; PIK3CA/PI3K: phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; PLA: proximity ligation assays; PSMA1: proteasome 20S subunit alpha 1; PSMB5: proteasome 20S subunit beta 5; qVICs: quiescent valve interstitial cells; qRT-PCR: quantitative real-time PCR; SA-GLB1/β-gal: SA-senescence-associated GLB1/β-galactosidase; ROS: reactive oxygen species; SASP: senescence-associated secretory phenotype; RPS6KB1/p70 S6K: ribosomal protein S6 kinase B1; SMAD: SMAD family member; SQSTM1/p62: sequestosome 1; STEM: scanning transmission electron microscopy; TGFB: transforming growth factor beta; TGFBR: transforming growth factor beta receptor; TP53/p53: tumor protein p53; UPS: ubiquitin-proteasome system; WT, wild-type.
Collapse
Affiliation(s)
- Qiyu Tang
- The Roslin Institute, The University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| | - Keyi Tang
- The Roslin Institute, The University of Edinburgh, Edinburgh, UK
| | - Greg R Markby
- The Roslin Institute, The University of Edinburgh, Edinburgh, UK
| | - Maciej Parys
- The Roslin Institute, The University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| | - Kanchan Phadwal
- The Roslin Institute, The University of Edinburgh, Edinburgh, UK
| | - Vicky E MacRae
- The Roslin Institute, The University of Edinburgh, Edinburgh, UK
- School of Life Sciences, Faculty of Science and Engineering, Anglia Ruskin University, Cambridge, UK
| | - Brendan M Corcoran
- The Roslin Institute, The University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Wen H, Yano N, Zhao T, Wei L, Zhao TC. The protective effect of irisin against hemorrhagic injury is mediated by PI3K and p38 pathways in hemorrhage/resuscitation. J Pharmacol Exp Ther 2025; 392:100027. [PMID: 39892988 DOI: 10.1124/jpet.124.002238] [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: 03/28/2024] [Revised: 09/03/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
The objective of this study was to investigate whether phosphoinositide 3-kinase (PI3K) and p38 mitogen-activated kinase contribute to the protection of irisin during hemorrhage/resuscitation. Experimental groups were divided based on the different treatments during resuscitation as follows: (1) hemorrhage: adult male CD-1 mice were subjected to hemorrhage at a mean arterial blood pressure of 35-45 mm Hg for 60 minutes, followed by resuscitation with shed blood and lactated Ringer's solution (n = 13); (2) hemorrhage + irisin: receiving irisin (5 μg/kg; n = 13); (3) hemorrhage + irisin + PI3K inhibitor: receiving both Ly294002 (1 mg/kg, i.v.) and irisin (n = 6); and (4) hemorrhage + irisin + p38 inhibitor: receiving SB202190 (1 mg/kg, i.v.) and irisin (n = 6). Compared with hemorrhage/resuscitation control, irisin improved cardiac function and the recovery of hemodynamics in association with the decreased systemic interleukin (IL)-1, IL-6, and tumor necrosis factor (TNF)-α, which were completely abrogated by PI3K or p38 inhibitions. Furthermore, the inhibition of PI3K or p38 abolished irisin-induced reduction of the inflammatory cell infiltration and terminal deoxynucleotidyl transferase-mediated digoxigenin-deoxyuridine nick-end labeling-positive apoptosis in the cardiac and skeletal muscles. Irisin reduced TNF-α and IL-6 expression in cardiac and skeletal muscles, which was abrogated by the inhibition of PI3K or p38. Irisin-treated hemorrhage increases the phosphorylation of PI3K and p38 in both cardiac and skeletal muscles, which was mitigated by the inhibition of PI3K or p38. PI3K and p38 play an important role in modulating the protective effect of irisin during the hemorrhage/resuscitation. SIGNIFICANCE STATEMENT: This study has identified a critical pathway in the regulation of trauma/hemorrhage by using a preclinical trauma model, in which irisin, as a hormone factor, stimulates PI3K and p38 pathways to induce protection against traumatic conditions. The study holds promise for developing a new therapeutic strategy to target irisin and its pathways related to PI3K and p38 to treat trauma and its comorbidities to reduce mortality for clinical implications.
Collapse
Affiliation(s)
- Huai Wen
- Department of Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Naohiro Yano
- Department of Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Thomas Zhao
- Department of Biology, Boston University, Boston, Massachusetts
| | - Lei Wei
- Department of Orthopedics, Rhode Island Hospital, Brown University, Providence, Rhode Island
| | - Ting C Zhao
- Department of Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island; Department of Plastic Surgery, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island.
| |
Collapse
|
6
|
You H, Gou Q, Dong M, Chang F, Xiu J. Exploring the role of iNOS in HFpEF-Related myocardial fibrosis: Involvement of PTEN-PI3K/AKT signaling pathway. Biochem Biophys Res Commun 2024; 734:150589. [PMID: 39245028 DOI: 10.1016/j.bbrc.2024.150589] [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: 07/04/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a challenging condition to treat with myocardial fibrosis being a pivotal pathological component. Previous studies have suggested a role for inducible nitric oxide synthase (iNOS) in the progression of this condition, but the precise mechanisms remain unclear. This study aimed to investigate the role of iNOS in HFpEF-related myocardial fibrosis and identify potential therapeutic targets. METHODS A 'two-hit' mouse model of HFpEF was established, and echocardiography, histopathology and biochemical analyses were performed. In vitro experiments were conducted in mouse cardiac fibroblasts, with iNOS overexpression and application of iNOS or phosphatidylinositol 3 kinase (PI3K) inhibitors. The iNOS-S-nitrosylated phosphatase and TENsin homolog (SNO-PTEN)-phosphorylated-protein kinase B (p-AKT) pathway was investigated, along with the effects on fibrotic markers and cell proliferation and migration. RESULTS HFpEF mice exhibited significant cardiac dysfunction and fibrosis, with increased expression of iNOS, SNO-PTEN, and p-AKT, indicative of the activation of the iNOS-SNO-PTEN-p-AKT pathway. iNOS overexpression in mouse cardiac fibroblasts led to increased SNO-PTEN, decreased PTEN, activated phosphorylated PI3K (p-PI3K) and p-AKT, and enhanced cell proliferation and migration, as well as increased collagen I and III expression. The use of an iNOS inhibitor (L-NIL) or a PI3K inhibitor (LY294002) partially reversed these changes. CONCLUSION Our findings suggest that the iNOS-SNO-PTEN-p-AKT pathway may play a crucial role in HFpEF-related myocardial fibrosis, with iNOS and PI3K inhibitors offering potential therapeutic benefits. These insights may pave the way for the development of effective drug therapies for HFpEF.
Collapse
Affiliation(s)
- Hongjun You
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China; Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Qiling Gou
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Mengya Dong
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Fengjun Chang
- Department of Cardiovascular Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Jiancheng Xiu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
7
|
Salah A, Bouzid F, Dhouib W, Benmarzoug R, Triki N, Rebai A, Kharrat N. Integrative Bioinformatics Approaches to Uncover Hub Genes and Pathways Involved in Cardiovascular Diseases. Cell Biochem Biophys 2024; 82:2107-2127. [PMID: 38809349 DOI: 10.1007/s12013-024-01319-4] [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] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
Cardiovascular diseases (CVD) represent a significant global health challenge resulting from a complex interplay of genetic, environmental, and lifestyle factors. However, the molecular pathways and genetic factors involved in the onset and progression of CVDs remain incompletely understood. Here, we performed an integrative bioinformatic analysis to highlight specific genes and signaling pathways implicated in the pathogenesis of 80 CVDs. Differentially expressed genes (DEGs) were identified through the integrated analysis of microarray and GWAS datasets. Then, hub genes were identified after gene ontology functional annotation analysis and protein-protein internet (PPI) analysis. In addition, pathways were identified through KEGG and gene ontology enrichment analyses. A total of 821 hub genes related to 80 CVDs were identified, including 135 common and frequent CVD-associated genes. TNF, IL6, VEGFA, and TGFB.1 genes were the central core genes expressed in 50% or more of CVDs, confirming that the inflammation is a key pathological feature of CVDs. Analysis of hub genes by KEGG enrichment revealed predominant enrichment in 201 KEGG pathways, of which the AGE-RAGE signaling pathway in diabetic complications was identified as the common key KEGG implicated in 62 CVDs. In addition, the outcomes showed an overrepresentation in pathways categorized under human diseases, particularly in the subcategories of infectious diseases and cancers, which may be common risk factors for CVDs. In conclusion, this powerful approach for in silico fine-mapping of genes and pathways allowed the identification of determinant hubs genes and pathways implicated in the pathogenesis of CVDs which could be employed in developing more targeted and effective interventions for preventing, diagnosing, and treating CVDs. The function of these hub genes in CVDs needs further exploration to elucidate their biological characteristics.
Collapse
Affiliation(s)
- Awatef Salah
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia.
| | - Fériel Bouzid
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Wala Dhouib
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Riadh Benmarzoug
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Nesrine Triki
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Ahmed Rebai
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Najla Kharrat
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
8
|
Liu C, Guo X, Zhang X. Modulation of atherosclerosis-related signaling pathways by Chinese herbal extracts: Recent evidence and perspectives. Phytother Res 2024; 38:2892-2930. [PMID: 38577989 DOI: 10.1002/ptr.8203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/06/2024]
Abstract
Atherosclerotic cardiovascular disease remains a preeminent cause of morbidity and mortality globally. The onset of atherosclerosis underpins the emergence of ischemic cardiovascular diseases, including coronary heart disease (CHD). Its pathogenesis entails multiple factors such as inflammation, oxidative stress, apoptosis, vascular endothelial damage, foam cell formation, and platelet activation. Furthermore, it triggers the activation of diverse signaling pathways including Phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt), NF-E2-related factor 2/antioxidant response element (Nrf2/ARE), the Notch signaling pathway, peroxisome proliferator-activated receptor (PPAR), nucleotide oligo-structural domain-like receptor thermoprotein structural domain-associated protein 3 (NLRP3), silencing information regulator 2-associated enzyme 1 (Sirt1), nuclear transcription factor-κB (NF-κB), Circular RNA (Circ RNA), MicroRNA (mi RNA), Transforming growth factor-β (TGF-β), and Janus kinase-signal transducer and activator of transcription (JAK/STAT). Over recent decades, therapeutic approaches for atherosclerosis have been dominated by the utilization of high-intensity statins to reduce lipid levels, despite significant adverse effects. Consequently, there is a growing interest in the development of safer and more efficacious drugs and therapeutic modalities. Traditional Chinese medicine (TCM) offers a vital strategy for the prevention and treatment of cardiovascular diseases. Numerous studies have detailed the mechanisms through which TCM active ingredients modulate signaling molecules and influence the atherosclerotic process. This article reviews the signaling pathways implicated in the pathogenesis of atherosclerosis and the advancements in research on TCM extracts for prevention and treatment, drawing on original articles from various databases including Google Scholar, Medline, CNKI, Scopus, and Pubmed. The objective is to furnish a reference for the clinical management of cardiovascular diseases.
Collapse
Affiliation(s)
- Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinyi Guo
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xulong Zhang
- Shaanxi Provincial Rehabilitation Hospital, Xi'an, China
| |
Collapse
|
9
|
Shi R, Gao S, Huang H, Jiang K, Wang D. Integrating network pharmacology with microRNA microarray analysis to identify the role of miRNAs in thrombosis treated by the Dahuang Zhechong pill. Comput Biol Med 2024; 173:108338. [PMID: 38531252 DOI: 10.1016/j.compbiomed.2024.108338] [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: 11/28/2023] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 03/28/2024]
Abstract
BACKGROUND Thrombotic diseases are the leading causes of death worldwide, urging for improvements in treatment strategies. Dahuang Zhechong pill (DHZCP) is a traditional Chinese medicine widely used for treating thrombotic diseases; however, the underlying mechanisms remain unclear. This study aimed to explore the potential mechanisms of DHZCP in treating thrombosis with a focus on bioinformatics and miRNAs. METHODS We used network pharmacology to explore the targets of thrombosis treated with DHZCP and performed microarray analysis to acquire miRNA profiles and predict the target genes in thrombin-stimulated MEG-01 cells treated with DHZCP. Based on the overlapping of targets, we carried out a component-target-miRNA network and enrichment analysis and validated the selected miRNAs and mRNAs using quantitative reverse transcription-polymerase chain reaction. RESULTS Our data showed 850 targets of 230 active ingredients of DHZCP and 1214 thrombosis-related genes; 235 targets were common. We identified 32 miRNAs that were regulated by thrombin stimulation but regulated reversely by DHZCP treatment in MEG-01 cells, and predicted 1846 targets with function annotation. We analyzed conjointly 23 integrating targets from network pharmacology and microarray. HIF1A, PIK3CA, MAPK1 and BCL2L1 emerged as key nodes in the network diagrams. We confirmed the differential expression of seven miRNAs, one mRNA (BCL2L1) and platelet surface protein. CONCLUSIONS This study showed that miRNAs and their targets, such as BCL2L1, played crucial roles in platelet activation during DHZCP intervention in thrombosis, highlighting their potential to alleviate platelet activation and increase cell apoptosis. The study's findings could help develop new strategies for improving thrombosis treatment.
Collapse
Affiliation(s)
- Rui Shi
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, 410008, China.
| | - Shan Gao
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, 410008, China
| | - Huichao Huang
- Department of Infectious Disease, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ke Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, 410008, China
| | - Dongsheng Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China; Hunan Key Laboratory of Traditional Chinese Medicine for Gan of State Administration, Central South University, Changsha, 410008, China.
| |
Collapse
|
10
|
Zhu M, Chen Y, Cheng L, Li X, Shen Y, Guo G, Xu X, Li H, Yang H, Liu C, He K. Calsyntenin-1 Promotes Doxorubicin-induced Dilated Cardiomyopathy in Rats. Cardiovasc Drugs Ther 2024; 38:237-252. [PMID: 36350487 PMCID: PMC10959838 DOI: 10.1007/s10557-022-07389-x] [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] [Accepted: 09/30/2022] [Indexed: 11/10/2022]
Abstract
PURPOSE Doxorubicin is an important cancer chemotherapeutic agent with severe cardiotoxic effects that eventually lead to dilated cardiomyopathy (DCM). Calsyntenin-1(CLSTN1) plays a critical role in the nervous system, but its relevance in cardiovascular diseases is unknown. We investigated the significance of CLSTN1 in doxorubicin-induced DCM. METHODS CLSTN1 expression in doxorubicin-induced DCM rats and H9c2 cells was determined using western blotting. To further explore the functions of CLSTN1, a cardiac-specific CLSTN1 overexpression rat model was constructed. The rats were subjected to analysis using echocardiographic, hemodynamic, and electrocardiographic parameters. Potential downstream molecules in CLSTN1 overexpression heart tissue were investigated using proteomics and western blotting. Finally, a knockdown of CLSTN1 was constructed to investigate the rescue function on doxorubicin-induced cell toxicity. RESULTS CLSTN1 protein expression increased drastically in doxorubicin-induced DCM rats and H9c2 cells. Under doxorubicin treatment, CLSTN1 protein-specific overexpression in the heart muscle promoted cardiac chamber enlargement and heart failure, while the knockdown of CLSTN1 reduced doxorubicin-induced cardiomyocyte toxicity in vitro. At the mechanistic level, overexpression of CLSTN1 downregulated SERCA2 expression and increased the phosphorylation levels of PI3K-Akt and CaMK2. CONCLUSION Our findings demonstrated that CLSTN1 promotes the pathogenesis of doxorubicin-induced DCM. CLSTN1 could be a therapeutic target to prevent the development of doxorubicin-induced DCM.
Collapse
Affiliation(s)
- Mingxiang Zhu
- Medical School of Chinese PLA, Beijing, 100853, China
- Medical Big Data Research Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yibing Chen
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Liting Cheng
- Medical School of Chinese PLA, Beijing, 100853, China
- Medical Big Data Research Center, Chinese PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xin Li
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanying Shen
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Ge Guo
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiang Xu
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Hanlu Li
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Hao Yang
- Department of Radiation Oncology, Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Huhhot, China
| | - Chunlei Liu
- Translational Medicine Research Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Kunlun He
- Medical Big Data Research Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
11
|
Gołaszewska A, Misztal T, Kazberuk A, Rusak T. Study on the Mechanism of the Adrenaline-Evoked Procoagulant Response in Human Platelets. Int J Mol Sci 2024; 25:2997. [PMID: 38474244 PMCID: PMC10932417 DOI: 10.3390/ijms25052997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/23/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Adrenaline has recently been found to trigger phosphatidylserine (PS) exposure on blood platelets, resulting in amplification of the coagulation process, but the mechanism is only fragmentarily established. Using a panel of platelet receptors' antagonists and modulators of signaling pathways, we evaluated the importance of these in adrenaline-evoked PS exposure by flow cytometry. Calcium and sodium ion influx into platelet cytosol, after adrenaline treatment, was examined by fluorimetric measurements. We found a strong reduction in PS exposure after blocking of sodium and calcium ion influx via Na+/H+ exchanger (NHE) and Na+/Ca2+ exchanger (NCX), respectively. ADP receptor antagonists produced a moderate inhibitory effect. Substantial limitation of PS exposure was observed in the presence of GPIIb/IIIa antagonist, phosphoinositide-3 kinase (PI3-K) inhibitors, or prostaglandin E1, a cyclic adenosine monophosphate (cAMP)-elevating agent. We demonstrated that adrenaline may develop a procoagulant response in human platelets with the substantial role of ion exchangers (NHE and NCX), secreted ADP, GPIIb/IIIa-dependent outside-in signaling, and PI3-K. Inhibition of the above mechanisms and increasing cytosolic cAMP seem to be the most efficient procedures to control adrenaline-evoked PS exposure in human platelets.
Collapse
Affiliation(s)
- Agata Gołaszewska
- Department of General and Experimental Pathology, Medical University of Bialystok, Mickiewicza 2C, 15-230 Bialystok, Poland
| | - Tomasz Misztal
- Department of Physical Chemistry, Medical University of Bialystok, Mickiewicza 2A, 15-369 Bialystok, Poland; (T.M.); (T.R.)
| | - Adam Kazberuk
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-959 Bialystok, Poland;
| | - Tomasz Rusak
- Department of Physical Chemistry, Medical University of Bialystok, Mickiewicza 2A, 15-369 Bialystok, Poland; (T.M.); (T.R.)
| |
Collapse
|
12
|
Sabbah DA, Hajjo R, Bardaweel SK, Zhong HA. Targeting the PI3K/AKT signaling pathway in anticancer research: a recent update on inhibitor design and clinical trials (2020-2023). Expert Opin Ther Pat 2024; 34:141-158. [PMID: 38557273 DOI: 10.1080/13543776.2024.2338100] [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: 09/18/2023] [Accepted: 03/27/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Recent years have witnessed great achievements in drug design and development targeting the phosphatidylinositol 3-kinase/protein kinase-B (PI3K/AKT) signaling pathway, a pathway central to cell growth and proliferation. The nearest neighbor protein-protein interaction networks for PI3K and AKT show the interplays between these target proteins which can be harnessed for drug discovery. In this review, we discuss the drug design and clinical development of inhibitors of PI3K/AKT in the past three years. We review in detail the structures, selectivity, efficacy, and combination therapy of 35 inhibitors targeting these proteins, classified based on the target proteins. Approaches to overcoming drug resistance and to minimizing toxicities are discussed. Future research directions for developing combinational therapy and PROTACs of PI3K and AKT inhibitors are also discussed. AREA COVERED This review covers clinical trial reports and patent literature on inhibitors of PI3K and AKT published between 2020 and 2023. EXPERT OPINION To address drug resistance and drug toxicity of inhibitors of PI3K and AKT, it is highly desirable to design and develop subtype-selective PI3K inhibitors or subtype-selective AKT1 inhibitors to minimize toxicity or to develop allosteric drugs that can form covalent bonds. The development of PROTACs of PI3Kα or AKT helps to reduce off-target toxicities.
Collapse
Affiliation(s)
- Dima A Sabbah
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Rima Hajjo
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- National Center for Epidemics and Communicable Disease Control (JCDC), Amman, Jordan
| | - Sanaa K Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman, Jordan
| | - Haizhen A Zhong
- DSC 309, Department of Chemistry, The University of Nebraska at Omaha, Omaha, NE, USA
| |
Collapse
|
13
|
Hsia C, Shu L, Lee A, Tran O, Yang C, Yen T, Huang W, Hsia C, Jayakumar T, Chiou K, Sheu J. Ginkgetin effectively mitigates collagen and AA-induced platelet activation via PLCγ2 but not cyclic nucleotide-dependent pathway in human. J Cell Mol Med 2024; 28:e18139. [PMID: 38334198 PMCID: PMC10853947 DOI: 10.1111/jcmm.18139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/22/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Platelets assume a pivotal role in the cardiovascular diseases (CVDs). Thus, targeting platelet activation is imperative for mitigating CVDs. Ginkgetin (GK), from Ginkgo biloba L, renowned for its anticancer and neuroprotective properties, remains unexplored concerning its impact on platelet activation, particularly in humans. In this investigation, we delved into the intricate mechanisms through which GK influences human platelets. At low concentrations (0.5-1 μM), GK exhibited robust inhibition of collagen and arachidonic acid (AA)-induced platelet aggregation. Intriguingly, thrombin and U46619 remained impervious to GK's influence. GK's modulatory effect extended to ATP release, P-selectin expression, intracellular calcium ([Ca2+ ]i) levels and thromboxane A2 formation. It significantly curtailed the activation of various signaling cascades, encompassing phospholipase Cγ2 (PLCγ2)/protein kinase C (PKC), phosphoinositide 3-kinase/Akt/glycogen synthase kinase-3β and mitogen-activated protein kinases. GK's antiplatelet effect was not reversed by SQ22536 (an adenylate cyclase inhibitor) or ODQ (a guanylate cyclase inhibitor), and GK had no effect on the phosphorylation of vasodilator-stimulated phosphoproteinSer157 or Ser239 . Moreover, neither cyclic AMP nor cyclic GMP levels were significantly increased after GK treatment. In mouse studies, GK notably extended occlusion time in mesenteric vessels, while sparing bleeding time. In conclusion, GK's profound impact on platelet activation, achieved through inhibiting PLCγ2-PKC cascade, culminates in the suppression of downstream signaling and, ultimately, the inhibition of platelet aggregation. These findings underscore the promising therapeutic potential of GK in the CVDs.
Collapse
Affiliation(s)
- Chih‐Wei Hsia
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Department of Medical ResearchTaipei Medical University HospitalTaipeiTaiwan
| | - Lan‐Hsin Shu
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Pharmacology, College of MedicineNational Taiwan UniversityTaipeiTaiwan
| | - Ai‐Wei Lee
- Department of Anatomy and Cell Biology, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Oanh‐Thi Tran
- International Master/Ph.D. Program in Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Chih‐Hao Yang
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Ting‐Lin Yen
- Department of Medical ResearchCathay General HospitalTaipeiTaiwan
| | - Wei‐Chieh Huang
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Chih‐Hsuan Hsia
- Translational Medicine CenterShin Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
| | | | - Kuan‐Rau Chiou
- Division of Cardiology, Department of Internal Medicine, Shuang Ho HospitalTaipei Medical UniversityNew Taipei CityTaiwan
| | - Joen‐Rong Sheu
- Graduate Institute of Medical Sciences, College of MedicineTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
14
|
Hassan HA, Mohamed Abdelhamid A, Samy W, Osama Mohammed H, Mortada Mahmoud S, Fawzy Abdel Mageed A, Abbas NAT. Ameliorative effects of androstenediol against acetic acid-induced colitis in male wistar rats via inhibiting TLR4-mediated PI3K/Akt and NF-κB pathways through estrogen receptor β activation. Int Immunopharmacol 2024; 127:111414. [PMID: 38141404 DOI: 10.1016/j.intimp.2023.111414] [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: 09/27/2023] [Revised: 12/13/2023] [Accepted: 12/16/2023] [Indexed: 12/25/2023]
Abstract
5-androstenediol (ADIOL) functions as a selective estrogen receptor β (ERβ) ligand with a protective effect against many diseases. So, we conducted a novel insight into its role in acetic acid (AA)-induced colitis and investigated its effect on TLR4-Mediated PI3K/Akt and NF-κB Pathways and the potential role of ERβ as contributing mechanisms. METHODS Rats were randomized into 5 Groups; Control, Colitis, Colitis + mesalazine (MLZ), Colitis + ADIOL, and Colitis + ADIOL + PHTPP (ER-β antagonist). The colitis was induced through a rectal enema of acetic acid (AA) on the 8th day. At the end of treatment, colons were collected for macroscopic assessment. Tissue levels of malondialdehyde (MDA), superoxide dismutase (SOD), nuclear factor kappa b (NF-κB), toll-like receptor (TLR4), and phosphorylated Protein kinase B (pAKT) were measured. Besides, Gene expression of interleukin-1beta (IL-1β), metalloproteases 9 (Mmp9), inositol 3 phosphate kinase (PI3K), Neutrophil gelatinase-associated lipocalin (NGAL), ERβ and NLRP6 were assessed. Histopathological and immunohistochemical studies were also investigated. RESULTS Compared to the untreated AA group, the disease activity index (DAI) and macroscopic assessment indicators significantly decreased with ADIOL injections. Indeed, ADIOL significantly decreased colonic tissue levels of MDA, TLR4, pAKT, and NF-κB immunostainig while increased SOD activity and β catenin immunostainig. ADIOL mitigated the high genetic expressions of IL1β, NGAL, MMP9, and PI3K while increased ERβ and NLRP6 gene expression. Also, the pathological changes detected in AA groups were markedly ameliorated with ADIOL. The specific ERβ antagonist, PHTPP, largely diminished these protective effects of ADIOL. CONCLUSION ADIOL could be beneficial against AA-induced colitis mostly through activating ERβ.
Collapse
Affiliation(s)
- Heba A Hassan
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt; Pharmacology Department, Faculty of Medicine, Mutah University, Mutah, Al-karak 61710, Jordan.
| | - Amira Mohamed Abdelhamid
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Walaa Samy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine -Zagazig University, Zagazig 45519, Egypt.
| | - Heba Osama Mohammed
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Samar Mortada Mahmoud
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Amal Fawzy Abdel Mageed
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine -Zagazig University, Zagazig 45519, Egypt.
| | - Noha A T Abbas
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt.
| |
Collapse
|
15
|
Setiabakti NM, Tarlac V, Larsson P, Hamilton JR. PI3KC2α inhibition is antithrombotic in blood from hypercholesterolemic mice. J Thromb Haemost 2024; 22:249-254. [PMID: 37827379 DOI: 10.1016/j.jtha.2023.09.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 09/22/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Current antiplatelet agents exhibit reduced antithrombotic efficacy in high-risk populations such as populations with hypercholesterolemia. The class II PI3-kinase, PI3KC2α, is a recently discovered target for novel antiplatelet therapy. PI3KC2α inhibition is antithrombotic in healthy mouse models, but whether this is preserved in hypercholesterolemia remains unknown. OBJECTIVES This study aimed to examine whether genetic deficiency or pharmacologic inhibition of PI3KC2α provides antithrombotic effects in blood from hypercholesterolemic mice. METHODS Hypercholesterolemic PI3KC2α-deficient mice were generated by breeding into an ApoE-/- background. Thrombosis was examined using an ex vivo whole blood thrombosis assay. The effect of pharmacologic inhibition of PI3KC2α was examined in whole blood from ApoE-/- mice treated with the PI3KC2α inhibitor MIPS-21335. RESULTS ApoE-/- mice exhibited the anticipated prothrombotic effect of hypercholesterolemia, with a 1.5-fold increase in thrombus volume in blood from ApoE-/- vs wild-type mice. This prothrombotic phenotype in blood from hypercholesterolemic mice was significantly reduced with PI3KC2α deficiency. Acute pharmacologic inhibition of PI3KC2α with MIPS-21335 similarly reduced thrombosis in blood from ApoE-/- mice. CONCLUSION These findings demonstrate that targeting PI3KC2α results in a potent antithrombotic effect in hypercholesterolemic mice and suggest that PI3KC2α is a promising target for antithrombotic therapy in patients with hypercholesterolemia at a high risk of thrombotic events.
Collapse
Affiliation(s)
- Natasha M Setiabakti
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Volga Tarlac
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Pia Larsson
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia
| | - Justin R Hamilton
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
16
|
Qi R, Hou J, Yang Y, Yang Z, Wu L, Qiao T, Wang X, Song D. Integrin beta1 mediates the effect of telocytes on mesenchymal stem cell proliferation and migration in the treatment of acute lung injury. J Cell Mol Med 2023; 27:3980-3994. [PMID: 37855260 PMCID: PMC10746951 DOI: 10.1111/jcmm.17976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 08/22/2023] [Accepted: 09/16/2023] [Indexed: 10/20/2023] Open
Abstract
Co-transplantation of mesenchymal stem cells (MSCs) with telocytes (TCs) was found to have therapeutic effects, although the mechanism of intercellular communication is still unknown. Our current studies aim at exploring the potential molecular mechanisms of TCs interaction and communication with MSCs with a focus on integrin beta1 (ITGB1) in TCs. We found that the co-culture of MSCs with ITGB1-deleted TCs (TCITGB1-ko ) changed the proliferation, differentiation and growth dynamics ability of MSC in responses to LPS or PI3K inhibitor. Changes of MSC proliferation and apoptosis were accompanied with the dysregulation of cytokine mRNA expression in MSCs co-cultured with TCITGB1-ko during the exposure of PI3Kα/δ/β inhibitor, of which IL-1β, IL-6 and TNF-α increased, while IFN-γ, IL-4 and IL-10 decreased. The responses of PI3K p85, PI3K p110 and pAKT of MSCs co-cultured with TCITGB1-ko to LPS or PI3K inhibitor were opposite to those with ITGB1-presented TCs. The intraperitoneal injection of TCITGB1-ko , TCvector or MSCs alone, as well as the combination of MSCs with TCITGB1-ko or TCvector exhibited therapeutic effects on LPS-induced acute lung injury. Thus, our data indicate that telocyte ITGB1 contributes to the interaction and intercellular communication between MSCs and TCs, responsible for influencing other cell phenomes and functions.
Collapse
Affiliation(s)
- Ruixue Qi
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan HospitalFudan University Shanghai Medical SchoolShanghaiChina
| | - Jiayun Hou
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Institute of Clinical ScienceFudan University Shanghai Medical SchoolShanghaiChina
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Department of Pulmonary MedicineShanghai Xuhui Central Hospital, Fudan UniversityShanghaiChina
| | - Ying Yang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan HospitalFudan University Shanghai Medical SchoolShanghaiChina
| | - Zhicheng Yang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan HospitalFudan University Shanghai Medical SchoolShanghaiChina
| | - Lihong Wu
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan HospitalFudan University Shanghai Medical SchoolShanghaiChina
| | - Tiankui Qiao
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan HospitalFudan University Shanghai Medical SchoolShanghaiChina
| | - Xiangdong Wang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan HospitalFudan University Shanghai Medical SchoolShanghaiChina
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Institute of Clinical ScienceFudan University Shanghai Medical SchoolShanghaiChina
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Department of Pulmonary MedicineShanghai Xuhui Central Hospital, Fudan UniversityShanghaiChina
| | - Dongli Song
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Institute of Clinical ScienceFudan University Shanghai Medical SchoolShanghaiChina
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghai Key Laboratory of Lung Inflammation and InjuryShanghaiChina
- Department of Pulmonary MedicineShanghai Xuhui Central Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
17
|
Wu T, Chen Y, Yang C, Lu M, Geng F, Guo J, Pi Y, Ling Y, Xu J, Cai T, Lu L, Zhou Y. Systematical Evaluation of the Structure-Cardiotoxicity Relationship of 7-Azaindazole-based PI3K Inhibitors Designed by Bioisosteric Approach. Cardiovasc Toxicol 2023; 23:364-376. [PMID: 37787964 DOI: 10.1007/s12012-023-09809-2] [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: 07/17/2023] [Accepted: 09/07/2023] [Indexed: 10/04/2023]
Abstract
A growing concern of cardiotoxicity induced by PI3K inhibitors has raised the requirements to evaluate the structure-cardiotoxicity relationship (SCR) in the development process of novel inhibitors. Based on three bioisosteric 7-azaindazole-based candidate inhibitors namely FD269, FD268 and FD274 that give same order of inhibitory concentration 50% (IC50) magnitude against PI3Ks, in this work, we proposed to systematically evaluate the SCR of 7-azaindazole-based PI3K inhibitors designed by bioisosteric approach. The 24-h lethal concentrations 50% (LC50) of FD269, FD268 and FD274 against zebrafish embryos were 0.35, 4.82 and above 50 μM (not detected), respectively. Determination of the heart rate, pericardial and yolk-sac areas and vascular malformation confirmed the remarkable reduction in the cardiotoxicity of from FD269 to FD268 and to FD274. The IC50s of all three compounds against the hERG channel were tested on the CHO cell line that constitutively expressing hERG channel, which were all higher than 20 μM. The transcriptomic analysis revealed that FD269 and FD268 induced the up-regulation of noxo1b, which encodes a subunit of an NADPH oxidase evoking the oxidative stress. Furthermore, immunohistochemistry tests confirmed the structure-dependent attenuation of the overproduction of ROS and cardiac apoptosis. Our results verified the feasibility of bioisosteric replacement to attenuate the cardiotoxicity of 7-azaindazole-based PI3K inhibitors, suggesting that the screening for PI3K inhibitors with both high potency and low cardiotoxicity from bioisosteres would be a beneficial trial.
Collapse
Affiliation(s)
- Tianze Wu
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Yi Chen
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Chengbin Yang
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Mingzhu Lu
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Fang Geng
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Jianhua Guo
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yan Pi
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yun Ling
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Jun Xu
- ABA Chemicals Co., Ltd, Taicang, 215400, Jiangsu, China
| | - Tong Cai
- ABA Chemicals Co., Ltd, Taicang, 215400, Jiangsu, China
| | - Lei Lu
- School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yaming Zhou
- Department of Chemistry, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
18
|
Morris BJ, Donlon TA. Genes That Extend Lifespan May Do So by Mitigating the Increased Risk of Death Posed by Having Hypertension. Am J Hypertens 2023; 36:631-640. [PMID: 37561089 PMCID: PMC10647014 DOI: 10.1093/ajh/hpad070] [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: 05/12/2023] [Revised: 07/22/2023] [Accepted: 08/09/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Genetic factors influence lifespan. In humans, there appears to be a particularly strong genetic effect in those aged ≥ 90 years. An important contribution is nutrient sensing genes which confer cell resilience. METHODS Our research has been investigating the genetic factors by longitudinal studies of American men of Japanese descent living on the island of Oahu in Hawaii. This cohort began as the Honolulu Heart Program in the mid-1960s and most subjects are now deceased. RESULTS We previously discovered various genes containing polymorphisms associated with longevity. In recent investigations of the mechanism involved we found that the longevity genotypes ameliorated the risk of mortality posed by having a cardiometabolic disease (CMD)-most prominently hypertension. For the gene FOXO3 the protective alleles mitigated the risk of hypertension, coronary heart disease (CHD) and diabetes. For the kinase MAP3K5 it was hypertension, CHD and diabetes, for the kinase receptor PIK3R1 hypertension, CHD and stroke, and for the growth hormone receptor gene (GHR) and vascular endothelial growth factor receptor 1 gene (FLT1), it was nullifying the higher mortality risk posed by hypertension. Subjects with a CMD who had a longevity genotype had similar survival as men without CMD. No variant protected against risk of death from cancer. We have postulated that the longevity-associated genotypes reduced mortality risk by effects on intracellular resilience mechanisms. In a proteomics study, 43 "stress" proteins and associated biological pathways were found to influence the association of FOXO3 genotype with reduced mortality. CONCLUSIONS Our landmark findings indicate how heritable genetic components affect longevity.
Collapse
Affiliation(s)
- Brian J Morris
- Department of Research, NIH Center of Biomedical Research Excellence on Aging, Kuakini Medical Center, Honolulu, Hawaii 96817, USA
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, USA
- School of Medical Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Timothy A Donlon
- Department of Research, NIH Center of Biomedical Research Excellence on Aging, Kuakini Medical Center, Honolulu, Hawaii 96817, USA
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96813, USA
| |
Collapse
|
19
|
Vishwakarma P, Siddiqui NF, Thakur S, Jadhav H. FDA approved fused-pyrimidines as potential PI3K inhibitors: a computational repurposing approach. J Biomol Struct Dyn 2023; 42:13497-13514. [PMID: 37909480 DOI: 10.1080/07391102.2023.2276315] [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: 06/26/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
Fused pyrimidine scaffold is present in several US FDA-approved drugs for various therapeutic indications. Drug repurposing (or drug repositioning) involves the analysis of existing clinically approved drugs for new therapeutic indications. Phosphoinositide-3-kinase (PI3K), via the regulatory PI3K pathway, is involved in cell growth, proliferation, differentiation, survival, and angiogenesis. It is also considered a target in anticancer drug development as it promotes the growth of cancerous cells and increases resistance to anticancer therapy. The present work employed computational techniques like molecular docking, MMGBSA analysis, and molecular dynamics simulations to explore the PI3K inhibition by FDA-approved drugs with fused pyrimidine scaffold. The work identifies Lapatinib as a pan-class I PI3K inhibitor and Dipyridamole as an γ isoform-specific PI3K inhibitor and is reported here.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pinky Vishwakarma
- Pharmaceutical Chemistry Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani, Rajasthan, India
| | - Noor Fatima Siddiqui
- Pharmaceutical Chemistry Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani, Rajasthan, India
| | - Shikha Thakur
- Pharmaceutical Chemistry Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani, Rajasthan, India
| | - Hemant Jadhav
- Pharmaceutical Chemistry Laboratory, Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Pilani, Rajasthan, India
| |
Collapse
|
20
|
Chahbaoui N, Khamouli S, Alaqarbeh M, Belaidi S, Sinha L, Chtita S, Bouachrine M. Identification of novel curcumin derivatives against pancreatic cancer: a comprehensive approach integrating 3D-QSAR pharmacophore modeling, virtual screening, and molecular dynamics simulations. J Biomol Struct Dyn 2023; 42:12021-12039. [PMID: 37811784 DOI: 10.1080/07391102.2023.2266502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/27/2023] [Indexed: 10/10/2023]
Abstract
Pancreatic cancer, known as the "silent killer," poses a daunting challenge in cancer therapy. The dysregulation of the PI3Kα signaling pathway in pancreatic cancer has attracted considerable interest as a promising target for therapeutic intervention. In this regard, the use of curcumin derivatives as inhibitors of PI3Kα has emerged, providing a novel and promising avenue for developing effective treatments for this devastating disease. Computational approaches were employed to explore this potential and investigate 58 curcumin derivatives with cytotoxic activity against the Panc-1 cell line. Our approach involved ligand-based pharmacophore modeling and atom-based 3D-QSAR analysis. The resulting QSAR model derived from the best-fitted pharmacophore hypothesis (AAHRR_1) demonstrated remarkable performance with high correlation coefficients (R2) of 0.990 for the training set and 0.977 for the test set. The cross-validation coefficient (Q2) of 0.971 also validated the model's predictive power. Tropsha's recommended criteria, including the Y-randomization test, were employed to ensure its reliability. Furthermore, an enrichment study was conducted to evaluate the model's performance in identifying active compounds. AAHRR_1 was used to screen a curated PubChem database of curcumin-related compounds. Two molecules (CID156189304 and CID154728220) exhibited promising pharmacokinetic properties and higher docking scores than Alpelisib, warranting further investigation. Extensive molecular dynamics simulations provided crucial insights into the conformational dynamics within the binding site, validating their stability and behavior. These findings contribute to our understanding of the potential therapeutic effectiveness of these compounds as PI3Kα inhibitors in pancreatic cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Narimene Chahbaoui
- Group of Computational and Pharmaceutical Chemistry, LMCE Laboratory, University of Biskra, Biskra, Algeria
| | - Saida Khamouli
- Group of Computational and Pharmaceutical Chemistry, LMCE Laboratory, University of Biskra, Biskra, Algeria
| | - Marwa Alaqarbeh
- Basic Science Department, Prince Al Hussein Bin Abdullah II Academy for Civil Protection, Al-Balqa Applied University, Al-Salt, Jordan
| | - Salah Belaidi
- Group of Computational and Pharmaceutical Chemistry, LMCE Laboratory, University of Biskra, Biskra, Algeria
| | - Leena Sinha
- Physics Department, University of Lucknow, Lucknow, India
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Casablanca, Morocco
| | - Mohammed Bouachrine
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, University Moulay Ismail, Meknes, Morocco
- Superior School of Technology - Khenifra (EST-Khenifra), University of Sultan Moulay Sliman, Khenifra, Morocco
| |
Collapse
|
21
|
Jia W, Luo S, Guo H, Kong D. Development of PI3Kα inhibitors for tumor therapy. J Biomol Struct Dyn 2023; 41:8587-8604. [PMID: 36221910 DOI: 10.1080/07391102.2022.2132293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/28/2022] [Indexed: 10/17/2022]
Abstract
The PI3K/AKT/mTOR signaling pathway is well known to be involved in cell growth, proliferation, metabolism and other cellular physiological processes. Abnormal activation of this pathway is closely related to tumorigenesis and metastasis. As the starting node of the pathway, PI3K is known to contain 4 isoforms, including PI3Kα, a heterodimer composed of the catalytic subunit p110α and the regulatory subunit p85. PIK3CA, which encodes p110α, is frequently mutated in cancer, especially breast cancer. Abnormal activation of PI3Kα promotes cancer cell proliferation, migration, invasion, and angiogenesis; therefore, PI3Kα has become a key target for the development of anticancer drugs. The hinge region and the region of the mutation site in the PI3Kα protein are important for designing PI3Kα-specific inhibitors. As the group shared by the most PI3Kα-specific inhibitors reported thus far, carboxamide can produce hydrogen bonds with Gln859 and Ser854. Gln859 is specific to the p110α protein in producing hydrogen bond interactions with PI3Kα-specific inhibitors and this is a key point for designing PI3Kα inhibitors. To date, alpelisib is the only PI3Kα inhibitor approved for the treatment of breast cancer. Several other PI3Kα inhibitors are under evaluation in clinical trials. In this review, we briefly describe PI3Kα and its role in tumorigenesis, summarize the clinical trial results of some PI3Kα inhibitors as well as the synthetic routes of alpelisib, and finally give our proposal for the development of novel PI3Kα inhibitors for tumor therapy. HighlightsWe summarize the progress of PI3Kα and PI3Kα inhibitors in cancer from the second half of the 20th century to the present.We describe the clinical trial results of PI3Kα inhibitors as well as the synthetic routes of the only approved PI3Kα inhibitor alpelisib.Crystal structure of alpelisib bound to the PI3Kα receptor binding domain.This review gives proposal for the development of novel PI3Kα inhibitors and will serve as a complementary summary to other reviews in the research field of PI3K inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Wenqing Jia
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Shuyu Luo
- School of Stomatology, Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Han Guo
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dexin Kong
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
22
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
23
|
Burley K, Fitzgibbon L, van Heel D, Vuckovic D, Mumford AD. PIK3R3 is a candidate regulator of platelet count in people of Bangladeshi ancestry. Res Pract Thromb Haemost 2023; 7:100175. [PMID: 37538507 PMCID: PMC10394561 DOI: 10.1016/j.rpth.2023.100175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 08/05/2023] Open
Abstract
Background Blood platelets are mediators of atherothrombotic disease and are regulated by complex sets of genes. Association studies in European ancestry populations have already detected informative platelet regulatory loci. Studies in other ancestries can potentially reveal new associations because of different allele frequencies, linkage structures, and variant effects. Objectives To reveal new regulatory genes for platelet count (PLT). Methods Genome-wide association studies (GWAS) were performed in 20,218 Bangladeshi and 9198 Pakistani individuals from the Genes & Health study. Loci significantly associated with PLT underwent fine-mapping to identify candidate genes. Results Of 1588 significantly associated variants (P < 5 × 10-8) at 20 loci in the Bangladeshi analysis, most replicated findings in prior transancestry GWAS and in the Pakistani analysis. However, the Bangladeshi locus defined by rs946528 (chr1:46019890) did not associate with PLT in the Pakistani analysis but was in the same linkage disequilibrium block (r2 ≥ 0.5) as PLT-associated variants in prior East Asian GWAS. The single independent association signal was refined to a 95% credible set of 343 variants spanning 8 coding genes. Functional annotation, mapping to megakaryocyte regulatory regions, and colocalization with blood expression quantitative trait loci identified the likely mediator of the PLT phenotype to be PIK3R3 encoding a regulator of phosphoinositol 3-kinase (PI3K). Conclusion Abnormal PI3K activity in the vessel wall is already implicated in the pathogenesis of atherothrombosis. Our identification of a new association between PIK3R3 and PLT provides further mechanistic insights into the contribution of the PI3K pathway to platelet biology.
Collapse
Affiliation(s)
- Kate Burley
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Lucy Fitzgibbon
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - David van Heel
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Dragana Vuckovic
- Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Andrew D. Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
24
|
Lobo V, Rocha A, Castro TG, Carvalho MA. Synthesis of Novel 2,9-Disubstituted-6-morpholino Purine Derivatives Assisted by Virtual Screening and Modelling of Class I PI3K Isoforms. Polymers (Basel) 2023; 15:polym15071703. [PMID: 37050317 PMCID: PMC10096987 DOI: 10.3390/polym15071703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
The phosphatidylinositol-3 kinase (PI3K) pathway is one of the most frequently activated pathogenic signalling cascades in a wide variety of cancers. In the last 15 years, there has been an increase in the search for selective inhibitors of the four class I isoforms of PI3K, as they demonstrate better specificity and reduced toxicity in comparison to existing inhibitors. A ligand-based and target-based rational drug design strategy was employed to build a virtual library of 105 new compounds. Through this strategy, the four isoforms were compared regarding their activity pocket availability, amino acid sequences, and prone interactions. Additionally, a known active scaffold was used as a molecular base to design new derivatives. The virtual screening of the resultant library toward the four isoforms points to the obtention of 19 selective inhibitors for the PI3Kα and PI3Kγ targets. Three selective ligands, one for α-isoform and two for γ-isoform, present a ∆ (∆Gbinding) equal or greater than 1.5 Kcal/mol and were identified as the most promising candidates. A principal component analysis was used to establish correlations between the affinity data and some of the physicochemical and structural properties of the ligands. The binding modes and interactions established by the selective ligands in the active centre of the α and γ isoforms of PI3K were also investigated. After modelling studies, a synthetic approach to generate selective ligands was developed and applied in synthesising a set of derivatives that were obtained in good to excellent yield.
Collapse
|
25
|
Brioschi M, D’Alessandra Y, Mapelli M, Mattavelli I, Salvioni E, Eligini S, Mallia A, Ricci V, Gianazza E, Ghilardi S, Agostoni P, Banfi C. Impact of Sacubitril/Valsartan on Circulating microRNA in Patients with Heart Failure. Biomedicines 2023; 11:1037. [PMID: 37189655 PMCID: PMC10136141 DOI: 10.3390/biomedicines11041037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Sacubitril/Valsartan, used for the treatment of heart failure (HF), is a combination of two drugs, an angiotensin receptor inhibitor, and a neprilysin inhibitor, which activates vasoactive peptides. Even though its beneficial effects on cardiac functions have been demonstrated, the mechanisms underpinning these effects remain poorly understood. To achieve more mechanistic insights, we analyzed the profiles of circulating miRNAs in plasma from patients with stable HF with reduced ejection function (HFrEF) and treated with Sacubitril/Valsartan for six months. miRNAs are short (22-24 nt) non-coding RNAs, which are not only emerging as sensitive and stable biomarkers for various diseases but also participate in the regulation of several biological processes. We found that in patients with high levels of miRNAs, specifically miR-29b-3p, miR-221-3p, and miR-503-5p, Sacubitril/Valsartan significantly reduced their levels at follow-up. We also found a significant negative correlation of miR-29b-3p, miR-221-3p, and miR-503-5p with VO2 at peak exercise, whose levels decrease with HF severity. Furthermore, from a functional point of view, miR-29b-3p, miR-221-3p, and miR-503-5p all target Phosphoinositide-3-Kinase Regulatory Subunit 1, which encodes regulatory subunit 1 of phosphoinositide-3-kinase. Our findings support that an additional mechanism through which Sacubitril/Valsartan exerts its functions is the modulation of miRNAs with potentially relevant roles in HFrEF pathophysiology.
Collapse
Affiliation(s)
- Maura Brioschi
- Centro Cardiologico Monzino, Functional Proteomics, Metabolomics, and Network Analysis, IRCCS, 20138 Milano, Italy
| | - Yuri D’Alessandra
- Centro Cardiologico Monzino, Functional Proteomics, Metabolomics, and Network Analysis, IRCCS, 20138 Milano, Italy
| | - Massimo Mapelli
- Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
- Cardiovascular Section, Department of Clinical Sciences and Community Health, University of Milan, 20122 Milano, Italy
| | | | | | - Sonia Eligini
- Centro Cardiologico Monzino, Functional Proteomics, Metabolomics, and Network Analysis, IRCCS, 20138 Milano, Italy
| | - Alice Mallia
- Centro Cardiologico Monzino, Functional Proteomics, Metabolomics, and Network Analysis, IRCCS, 20138 Milano, Italy
- Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Università di Pavia, 27100 Pavia, Italy
| | - Veronica Ricci
- Centro Cardiologico Monzino, Functional Proteomics, Metabolomics, and Network Analysis, IRCCS, 20138 Milano, Italy
| | - Erica Gianazza
- Centro Cardiologico Monzino, Functional Proteomics, Metabolomics, and Network Analysis, IRCCS, 20138 Milano, Italy
| | - Stefania Ghilardi
- Centro Cardiologico Monzino, Functional Proteomics, Metabolomics, and Network Analysis, IRCCS, 20138 Milano, Italy
| | - Piergiuseppe Agostoni
- Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy
- Cardiovascular Section, Department of Clinical Sciences and Community Health, University of Milan, 20122 Milano, Italy
| | - Cristina Banfi
- Centro Cardiologico Monzino, Functional Proteomics, Metabolomics, and Network Analysis, IRCCS, 20138 Milano, Italy
| |
Collapse
|
26
|
Cheng Q, Wang M, Jin R, Li G. Blocking of PI3-kinase beta protects against cerebral ischemia/reperfusion injury by reducing platelet activation and downstream microvascular thrombosis in rats. Sci Rep 2023; 13:2030. [PMID: 36739310 PMCID: PMC9899241 DOI: 10.1038/s41598-023-29235-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/01/2023] [Indexed: 02/06/2023] Open
Abstract
Phosphoinositide 3-kinase beta (PI3Kβ) plays an important role in platelet activation and thrombosis, but its role in stroke pathology remains unknown. In this study, we investigated whether inhibition of PI3Kβ protects against cerebral ischemia/reperfusion (I/R) injury by preventing circulating platelet activation and downstream microvascular thrombosis. We used a rat intraluminal filament model of transient middle cerebral artery occlusion (tMCAO) because the rapid restoration of cerebral blood flow to the ischemic area in both tMCAO and endovascular thrombectomy provides clinical relevance for this model. The results showed that TGX221, a selective PI3Kβ inhibitor, treatment immediately before the onset of reperfusion dose-dependently reduced infarct volume and improved neurological function. The protective effects were associated with blocking platelet activation and thrombotic response, thereby reducing downstream microvascular thrombosis, and maintaining reperfusion efficiency. These results suggest that PI3Kβ might be a promising target for treating downstream microvascular thrombosis induced by cerebral I/R injury and offer a novel adjunctive treatment to improve reperfusion therapy for acute ischemic stroke.
Collapse
Affiliation(s)
- Qiong Cheng
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226000, China
| | - Min Wang
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA
| | - Rong Jin
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA.
| | - Guohong Li
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, PA, 17033, USA.
| |
Collapse
|
27
|
The genetic basis of exercise and cardiorespiratory fitness – Relation to cardiovascular disease. CURRENT OPINION IN PHYSIOLOGY 2023. [DOI: 10.1016/j.cophys.2023.100649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
28
|
Gromadziński L, Paukszto Ł, Lepiarczyk E, Skowrońska A, Lipka A, Makowczenko KG, Łopieńska-Biernat E, Jastrzębski JP, Holak P, Smoliński M, Majewska M. Pulmonary artery embolism: comprehensive transcriptomic analysis in understanding the pathogenic mechanisms of the disease. BMC Genomics 2023; 24:10. [PMID: 36624378 PMCID: PMC9830730 DOI: 10.1186/s12864-023-09110-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Pulmonary embolism (PE) is a severe disease that usually originates from deep vein thrombosis (DVT) of the lower extremities. This study set out to investigate the changes in the transcriptome of the pulmonary artery (PA) in the course of the PE in the porcine model. METHODS The study was performed on 11 male pigs: a thrombus was formed in each right femoral vein in six animals, and then was released to induce PE, the remaining five animals served as a control group. In the experimental animals total RNA was isolated from the PA where the blood clot lodged, and in the control group, from the corresponding PA segments. High-throughput RNA sequencing was used to analyse the global changes in the transcriptome of PA with induced PE (PA-E). RESULTS Applied multistep bioinformatics revealed 473 differentially expressed genes (DEGs): 198 upregulated and 275 downregulated. Functional Gene Ontology annotated 347 DEGs into 27 biological processes, 324 to the 11 cellular components and 346 to the 2 molecular functions categories. In the signaling pathway analysis, KEGG 'protein processing in endoplasmic reticulum' was identified for the mRNAs modulated during PE. The same KEGG pathway was also exposed by 8 differentially alternative splicing genes. Within single nucleotide variants, the 61 allele-specific expression variants were localised in the vicinity of the genes that belong to the cellular components of the 'endoplasmic reticulum'. The discovered allele-specific genes were also classified as signatures of the cardiovascular system. CONCLUSIONS The findings of this research provide the first thorough investigation of the changes in the gene expression profile of PA affected by an embolus. Evidence from this study suggests that the disturbed homeostasis in the biosynthesis of proteins in the endoplasmic reticulum plays a major role in the pathogenesis of PE.
Collapse
Affiliation(s)
- Leszek Gromadziński
- grid.412607.60000 0001 2149 6795Department of Cardiology and Internal Medicine, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| | - Łukasz Paukszto
- grid.412607.60000 0001 2149 6795Department of Botany and Nature Protection, University of Warmia and Mazury in Olsztyn, Plac Łódzki 1, 10-727 Olsztyn, Poland
| | - Ewa Lepiarczyk
- grid.412607.60000 0001 2149 6795Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| | - Agnieszka Skowrońska
- grid.412607.60000 0001 2149 6795Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| | - Aleksandra Lipka
- grid.412607.60000 0001 2149 6795Department of Gynecology, and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Żołnierska Str 18, 10-561 Olsztyn, Poland
| | - Karol G. Makowczenko
- grid.412607.60000 0001 2149 6795Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| | - Elżbieta Łopieńska-Biernat
- grid.412607.60000 0001 2149 6795Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str 1A, 10-719 Olsztyn, Poland
| | - Jan P. Jastrzębski
- grid.412607.60000 0001 2149 6795Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str 1A, 10-719 Olsztyn-Kortowo, Poland
| | - Piotr Holak
- grid.412607.60000 0001 2149 6795Department of Surgery and Radiology With Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Str 14, 10-719 Olsztyn, Poland
| | - Michał Smoliński
- grid.460107.4Clinic of Cardiology and Internal Diseases, University Clinical Hospital in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| | - Marta Majewska
- grid.412607.60000 0001 2149 6795Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str 30, 10-082 Olsztyn, Poland
| |
Collapse
|
29
|
Zhou Y, Zhang D, Tan P, Xian B, Jiang H, Wu Q, Huang X, Zhang P, Xiao X, Pei J. Mechanism of platelet activation and potential therapeutic effects of natural drugs. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154463. [PMID: 36347177 DOI: 10.1016/j.phymed.2022.154463] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/20/2022] [Accepted: 09/18/2022] [Indexed: 05/09/2023]
Abstract
BACKGROUND Cardiovascular disease is one of the most concerning chronic diseases in the world. Many studies have shown that platelet overactivation is a very important factor in the occurrence and development of cardiovascular diseases. At present, the widely used antiplatelet drugs have some defects, such as drug resistance and adverse reactions. PURPOSE The purpose of this article is to summarize the main mechanisms and pathways of platelet activation, the main targets of antiplatelet aggregation, and the antiplatelet aggregation components of natural drugs and their mechanisms of action to provide new research ideas for the development and application of antiplatelet drugs. STUDY DESIGN AND METHODS In this review, we systematically searched the PubMed, Google Scholar, Web of Science, and CNKI databases and selected studies based on predefined eligibility criteria. We then assessed their quality and extracted data. RESULTS ADP, AA, THR, AF, collagen, SDF-1α, and Ca2+ can induce platelet aggregation and trigger thrombosis. Natural drugs have a good inhibitory effect on platelet activation. More than 50 kinds of natural drugs and over 120 kinds of chemical compounds, including flavonoids, alkaloids, saponins, terpenoids, coumarins, and organic acids, have significantly inhibited platelet activation activity. The MAPK pathway, cGMP-PKG pathway, cAMP-PKA pathway, PI3K-AKT pathway, PTK pathway, PLC pathway, and AA pathway are the main mechanisms and pathways of platelet activation. CONCLUSION Natural drugs and their active ingredients have shown good activity and application prospects in anti-platelet aggregation. We hope that this review provides new research ideas for the development and application of antiplatelet drugs.
Collapse
Affiliation(s)
- Yongfeng Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dingkun Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Peng Tan
- Sichuan Academy of Traditional Chinese Medicine, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Chengdu 610041, China
| | - Bin Xian
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huajuan Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qinghua Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xulong Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ping Zhang
- Medical Supplies Centre of PLA General Hospital, Beijing 100036, China.
| | - Xiaohe Xiao
- Department of Liver Disease, Fifth Medical Center of PLA General Hospital, Beijing 10039, China.
| | - Jin Pei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
30
|
Manke MC, Ahrends R, Borst O. Platelet lipid metabolism in vascular thrombo-inflammation. Pharmacol Ther 2022; 237:108258. [DOI: 10.1016/j.pharmthera.2022.108258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022]
|
31
|
van Bavel JJA, Pham C, Beekman HDM, Houtman MJC, Bossu A, Sparidans RW, van der Heyden MAG, Vos MA. PI3K/mTOR inhibitor omipalisib prolongs cardiac repolarization along with a mild proarrhythmic outcome in the AV block dog model. Front Cardiovasc Med 2022; 9:956538. [PMID: 35990966 PMCID: PMC9381882 DOI: 10.3389/fcvm.2022.956538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background The phosphoinositide 3-kinase (PI3K) signaling pathway is an interesting target in cancer treatment. The awareness of the proarrhythmic risk of PI3K inhibitors was raised because PI3K is also involved in regulating signaling toward cardiac ion channels. Canine cardiomyocytes treated with PI3K inhibitors show an increased action potential duration and reduced cardiac repolarizing currents. Now, the potential proarrhythmic effect of chronic treatment of PI3K/mTOR inhibitor GSK2126458 (omipalisib) was investigated in the atrioventricular (AV) block dog model. Methods Purpose-bred Mongrel dogs received complete AV block by ablation of the bundle of His and their hearts were paced in the right ventricular apex at VDD-mode (RVA-VDD). In this way, sinus rhythm was maintained for 15 ± 1 days and thereby bradycardia-induced cardiac remodeling was prevented. Dogs received 1 mg/kg omipalisib once (n = 3) or twice (n = 10) a day via oral administration for 7 days. Under standardized conditions (anesthesia, bradycardia at 60 beats/min, and a dofetilide challenge), potential proarrhythmic effects of omipalisib were investigated. Results Twice daily dosing of omipalisib increased accumulative plasma levels compared to once daily dosing accompanied with adverse events. Omipalisib prolonged the QT interval at baseline and more strongly after the dofetilide challenge (490 ± 37 to 607 ± 48 ms). The arrhythmic outcome after omipalisib resulted in single ectopic beats in 30% of dogs perpetuating in multiple ectopic beats and TdP arrhythmia in 20% of dogs. Isolated ventricular cardiomyocytes from omipalisib-treated dogs showed a diminished IKs current density. Conclusion Chronic treatment of PI3K/mTOR inhibitor omipalisib prolonged the QT interval in a preclinical model under standardized proarrhythmic conditions. Furthermore, this study showed that electrical remodeling induced by omipalisib had a mild proarrhythmic outcome.
Collapse
Affiliation(s)
- J. J. A. van Bavel
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - C. Pham
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - H. D. M. Beekman
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - M. J. C. Houtman
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - A. Bossu
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - R. W. Sparidans
- Division Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - M. A. G. van der Heyden
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
- *Correspondence: M. A. G. van der Heyden
| | - M. A. Vos
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
32
|
Hou Y, Zhang F, Min W, Yuan K, Kuang W, Wang X, Zhu Y, Sun C, Xia F, Wang Y, Zhang H, Wang L, Yang P. Discovery of Novel Phosphoinositide-3-Kinase α Inhibitors with High Selectivity, Excellent Bioavailability, and Long-Acting Efficacy for Gastric Cancer. J Med Chem 2022; 65:9873-9892. [PMID: 35834807 DOI: 10.1021/acs.jmedchem.2c00549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Phosphoinositide-3-kinase (PI3K) overexpressed in many tumors is a promising target for cancer therapy. However, due to toxicity from the ubiquitous expression of PI3K in many tissues, the development of PI3K inhibitors with high selectivity and low toxicity has become an urgent need for tumor treatment. Herein, based on the HipHop, we designed and synthesized a series of 6-(4,6-dimorpholino-1,3,5-triazin-2-yl)benzo[d]oxazol-2-amine derivatives as potent, selective, and long-acting PI3Kα inhibitors. Compound 27 was determined with potent PI3Kα inhibitory activity (IC50 = 4.4 nM), which exhibited excellent selectivity for homologous PI3K enzymes and a 370 kinome panel. Meanwhile, 27 featured favorable stability (T1/2 > 10 h) and high bioavailability (130%). Importantly, compound 27 exerted great antigastric cancer activity in vivo when combined with taxol. Collectively, these characteristics suggested 27 to be a promising PI3K agent for cancer treatment.
Collapse
Affiliation(s)
- Yi Hou
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fang Zhang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wenjian Min
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wenbin Kuang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yasheng Zhu
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fei Xia
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yanyin Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Haolin Zhang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Linping Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China.,Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
33
|
Zhang X, Li Z, Liu X, Qin X, Luo J, Zhang W, Liu B, Wei Y. ZPI prevents ox-LDL-mediated endothelial injury leading to inhibition of EndMT, inflammation, apoptosis, and oxidative stress through activating Pi3k/Akt signal pathway. Drug Dev Res 2022; 83:1212-1225. [PMID: 35656597 DOI: 10.1002/ddr.21952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/31/2022] [Accepted: 05/02/2022] [Indexed: 11/09/2022]
Abstract
Oxidized low-density lipoprotein (ox-LDL)-mediated endothelial dysfunction exerts an essential role in the development of atherosclerosis. Protein Z-dependent protease inhibitor (ZPI), a member of the serine protease inhibitor superfamily, could inhibit the function of activated coagulation factor X (FXa) via interaction with protein Z (PZ). Studies have pointed out that ZPI was statistically related to atherosclerotic diseases, which may have a robust cardiovascular protective effect. However, the underlying mechanism of ZPI on ox-LDL-mediated endothelial injury requires further elucidation. Human umbilical vein endothelial cells (HUVECs) were treated with ox-LDL (100 μg/ml) and ZPI (10 μg/ml). Cell viability was measured by the Cell Counting Kit-8 (CCK-8) assay. Cell apoptosis, oxidative stress, and endothelial-to-mesenchymal transition (EndMT) were analyzed by immunofluorescence (IF). Cell migration was measured using a wound-healing assay. Quantitative real-time polymerase chain reaction and western blot analysis were performed to determine messenger RNA and protein expression. Ox-LDL (100 μg/ml, 48 h) significantly reduced cell viability and migration, increased EndMT, inflammation, apoptosis, and oxidative stress. The related protein expression of phosphatidylinositol 3 kinase/protein kinase B (Pi3k/Akt) signal pathway in HUVECs was also simultaneously decreased. We also discovered that ZPI treatment could prevent ox-LDL-mediated endothelial injury through the improvement of cell viability and alleviation of apoptosis, oxidative stress, EndMT, and inflammation. Thus, the protective effect of ZPI on HUVECs may be mediated by activation of the Pi3k/Akt signal pathway. ZPI may exert an important protective role in HUVECs dysfunction triggered by ox-LDL via activation of the Pi3k/Akt signal pathway. Therefore, ZPI may possess potential therapeutic effects on atherosclerotic endothelial injury-related diseases.
Collapse
Affiliation(s)
- Xingxu Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiqiang Li
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiangdong Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoming Qin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiachen Luo
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenming Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Baoxin Liu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yidong Wei
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Wang H, Li C, Liu X, Ma M. Design, synthesis and activity study of a novel PI3K degradation by hijacking VHL E3 ubiquitin ligase. Bioorg Med Chem 2022; 61:116707. [PMID: 35344835 DOI: 10.1016/j.bmc.2022.116707] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 11/25/2022]
Abstract
PI3K kinase plays an important role in regulating key processes in cells, such as cell growth, metabolism, proliferation, and apoptosis. The overexpression of PI3K kinase exists in many cancers. The proteolytic target chimera (PROTAC) technology is a new technology that uses the ubiquitin-proteasome system to degrade a given target protein. It has been described that CRBN-based PROTAC targets the degradation of PI3K kinase. However, PROTAC based on VHL has not been reported yet. Here, we connected the previously obtained highly active PI3K inhibitor to the VHL ligand through different small molecules, and obtained a series of PROTAC molecules targeting PI3K kinase. Obtain the most active compound through screening. It provides evidence for the feasibility of PROTAC technology to recruit VHL E3 ligase in PI3K kinase.
Collapse
Affiliation(s)
- Haili Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Chuchu Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Xiaoqing Liu
- Shanghai University of Medicine & Health Sciences affiliated Zhoupu Hospital, Shanghai 201318, China.
| | - Mingliang Ma
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China; Key Laboratory of Brain Functional Genomics-Ministry of Education, School of Life Science, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
35
|
Structural Insights from Molecular Modeling of Isoindolin-1-One Derivatives as PI3Kγ Inhibitors against Gastric Carcinoma. Biomedicines 2022; 10:biomedicines10040813. [PMID: 35453562 PMCID: PMC9030798 DOI: 10.3390/biomedicines10040813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 01/15/2023] Open
Abstract
The upregulation of phosphoinositol-3-kinase γ (PI3Kγ) is deemed to be positively correlated with tumor-associated-macrophage (TAM)-mediated gastric carcinoma (GC). PI3Kγ suppresses tumor necrosis factor-alpha (TNF-α) and interleukin-12 (IL-12) through activation of the AKT/mTOR pathway, which promotes the immunosuppressant phenotype of TAM. Unlike α and β isoforms, δ and γ isoforms are primarily distributed in leucocytes and macrophages. Dual inhibitors against PI3Kδ and PI3Kγ have been proven to have merits in targeting solid tumors. Furthermore, it has been found that PI3Kδ is activated by cytokines, while PI3Kγ is activated by G-protein-coupled receptors (GPCRs). This facilitates determining the functional difference between these two isoforms. For this goal, selective inhibitors would be immensely helpful. In the current manuscript, we conducted various molecular modeling studies with a series of isoindolin-1-one derivatives as potent PI3Kγ inhibitors by combining molecular docking, molecular dynamics (MD), molecular mechanics, Poisson–Boltzmann/generalized Born surface area (MM-PB/GBSA) binding free energy calculation, and three-dimensional structure–activity relationship (3D-QSAR) study. To evaluate the selectivity of γ isoform over δ, the molecular modeling studies of idelalisib analogs reported as PI3Kδ inhibitors were also investigated. The contour polyhedrons were generated from the comparative molecular field analysis (CoMFA) and comparative molecular similarity index analysis (CoMSIA) around the ligand-bound active site for both isoforms, which could emphasize plausible explanations for the physicochemical factors that affect selective ligand recognition. The binding modalities of the two isoforms using CoMFA and MD models were compared, which suggested some key differences in the molecular interactions with the ligands and could be summarized as three subsites (one affinity subsite near the C-helix and DFG and two hydrophobic subsites). In the context of the structure–activity relationship (SAR), several new compounds were designed using a fragment-substitution strategy with the aim of selectively targeting PI3Kγ. The pIC50 values of the designed compounds were predicted by the 3D-QSAR models, followed by the MM-PB/GBSA binding energy estimation. The overall findings suggest that the designed compounds have the potential to be used as PI3Kγ inhibitors with a higher binding affinity and selectivity.
Collapse
|
36
|
Wang X, Li W, Zhang Y, Sun Q, Cao J, Tan N, Yang S, Lu L, Zhang Q, Wei P, Ma X, Wang W, Wang Y. Calycosin as a Novel PI3K Activator Reduces Inflammation and Fibrosis in Heart Failure Through AKT-IKK/STAT3 Axis. Front Pharmacol 2022; 13:828061. [PMID: 35264961 PMCID: PMC8899514 DOI: 10.3389/fphar.2022.828061] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/24/2022] [Indexed: 01/20/2023] Open
Abstract
Aim: Inflammation and fibrosis have been shown to be critical factors in heart failure (HF) progression. Calycosin (Cal) is the major active component of Astragalus mongholicus Bunge and has been reported to have therapeutic effects on the cardiac dysfunction after myocardial infarction. However, whether Cal could ameliorate myocardial infarction (MI)-induced inflammation and fibrosis and precise mechanisms remain uncertain. The aim of this study is to explore the role of Cal in HF and to clarify the underlying mechanisms. Methods: For in vivo experiments, rats underwent left anterior descending artery ligation for heart failure model, and the cardioprotective effects of Cal were measured by echocardiographic assessment and histological examination. RNA-seq approach was applied to explore potential differential genes and pathways. For further mechanistic study, proinflammatory-conditioned media (conditioned media)-induced H9C2 cell injury model and TGFβ-stimulated cardiac fibroblast model were applied to determine the regulatory mechanisms of Cal. Results: In the in vivo experiments, echocardiography results showed that Cal significantly improved heart function. GO and reactome enrichment revealed that inflammation and fibrosis pathways are involved in the Cal-treated group. KEGG enrichment indicated that the PI3K–AKT pathway is enriched in the Cal-treated group. Further experiments proved that Cal alleviated cardiomyocyte inflammatory responses evidenced by downregulating the expressions of phosphorylated IκB kinase α/β (p-IKKα/β), phosphorylated nuclear factor kapa B (p-NFκB), and tumor necrosis factor α (TNFα). Besides, Cal effectively attenuated cardiac fibrosis through the inhibitions of expressions and depositions of collagen I and collagen III. In the in vitro experiments, the phosphatidylinositol three kinase (PI3K) inhibitor LY294002 could abrogate the anti-inflammation and antifibrosis therapeutic effects of Cal, demonstrating that the cardioprotective effects of Cal were mediated through upregulations of PI3K and serine/threonine kinase (AKT). Conclusion: Cal inhibited inflammation and fibrosis via activation of the PI3K–AKT pathway in H9C2 cells, fibroblasts, and heart failure in postacute myocardial infarction rats.
Collapse
Affiliation(s)
- Xiaoping Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weili Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yawen Zhang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qianbin Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Cao
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - NanNan Tan
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuangjie Yang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Linghui Lu
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Zhang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Peng Wei
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Wei Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China.,Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, Beijing, China
| | - Yong Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China.,Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, Beijing, China.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
37
|
PAR4-Mediated PI3K/Akt and RhoA/ROCK Signaling Pathways Are Essential for Thrombin-Induced Morphological Changes in MEG-01 Cells. Int J Mol Sci 2022; 23:ijms23020776. [PMID: 35054966 PMCID: PMC8775998 DOI: 10.3390/ijms23020776] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 02/05/2023] Open
Abstract
Thrombin stimulates platelets via a dual receptor system of protease-activated receptors (PARs): PAR1 and PAR4. PAR1 activation induces a rapid and transient signal associated with the initiation of platelet aggregation, whereas PAR4 activation results in a prolonged signal, required for later phases, that regulates the stable formation of thrombus. In this study, we observed differential signaling pathways for thrombin-induced PAR1 and PAR4 activation in a human megakaryoblastic leukemia cell line, MEG-01. Interestingly, thrombin induced both calcium signaling and morphological changes in MEG-01 cells via the activation of PAR1 and PAR4, and these intracellular events were very similar to those observed in platelets shown in previous studies. We developed a novel image-based assay to quantitatively measure the morphological changes in living cells, and observed the underlying mechanism for PAR1- and PAR4-mediated morphological changes in MEG-01 cells. Selective inhibition of PAR1 and PAR4 by vorapaxar and BMS-986120, respectively, showed that thrombin-induced morphological changes were primarily mediated by PAR4 activation. Treatment of a set of kinase inhibitors and 2-aminoethoxydiphenyl borate (2-APB) revealed that thrombin-mediated morphological changes were primarily regulated by calcium-independent pathways and PAR4 activation-induced PI3K/Akt and RhoA/ROCK signaling pathways in MEG-01 cells. These results indicate the importance of PAR4-mediated signaling pathways in thrombin-induced morphological changes in MEG-01 cells and provide a useful in vitro cellular model for platelet research.
Collapse
|
38
|
Multi-scale mechanism of antiviral drug-alike phytoligands from Ayurveda in managing COVID-19 and associated metabolic comorbidities: insights from network pharmacology. Mol Divers 2022; 26:2575-2594. [PMID: 34993740 PMCID: PMC8736312 DOI: 10.1007/s11030-021-10352-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/14/2021] [Indexed: 02/07/2023]
Abstract
The novel coronavirus disease (COVID-19), which emerged in Wuhan, China, is continuously spreading worldwide, creating a huge burden on public health and economy. Ayurveda, the oldest healing schema of Traditional Indian Medicinal (TIM) system, is considered as a promising CAM therapy to combat various diseases/ disorders. To explore the regulatory mechanisms of 3038 Ayurvedic herbs (AHs) against SARS-CoV-2, in this study, multi-targeting and synergistic actions of constituent 34,472 phytochemicals (APCs) are investigated using a comprehensive approach comprising of network pharmacology and molecular docking. Immunomodulatory prospects of antiviral drug-alike potentially effective phytochemicals (PEPs) are presented as a special case study, highlighting the importance of 6 AHs in eliciting the antiviral immunity. By evaluating binding affinity of 292 PEPs against 24 SARS-CoV-2 proteins, we develop and analyze a high-confidence "bi-regulatory network" of 115 PEPs having ability to regulate protein targets in both virus and its host human system. Furthermore, mechanistic actions of PEPs against cardiovascular complications, diabetes mellitus and hypertension are also investigated to address the regulatory potential of AHs in dealing with COVID-19-associated metabolic comorbidities. The study further reports 12 PEPs as promising source of COVID-19 comorbidity regulators.
Collapse
|
39
|
Pan N, Li ZC, Li ZH, Chen SH, Jiang MH, Yang HY, Liu YS, Hu R, Zeng YW, Dai LH, Liu L, Wang GL. Antiplatelet and Antithrombotic Effects of Isaridin E Isolated from the Marine-Derived Fungus via Downregulating the PI3K/Akt Signaling Pathway. Mar Drugs 2021; 20:23. [PMID: 35049878 PMCID: PMC8780978 DOI: 10.3390/md20010023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023] Open
Abstract
Isaridin E, a cyclodepsipeptide isolated from the marine-derived fungus Amphichorda felina (syn. Beauveria felina) SYSU-MS7908, has been demonstrated to possess anti-inflammatory and insecticidal activities. Here, we first found that isaridin E concentration-dependently inhibited ADP-induced platelet aggregation, activation, and secretion in vitro, but did not affect collagen- or thrombin-induced platelet aggregation. Furthermore, isaridin E dose-dependently reduced thrombosis formation in an FeCl3-induced mouse carotid model without increasing the bleeding time. Mechanistically, isaridin E significantly decreased the ADP-mediated phosphorylation of PI3K and Akt. In conclusion, these results suggest that isaridin E exerts potent antithrombotic effects in vivo without increasing the risk of bleeding, which may be due to its important role in inhibiting ADP-induced platelet activation, secretion and aggregation via the PI3K/Akt pathways.
Collapse
Affiliation(s)
- Ni Pan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
- Institute of Pediatrics, Guangzhou Women and Children’s Medical Centre, Guangzhou Medical University, Guangzhou 510080, China
| | - Zi-Cheng Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Zhi-Hong Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Sen-Hua Chen
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510080, China; (S.-H.C.); (M.-H.J.)
| | - Ming-Hua Jiang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510080, China; (S.-H.C.); (M.-H.J.)
| | - Han-Yan Yang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Yao-Sheng Liu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Rui Hu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Yu-Wei Zeng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| | - Le-Hui Dai
- Department of Basic Medical Sciences, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China;
| | - Lan Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou 510080, China; (S.-H.C.); (M.-H.J.)
| | - Guan-Lei Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; (N.P.); (Z.-C.L.); (Z.-H.L.); (H.-Y.Y.); (Y.-S.L.); (R.H.); (Y.-W.Z.)
| |
Collapse
|
40
|
Screening Analysis of Platelet miRNA Profile Revealed miR-142-3p as a Potential Biomarker in Modeling the Risk of Acute Coronary Syndrome. Cells 2021; 10:cells10123526. [PMID: 34944034 PMCID: PMC8700136 DOI: 10.3390/cells10123526] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/04/2021] [Accepted: 12/11/2021] [Indexed: 12/18/2022] Open
Abstract
Transcriptome analysis constitutes one of the major methods of elucidation of the genetic basis underlying the pathogenesis of various diseases. The post-transcriptional regulation of gene expression is mainly provided by microRNAs. Their remarkable stability in biological fluids and their high sensitivity to disease alteration indicates their potential role as biomarkers. Given the high mortality and morbidity of cardiovascular diseases, novel predictive biomarkers are sorely needed. Our study focuses for the first time on assessing potential biomarkers of acute coronary syndrome (ACS) based on the microRNA profiles of platelets. The study showed the overexpression of eight platelet microRNAs in ACS (miR-142-3p; miR-107; miR-338-3p, miR-223-3p, miR-21-5p, miR-130b-3p, miR-301a-3p, miR-221-3p) associated with platelet reactivity and functionality. Our results show that the combined model based on miR-142-3p and aspartate transaminase reached 82% sensitivity and 88% specificity in the differentiation of the studied groups. Furthermore, the analyzed miRNAs were shown to cluster into two orthogonal groups, regulated by two different biological factors. Bioinformatic analysis demonstrated that one group of microRNAs may be associated with the physiological processes of platelets, whereas the other group may be linked to platelet-vascular environment interactions. This analysis paves the way towards a better understanding of the role of platelet microRNAs in ACS pathophysiology and better modeling of the risk of ACS.
Collapse
|
41
|
Chen W, Dai G, Qian Y, Wen L, He X, Liu H, Gao Y, Tang X, Dong B. PIK3CA mutation affects the proliferation of colorectal cancer cells through the PI3K-MEK/PDK1-GPT2 pathway. Oncol Rep 2021; 47:11. [PMID: 34751411 DOI: 10.3892/or.2021.8222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 07/07/2021] [Indexed: 11/06/2022] Open
Abstract
The phosphatidylinositol‑3‑kinase catalytic subunit α (PIK3CA) gene is mutated in numerous human cancers. This mutation promotes the proliferation of tumor cells; however, the underlying mechanism is still not clear. In the present study, it was revealed that the PIK3CA mutation in colorectal cancer (CRC) HCT116 (MUT) rendered the cells more dependent on glutamine by regulating the glutamic‑pyruvate transaminase 2 (GPT2). The dependence of glutamine increased the proliferation of cells in a normal environment and resistance to a suboptimal environment. Further study revealed that the mutated PIK3CA could regulate GPT2 expression not only through signal transduction molecule 3‑phosphoinositide‑dependent kinase (PDK1) but also through mitogen‑activated protein kinase (MEK) molecules. In HCT116 cells, MEK inhibitor treatment could reduce the expression of GPT2 signaling molecules, thereby inhibiting the proliferation of CRC cells. A new signal transduction pathway, the PI3K/MEK/GPT2 pathway was identified. Based on these findings, MEK and PDK1 inhibitors were combined to inhibit the aforementioned pathway. It was revealed that the combined application of MEK and PDK1 inhibitors could promisingly inhibit the proliferation of MUT compared with the application of PI3K inhibitors, PDK1 inhibitors, or MEK inhibitors alone. In vivo, MEK inhibitors alone and combined inhibitors had stronger tumor‑suppressing effects. There was no significant difference between the PDK1‑inhibitor group and normal group in vivo. Thus, these results indicated that mutated PI3K affected GPT2 mediated by the MEK/PDK1 dual pathway, and that the PI3K/MEK/GPT2 pathway was more important in vivo. Inhibiting MEK and PDK1 concurrently could effectively inhibit the proliferation of CRC cells. Targeting the MEK and PDK1 signaling pathway may provide a novel strategy for the treatment of PIK3CA‑mutated CRC.
Collapse
Affiliation(s)
- Wenli Chen
- Department of Biochemistry, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Guangli Dai
- Department of Obstetrics and Gynecology, Wuhu Traditional Chinese Medicine Hospital, Wuhu, Anhui 241003, P.R. China
| | - Yike Qian
- Department of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Lian Wen
- Department of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xueqing He
- Department of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Hui Liu
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Yunxing Gao
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xingli Tang
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Bohan Dong
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
42
|
Shi H, Ossip DJ, Mayo NL, Lopez DA, Block RC, Post WS, Bertoni AG, Ding J, Chen S, Yan C, Xie Z, Hoeschele I, Liu Y, Li D. Role of DNA methylation on the association between physical activity and cardiovascular diseases: results from the longitudinal multi-ethnic study of atherosclerosis (MESA) cohort. BMC Genomics 2021; 22:790. [PMID: 34732130 PMCID: PMC8567593 DOI: 10.1186/s12864-021-08108-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 10/14/2021] [Indexed: 12/03/2022] Open
Abstract
Background The complexity of physical activity (PA) and DNA methylation interaction in the development of cardiovascular disease (CVD) is rarely simultaneously investigated in one study. We examined the role of DNA methylation on the association between PA and CVD. Results The Multi-Ethnic Study of Atherosclerosis (MESA) cohort Exam 5 data with 1065 participants free of CVD were used for final analysis. The quartile categorical total PA variable was created by activity intensity (METs/week). During a median follow-up of 4.0 years, 69 participants developed CVD. Illumina HumanMethylation450 BeadChip was used to provide genome-wide DNA methylation profiles in purified human monocytes (CD14+). We identified 23 candidate DNA methylation loci to be associated with both PA and CVD. We used the structural equation modeling (SEM) approach to test the complex relationships among multiple variables and the roles of mediators. Three of the 23 identified loci (corresponding to genes VPS13D, PIK3CD and VPS45) remained as significant mediators in the final SEM model along with other covariates. Bridged by the three genes, the 2nd PA quartile (β = − 0.959; 95%CI: − 1.554 to − 0.449) and the 3rd PA quartile (β = − 0.944; 95%CI: − 1.628 to − 0.413) showed the greatest inverse associations with CVD development, while the 4th PA quartile had a relatively weaker inverse association (β = − 0.355; 95%CI: − 0.713 to − 0.124). Conclusions The current study is among the first to simultaneously examine the relationships among PA, DNA methylation, and CVD in a large cohort with long-term exposure. We identified three DNA methylation loci bridged the association between PA and CVD. The function of the identified genes warrants further investigation in the pathogenesis of CVD. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08108-w.
Collapse
Affiliation(s)
- Hangchuan Shi
- Department of Clinical and Translational Research, University of Rochester Medical Center, Rochester, NY, 14642-0708, USA.,Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Deborah J Ossip
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Nicole L Mayo
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Daniel A Lopez
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Robert C Block
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Alain G Bertoni
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jingzhong Ding
- Department of Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, 27157, USA
| | - Si Chen
- Aab Cardiovascular Research Institute, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA.,Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA.,Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Zidian Xie
- Department of Clinical and Translational Research, University of Rochester Medical Center, Rochester, NY, 14642-0708, USA
| | - Ina Hoeschele
- Department of Statistics, Fralin Life Sciences Institute at Virginia Tech, Blacksburg, VA, 24061, USA
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA.
| | - Dongmei Li
- Department of Clinical and Translational Research, University of Rochester Medical Center, Rochester, NY, 14642-0708, USA.
| |
Collapse
|
43
|
Moore SF, Agbani EO, Wersäll A, Poole AW, Williams CM, Zhao X, Li Y, Hutchinson JL, Hunter RW, Hers I. Opposing Roles of GSK3α and GSK3β Phosphorylation in Platelet Function and Thrombosis. Int J Mol Sci 2021; 22:10656. [PMID: 34638997 PMCID: PMC8508950 DOI: 10.3390/ijms221910656] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 11/25/2022] Open
Abstract
One of the mechanisms by which PI3 kinase can regulate platelet function is through phosphorylation of downstream substrates, including glycogen synthase kinase-3 (GSK3)α and GSK3β. Platelet activation results in the phosphorylation of an N-terminal serine residue in GSK3α (Ser21) and GSK3β (Ser9), which competitively inhibits substrate phosphorylation. However, the role of phosphorylation of these paralogs is still largely unknown. Here, we employed GSK3α/β phosphorylation-resistant mouse models to explore the role of this inhibitory phosphorylation in regulating platelet activation. Expression of phosphorylation-resistant GSK3α/β reduced thrombin-mediated platelet aggregation, integrin αIIbβ3 activation, and α-granule secretion, whereas platelet responses to the GPVI agonist collagen-related peptide (CRP-XL) were significantly enhanced. GSK3 single knock-in lines revealed that this divergence is due to differential roles of GSK3α and GSK3β phosphorylation in regulating platelet function. Expression of phosphorylation-resistant GSK3α resulted in enhanced GPVI-mediated platelet activation, whereas expression of phosphorylation-resistant GSK3β resulted in a reduction in PAR-mediated platelet activation and impaired in vitro thrombus formation under flow. Interestingly, the latter was normalised in double GSK3α/β KI mice, indicating that GSK3α KI can compensate for the impairment in thrombosis caused by GSK3β KI. In conclusion, our data indicate that GSK3α and GSK3β have differential roles in regulating platelet function.
Collapse
Affiliation(s)
- Samantha F. Moore
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
| | - Ejaife O. Agbani
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Andreas Wersäll
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
| | - Alastair W. Poole
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
| | - Chris M. Williams
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
| | - Xiaojuan Zhao
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
| | - Yong Li
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
| | - James L. Hutchinson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
| | - Roger W. Hunter
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
- NHS Blood and Transplant, North Bristol Park, Filton, Bristol BS34 7QH, UK
| | - Ingeborg Hers
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol BS8 1TD, UK; (S.F.M.); (E.O.A.); (A.W.); (A.W.P.); (C.M.W.); (X.Z.); (Y.L.); (J.L.H.); (R.W.H.)
| |
Collapse
|
44
|
Li H, Liu X, Sun N, Wang T, Zhu J, Yang S, Song X, Wang R, Wang X, Zhao Y, Zhang Y. Differentially Expressed Circular Non-coding RNAs in Atherosclerotic Aortic Vessels and Their Potential Functions in Endothelial Injury. Front Cardiovasc Med 2021; 8:657544. [PMID: 34307490 PMCID: PMC8294331 DOI: 10.3389/fcvm.2021.657544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/29/2021] [Indexed: 01/22/2023] Open
Abstract
Background: Circular non-coding RNA (circRNA) has a variety of biological functions. However, the expression profile and potential effects of circRNA on atherosclerosis (AS) and vascular endothelial injury have not been fully elucidated. This study aims to identify the differentially expressed circRNAs in atherosclerotic aortic vessels and predict their potential functions in endothelial injury. Method: ApoE-/- mice were fed with high-fat diet for 12 weeks to induce AS. Atherosclerotic plaques were evaluated by H&E and Masson staining and immunohistochemistry; differentially expressed circRNAs were detected by Arraystar Circular RNA Microarray and verified by RT-PCR; the potential target mircoRNAs of circRNAs were predicted by miRanda, Tarbase, Targetscan and their expression changes were verified by RT-PCR; the potential target genes of mircoRNAs were predicted by Targetscan and verified by Western blot; the signaling pathways that they might annotate or regulate and their potential functions in vascular endothelial injury were predicted by gene enrichment analysis. Results: Fifty two circRNAs were up-regulated more than twice and 47 circRNAs were down-regulated more than 1.5 times in AS aortic vessels. Mmmu_circRNA_36781 and 37699 were up-regulated both in AS aortic vessels and H2O2-treated mouse aortic endothelial cells (MAECs). The expression of miR-30d-3p and miR-140-3p, the target microRNA of circRNA_37699 and circRNA_36781, were downregulated both in AS vessels and H2O2-treated MAECs. On the contrary, MKK6 and TP53RK, the potential target gene of miR-140-3p and miR-30d-3p, were upregulated both in AS aortic roots and H2O2-treated MAECs. Besides, gene enrichment analysis showed that MAPK and PI3K-AKT signaling pathway were the most potential signaling pathways regulated by the differentially expressed circRNAs in atherosclerosis. Conclusions: Mmu_circRNA_36781 (circRNA ABCA1) and 37699 (circRNA KHDRBS1) were significantly up-regulated in AS aortic vessels and H2O2-treated MAECs. They have potential regulatory effects on atherosclerosis and vascular endothelial injury by targeting miR-30d-3p-TP53RK and miR-140-3p-MKK6 axis and their downstream signaling pathways.
Collapse
Affiliation(s)
- Houwei Li
- Department of Cardiology at the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Na Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tianshuo Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jia Zhu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Shuang Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xia Song
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ruishuai Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xinhui Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yixiu Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
45
|
Donlon TA, Chen R, Masaki KH, Willcox BJ, Morris BJ. Association with Longevity of Phosphatidylinositol 3-Kinase Regulatory Subunit 1 Gene Variants Stems from Protection against Mortality Risk in Men with Cardiovascular Disease. Gerontology 2021; 68:162-170. [PMID: 34077942 PMCID: PMC11910987 DOI: 10.1159/000515390] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/20/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Genetic variation in the phosphatidylinositol 3-kinase reregulatory subunit 1 gene (PIK3R1) is associated with longevity. OBJECTIVE The aim of the study was to determine whether cardiovascular disease (CVD) affects this association. METHODS We performed a longitudinal study of longevity-associated PIK3R1 single-nucleotide polymorphism rs7709243 genotype by CVD status in 3,584 elderly American men of Japanese ancestry. RESULTS At baseline (1991-1993), 2,254 subjects had CVD and 1,314 did not. The follow-up until Dec 31, 2019 found that overall, men with a CVD had higher mortality than men without a CVD (p = 1.7 × 10-5). However, survival curves of CVD subjects differed according to PIK3R1 genotype. Those with longevity-associated PIK3R1 TT/CC had survival curves similar to those of subjects without a CVD (p = 0.11 for TT/CC, and p = 0.054 for TC), whereas survival curves for CVD subjects with the CT genotype were significantly attenuated compared with survival curves of subjects without a CVD (p = 0.0000012 compared with TT/CC, and p = 0.0000028 compared with TC). Men without CVD showed no association of longevity-associated genotype with life span (p = 0.58). Compared to subjects without any CVD, hazard ratios for mortality risk were 1.26 (95% CI, 1.14-1.39; p = 0.0000043) for CT subject with CVD and 1.07 (95% CI 0.99-1.17; p = 0.097) for CC/TT subjects with CVD. There was no genotypic effect on life span for 1,007 subjects with diabetes and 486 with cancer. CONCLUSION Our study provides novel insights into the basis for PIK3R1 as a longevity gene. We suggest that the PIK3R1 longevity genotype attenuates mortality risk in at-risk individuals by protection against cellular stress caused by CVD.
Collapse
Affiliation(s)
- Timothy A. Donlon
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Cell and Molecular Biology and Department of Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Randi Chen
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
| | - Kamal H. Masaki
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Bradley J. Willcox
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| | - Brian J. Morris
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
- School of Medical Sciences and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
46
|
Fradera X, Deng Q, Achab A, Garcia Y, Kattar SD, McGowan MA, Methot JL, Wilson K, Zhou H, Shaffer L, Goldenblatt P, Tong V, Augustin MA, Altman MD, Lesburg CA, Shah S, Katz JD. Discovery of a new series of PI3K-δ inhibitors from Virtual Screening. Bioorg Med Chem Lett 2021; 42:128046. [PMID: 33865969 DOI: 10.1016/j.bmcl.2021.128046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 11/18/2022]
Abstract
PI3K-δ mediates key immune cell signaling pathways and is a target of interest for treatment of oncological and immunological disorders. Here we describe the discovery and optimization of a novel series of PI3K-δ selective inhibitors. We first identified hits containing an isoindolinone scaffold using a combined ligand- and receptor-based virtual screening workflow, and then improved potency and selectivity guided by structural data and modeling. Careful optimization of molecular properties led to compounds with improved permeability and pharmacokinetic profile, and high potency in a whole blood assay.
Collapse
Affiliation(s)
- Xavier Fradera
- Computational and Structural Chemistry, Merck & Co., Inc., Boston, MA, USA.
| | - Qiaolin Deng
- Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Yudith Garcia
- Discovery Chemistry, Merck & Co., Inc., Boston, MA, USA
| | | | | | - Joey L Methot
- Discovery Chemistry, Merck & Co., Inc., Boston, MA, USA
| | - Kevin Wilson
- Discovery Chemistry, Merck & Co., Inc., Boston, MA, USA
| | - Hua Zhou
- Discovery Chemistry, Merck & Co., Inc., Boston, MA, USA
| | - Lynsey Shaffer
- Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | | | | | | | - Michael D Altman
- Computational and Structural Chemistry, Merck & Co., Inc., Boston, MA, USA
| | - Charles A Lesburg
- Computational and Structural Chemistry, Merck & Co., Inc., Boston, MA, USA
| | - Sanjiv Shah
- Quantitative Biosciences, Merck & Co., Inc., Boston, MA, USA
| | - Jason D Katz
- Discovery Chemistry, Merck & Co., Inc., Boston, MA, USA
| |
Collapse
|
47
|
Barrachina MN, Izquierdo I, Hermida-Nogueira L, Morán LA, Pérez A, Arroyo AB, García-Barberá N, González-Conejero R, Troitiño S, Eble JA, Rivera J, Martínez C, Loza MI, Domínguez E, García Á. The PI3Kδ Inhibitor Idelalisib Diminishes Platelet Function and Shows Antithrombotic Potential. Int J Mol Sci 2021; 22:ijms22073304. [PMID: 33804911 PMCID: PMC8037016 DOI: 10.3390/ijms22073304] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Clinical management of ischemic events and prevention of vascular disease is based on antiplatelet drugs. Given the relevance of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) as a candidate target in thrombosis, the main goal of the present study was to identify novel antiplatelet agents within the existing inhibitors blocking PI3K isoforms. Methods: We performed a biological evaluation of the pharmacological activity of PI3K inhibitors in platelets. The effect of the inhibitors was evaluated in intracellular calcium release and platelet functional assays, the latter including aggregation, adhesion, and viability assays. The in vivo drug antithrombotic potential was assessed in mice undergoing chemically induced arterial occlusion, and the associated hemorrhagic risk evaluated by measuring the tail bleeding time. Results: We show that PI3K Class IA inhibitors potently block calcium mobilization in human platelets. The PI3K p110δ inhibitor Idelalisib inhibits platelet aggregation mediated by ITAM receptors GPVI and CLEC-2, preferentially by the former. Moreover, Idelalisib also inhibits platelet adhesion and aggregation under shear and adhesion to collagen. Interestingly, an antithrombotic effect was observed in mice treated with Idelalisib, with mild bleeding effects at high doses of the drug. Conclusion: Idelalisib may have antiplatelet effects with minor bleeding effects, which provides a rationale to evaluate its antithrombotic efficacy in humans.
Collapse
Affiliation(s)
- María N. Barrachina
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Irene Izquierdo
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Lidia Hermida-Nogueira
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Luis A. Morán
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Amparo Pérez
- Pharmacology Applied to Drug Discovery Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade Santiago de Compostela, 15705 Santiago de Compostela, Spain; (A.P.); (M.I.L.); (E.D.)
- Grupo Biofarma, Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Ana B. Arroyo
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - Nuria García-Barberá
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - Rocío González-Conejero
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - Sara Troitiño
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
| | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, 48149 Münster, Germany;
| | - José Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - Constantino Martínez
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Arrixaca, CIBERER-U765, 30003 Murcia, Spain; (A.B.A.); (N.G.-B.); (R.G.-C.); (J.R.); (C.M.)
| | - María I. Loza
- Pharmacology Applied to Drug Discovery Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade Santiago de Compostela, 15705 Santiago de Compostela, Spain; (A.P.); (M.I.L.); (E.D.)
- Grupo Biofarma, Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Eduardo Domínguez
- Pharmacology Applied to Drug Discovery Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas, Universidade Santiago de Compostela, 15705 Santiago de Compostela, Spain; (A.P.); (M.I.L.); (E.D.)
- Grupo Biofarma, Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain
| | - Ángel García
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases, Universidade Santiago de Compostela and Instituto de Investigación Sanitaria de Santiago, 15706 Santiago de Compostela, Spain; (M.N.B.); (I.I.); (L.H.-N.); (L.A.M.); (S.T.)
- Correspondence: ; Tel.: +34-881-815429
| |
Collapse
|
48
|
Cardiovascular toxicity of PI3Kα inhibitors. Clin Sci (Lond) 2021; 134:2595-2622. [PMID: 33063821 DOI: 10.1042/cs20200302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/27/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
The phosphoinositide 3-kinases (PI3Ks) are a family of intracellular lipid kinases that phosphorylate the 3'-hydroxyl group of inositol membrane lipids, resulting in the production of phosphatidylinositol 3,4,5-trisphosphate from phosphatidylinositol 4,5-bisphosphate. This results in downstream effects, including cell growth, proliferation, and migration. The heart expresses three PI3K class I enzyme isoforms (α, β, and γ), and these enzymes play a role in cardiac cellular survival, myocardial hypertrophy, myocardial contractility, excitation, and mechanotransduction. The PI3K pathway is associated with various disease processes but is particularly important to human cancers since many gain-of-function mutations in this pathway occur in various cancers. Despite the development, testing, and regulatory approval of PI3K inhibitors in recent years, there are still significant challenges when creating and utilizing these drugs, including concerns of adverse effects on the heart. There is a growing body of evidence from preclinical studies revealing that PI3Ks play a crucial cardioprotective role, and thus inhibition of this pathway could lead to cardiac dysfunction, electrical remodeling, vascular damage, and ultimately, cardiovascular disease. This review will focus on PI3Kα, including the mechanisms underlying the adverse cardiovascular effects resulting from PI3Kα inhibition and the potential clinical implications of treating patients with these drugs, such as increased arrhythmia burden, biventricular cardiac dysfunction, and impaired recovery from cardiotoxicity. Recommendations for future directions for preclinical and clinical work are made, highlighting the possible role of PI3Kα inhibition in the progression of cancer-related cachexia and female sex and pre-existing comorbidities as independent risk factors for cardiac abnormalities after cancer treatment.
Collapse
|
49
|
Phosphoinositide 3-kinases in platelets, thrombosis and therapeutics. Biochem J 2021; 477:4327-4342. [PMID: 33242335 DOI: 10.1042/bcj20190402] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/20/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022]
Abstract
Our knowledge on the expression, regulation and roles of the different phosphoinositide 3-kinases (PI3Ks) in platelet signaling and functions has greatly expanded these last twenty years. Much progress has been made in understanding the roles and regulations of class I PI3Ks which produce the lipid second messenger phosphatidylinositol 3,4,5 trisphosphate (PtdIns(3,4,5)P3). Selective pharmacological inhibitors and genetic approaches have allowed researchers to generate an impressive amount of data on the role of class I PI3Kα, β, δ and γ in platelet activation and in thrombosis. Furthermore, platelets do also express two class II PI3Ks (PI3KC2α and PI3KC2β), thought to generate PtdIns(3,4)P2 and PtdIns3P, and the sole class III PI3K (Vps34), known to synthesize PtdIns3P. Recent studies have started to reveal the importance of PI3KC2α and Vps34 in megakaryocytes and platelets, opening new perspective in our comprehension of platelet biology and thrombosis. In this review, we will summarize previous and recent advances on platelet PI3Ks isoforms. The implication of these kinases and their lipid products in fundamental platelet biological processes and thrombosis will be discussed. Finally, the relevance of developing potential antithrombotic strategies by targeting PI3Ks will be examined.
Collapse
|
50
|
Zhao Y, Qian Y, Sun Z, Shen X, Cai Y, Li L, Wang Z. Role of PI3K in the Progression and Regression of Atherosclerosis. Front Pharmacol 2021; 12:632378. [PMID: 33767629 PMCID: PMC7985550 DOI: 10.3389/fphar.2021.632378] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Phosphatidylinositol 3 kinase (PI3K) is a key molecule in the initiation of signal transduction pathways after the binding of extracellular signals to cell surface receptors. An intracellular kinase, PI3K activates multiple intracellular signaling pathways that affect cell growth, proliferation, migration, secretion, differentiation, transcription and translation. Dysregulation of PI3K activity, and as aberrant PI3K signaling, lead to a broad range of human diseases, such as cancer, immune disorders, diabetes, and cardiovascular diseases. A growing number of studies have shown that PI3K and its signaling pathways play key roles in the pathophysiological process of atherosclerosis. Furthermore, drugs targeting PI3K and its related signaling pathways are promising treatments for atherosclerosis. Therefore, we have reviewed how PI3K, an important regulatory factor, mediates the development of atherosclerosis and how targeting PI3K can be used to prevent and treat atherosclerosis.
Collapse
Affiliation(s)
- Yunyun Zhao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xinyi Shen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yaoyao Cai
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|