1
|
He W, Li ZQ, Gu HY, Pan QL, Lin FX. Targeted Therapy of Spinal Cord Injury: Inhibition of Apoptosis Is a Promising Therapeutic Strategy. Mol Neurobiol 2024; 61:4222-4239. [PMID: 38066400 DOI: 10.1007/s12035-023-03814-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/16/2023] [Indexed: 07/11/2024]
Abstract
Spinal cord injury (SCI) is a serious disabling central nervous system injury that can lead to motor, sensory, and autonomic dysfunction below the injury level. SCI can be divided into primary injury and secondary injury according to pathological process. Primary injury is mostly irreversible, while secondary injury is a dynamic regulatory process. Apoptosis is an important pathological event of secondary injury and has a significant effect on the recovery of nerve function after SCI. Nerve cell death can further aggravate the microenvironment of the injured site, leading to neurological dysfunction and thus affect the clinical outcome of patients. Therefore, apoptosis plays a crucial role in the pathological progression of secondary SCI, while inhibiting apoptosis may be a promising therapeutic strategy for SCI. This review will summarize and explore the factors that lead to cell death after SCI, the influence of cross talk between signaling pathways and pathways involved in apoptosis and discuss the influence of apoptosis on SCI, and the therapeutic significance of targeting apoptosis on SCI. This review helps us to understand the role of apoptosis in secondary SCI and provides a theoretical basis for the treatment of SCI based on apoptosis.
Collapse
Affiliation(s)
- Wei He
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
| | - Zhi-Qiang Li
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
| | - Hou-Yun Gu
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
| | - Qi-Lin Pan
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China
| | - Fei-Xiang Lin
- Department of Spine Surgery, Ganzhou People's Hospital, Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China.
- Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), Jiangxi Province, 16 Meiguan Avenue, Ganzhou, 341000, People's Republic of China.
| |
Collapse
|
2
|
Luo P, Ji Y, Liu X, Zhang W, Cheng R, Zhang S, Qian X, Huang C. Affected inflammation-related signaling pathways in snake envenomation: A recent insight. Toxicon 2023; 234:107288. [PMID: 37703930 DOI: 10.1016/j.toxicon.2023.107288] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/24/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023]
Abstract
Snake envenomation is well known to cause grievous pathological signs, including haemorrhagic discharge, necrosis, and respiratory distress. However, inflammatory reactions are also common envenoming manifestations that lead to successive damage, such as oedema, ulceration, lymphadenectasis, systemic inflammatory response syndrome (SIRS) and even multiple organ dysfunction syndrome (MODS). Interference with the inflammatory burst is hence important in the clinical treatment of snake envenomation. Here, we summarize the typical snake toxins (or venoms) that cause inflammatory reactions and the underlying signaling pathways. In brief, inflammatory reactions are usually triggered by snake venom phospholipase A2 (svPLA2), snake venom metalloprotease (SVMP), snake venom serine protease (SVSP) and C-type lectin/snaclec (CTL) as well as disintegrin (DIS) via multiple signaling pathways. They are nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing 3 (NLRP3), nuclear factor kappa-B (NF-κB), mitogen-activated protein kinase (MAPK), janus kinase/signal transducer and activator of transcription (JAK-STAT) and phosphoinositide 3-Kinase/protein kinase B (PI3K/PKB also called PI3K-AKT) signaling pathways. Activation of these pathways promotes the expression of pro-inflammatory molecules such as cytokines, especially interleukin-1β (IL-1β) which causes further inflammatory cascades and manifestations, such as swelling, fever, pain, and severe complications. Remarkably, almost half of introduced snake toxins (or venoms) have anti-inflammatory effects through blocking these pathways and suppressing the expression of pro-inflammatory molecules. Investigation of affected inflammation-related signaling pathways is meaningful to achieve better clinical treatment.
Collapse
Affiliation(s)
- Peiyi Luo
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Yuxin Ji
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Xiaohan Liu
- Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, PR China.
| | - Weiyun Zhang
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Ruoxi Cheng
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Shuxian Zhang
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Xiao Qian
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| | - Chunhong Huang
- College of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, 330000, PR China.
| |
Collapse
|
3
|
Xiao W, Xu Y, Baak JP, Dai J, Jing L, Zhu H, Gan Y, Zheng S. Network module analysis and molecular docking-based study on the mechanism of astragali radix against non-small cell lung cancer. BMC Complement Med Ther 2023; 23:345. [PMID: 37770919 PMCID: PMC10537544 DOI: 10.1186/s12906-023-04148-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/31/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Most lung cancer patients worldwide (stage IV non-small cell lung cancer, NSCLC) have a poor survival: 25%-30% patients die < 3 months. Yet, of those surviving > 3 months, 10%-15% patients survive (very) long. Astragali radix (AR) is an effective traditional Chinese medicine widely used for non-small cell lung cancer (NSCLC). However, the pharmacological mechanisms of AR on NSCLC remain to be elucidated. METHODS Ultra Performance Liquid Chromatography system coupled with Q-Orbitrap HRMS (UPLC-Q-Orbitrap HRMS) was performed for the qualitative analysis of AR components. Then, network module analysis and molecular docking-based approach was conducted to explore underlying mechanisms of AR on NSCLC. The target genes of AR were obtained from four databases including TCMSP (Traditional Chinese Medicine Systems Pharmacology) database, ETCM (The Encyclopedia of TCM) database, HERB (A high-throughput experiment- and reference-guided database of TCM) database and BATMAN-TCM (a Bioinformatics Analysis Tool for Molecular mechanism of TCM) database. NSCLC related genes were screened by GEO (Gene Expression Omnibus) database. The STRING database was used for protein interaction network construction (PIN) of AR-NSCLC shared target genes. The critical PIN were further constructed based on the topological properties of network nodes. Afterwards the hub genes and network modules were analyzed, and enrichment analysis were employed by the R package clusterProfiler. The Autodock Vina was utilized for molecular docking, and the Gromacs was utilized for molecular dynamics simulations Furthermore, the survival analysis was performed based on TCGA (The Cancer Genome Atlas) database. RESULTS Seventy-seven AR components absorbed in blood were obtained. The critical network was constructed with 1447 nodes and 28,890 edges. Based on topological analysis, 6 hub target genes and 7 functional modules were gained. were obtained including TP53, SRC, UBC, CTNNB1, EP300, and RELA. After module analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that AR may exert therapeutic effects on NSCLC by regulating JAK-STAT signaling pathway, PI3K-AKT signaling pathway, ErbB signaling pathway, as well as NFkB signaling pathway. After the intersection calculation of the hub targets and the proteins participated in the above pathways, TP53, SRC, EP300, and RELA were obtained. These proteins had good docking affinity with astragaloside IV. Furthermore, RELA was associated with poor prognosis of NSCLC patients. CONCLUSIONS This study could provide chemical component information references for further researches. The potential pharmacological mechanisms of AR on NSCLC were elucidated, promoting the clinical application of AR in treating NSCLC. RELA was selected as a promising candidate biomarker affecting the prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Wenke Xiao
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yaxin Xu
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jan P Baak
- Stavanger University Hospital, Stavanger, 4068, Norway
- Dr. Med Jan Baak AS, Tananger, 4056, Norway
| | - Jinrong Dai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lijia Jing
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hongxia Zhu
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yanxiong Gan
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Shichao Zheng
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
4
|
An HJ, Gwon MG, Gu H, Bae S, Leem J, Lee JB, Park KK. STAT3/NF‑κB decoy oligodeoxynucleotides inhibit atherosclerosis through regulation of the STAT/NF‑κB signaling pathway in a mouse model of atherosclerosis. Int J Mol Med 2023; 51:37. [PMID: 37026512 PMCID: PMC10094942 DOI: 10.3892/ijmm.2023.5240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/11/2023] [Indexed: 04/03/2023] Open
Abstract
Atherosclerosis is a progressive chronic inflammatory condition that is the cause of most cardiovascular and cerebrovascular diseases. The transcription factor nuclear factor‑κB (NF‑κB) regulates a number of genes involved in the inflammatory responses of cells that are critical to atherogenesis, and signal transducer and activator of transcription (STAT)3 is a key transcription factor in immunity and inflammation. Decoy oligodeoxynucleotides (ODNs) bind to sequence‑specific transcription factors and limit gene expression by interfering with transcription in vitro and in vivo. The present study aimed to investigate the beneficial functions of STAT3/NF‑κB decoy ODNs in liposaccharide (LPS)‑induced atherosclerosis in mice. Atherosclerotic injuries of mice were induced via intraperitoneal injection of LPS and the mice were fed an atherogenic diet. Ring‑type STAT3/NF‑κB decoy ODNs were designed and administered via an injection into the tail vein of the mice. To investigate the effect of STAT3/NF‑κB decoy ODNs, electrophoretic mobility shift assay, western blot analysis, histological analysis with hematoxylin and eosin staining, Verhoeff‑Van Gieson and Masson's trichrome staining were performed. The results revealed that STAT3/NF‑κB decoy ODNs were able to suppress the development of atherosclerosis by attenuating morphological changes and inflammation in atherosclerotic mice aortae, and by reducing pro‑inflammatory cytokine secretion through inhibition of the STAT3/NF‑κB pathway. In conclusion, the present study provided novel insights into the antiatherogenic molecular mechanism of STAT3/NF‑κB decoy ODNs, which may serve as an additional therapeutic intervention to combat atherosclerosis.
Collapse
Affiliation(s)
- Hyun-Jin An
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Mi-Gyeong Gwon
- Department of Immunology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Hyemin Gu
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Seongjae Bae
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Jin Bae Lee
- Department of Cardiology, Daegu Catholic University Medical Center, Daegu 42472, Republic of Korea
| | - Kwan-Kyu Park
- Department of Pathology, College of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| |
Collapse
|
5
|
Ceja-Gálvez HR, Renteria-Flores FI, Nicoletti F, Hernández-Bello J, Macedo-Ojeda G, Muñoz-Valle JF. Severe COVID-19: Drugs and Clinical Trials. J Clin Med 2023; 12:2893. [PMID: 37109231 PMCID: PMC10142549 DOI: 10.3390/jcm12082893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/08/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
By January of 2023, the COVID-19 pandemic had led to a reported total of 6,700,883 deaths and 662,631,114 cases worldwide. To date, there have been no effective therapies or standardized treatment schemes for this disease; therefore, the search for effective prophylactic and therapeutic strategies is a primary goal that must be addressed. This review aims to provide an analysis of the most efficient and promising therapies and drugs for the prevention and treatment of severe COVID-19, comparing their degree of success, scope, and limitations, with the aim of providing support to health professionals in choosing the best pharmacological approach. An investigation of the most promising and effective treatments against COVID-19 that are currently available was carried out by employing search terms including "Convalescent plasma therapy in COVID-19" or "Viral polymerase inhibitors" and "COVID-19" in the Clinicaltrials.gov and PubMed databases. From the current perspective and with the information available from the various clinical trials assessing the efficacy of different therapeutic options, we conclude that it is necessary to standardize certain variables-such as the viral clearance time, biomarkers associated with severity, hospital stay, requirement of invasive mechanical ventilation, and mortality rate-in order to facilitate verification of the efficacy of such treatments and to better assess the repeatability of the most effective and promising results.
Collapse
Affiliation(s)
- Hazael Ramiro Ceja-Gálvez
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Francisco Israel Renteria-Flores
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Jorge Hernández-Bello
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Gabriela Macedo-Ojeda
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - José Francisco Muñoz-Valle
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
6
|
He S, Wang T, Shi C, Wang Z, Fu X. Network pharmacology-based approach to understand the effect and mechanism of Danshen against anemia. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114615. [PMID: 34509606 DOI: 10.1016/j.jep.2021.114615] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/26/2021] [Accepted: 09/05/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danshen, the dried rhizome of Salvia miltiorrhiza Bge., is widely used to treat cardio-cerebrovascular diseases in China. However, its role in nourishing blood, which has been detailed in historical literature for thousands of years, is perpetually disputed in the academic field. Moreover, there is no systematic research on Danshen in treating anemia. This research aimed to investigate the effects and mechanisms of Danshen on anemia in a zebrafish model based on the results of a network pharmacology study. MATERIALS AND METHODS The network pharmacology study was based on the screening of chemical components and related targets from TCMSP and SwissADME database. The genes associated with anemia were obtained from DisgeNet database, and the genes with the intersection of Danshen target genes were screened out. The Cytoscape 3.7.2 software package was used to construct the "ingredient-target-pathway" network. The exploration of target interaction by String system and the enrichment analysis by Metascape system, was used to discover the possible anti-anemia action mechanism of Danshen. Then, a zebrafish anemia model was induced by vinorelbine followed by the administration of aqueous/ethanol extract of Danshen in contrast to SiWu Decoction (SWD), which is generally acknowledged as a positive drug for tonifying blood. Afterward, the red blood cell signal, cardiac output, and blood flow velocity were detected to evaluate their blood-enriching effects. Quantitative real-time polymerase chain reaction (qPCR) was used to analyze the mRNA levels of hematopoietic-related factors, which were predicted in network pharmacology. RESULTS Compounds and target screening hinted that 115 chemical targets from Danshen were related to anemia, KEGG pathway enrichment results suggested that the mechanism of Danshen in treating anemia was significantly related to the Jak-STAT signaling pathway. Pharmacodynamic results showed that aqueous extract of Danshen (DSAE) and ethanol extract of Danshen (DSEE) markedly enhanced the number of red blood cells, cardiac output, and blood flow velocity. Compared with DSAE, DSEE exerted anti-anemia effects at a lower dose; however, along with higher toxicity. PCR data demonstrated that DSAE and DSEE treatment both upregulated the mRNA expression of erythroid hematopoiesis-related factors in the Epo-JAK-STAT signaling pathway, such as Gata-1, Epo, EpoR, Jak2, STAT3, and STAT5. In general, DSAE exhibited higher activation of this signaling than DSEE. CONCLUSIONS These results indicated that DSAE and DSEE both possess blood-enriching functions related with their ability to promote Jak-STAT signaling. DSAE exerted lower toxicity and attenuated anemia over a wider dose range than DSEE, which suggests that DSAE may be more suitable for the treatment for anemia. These results presented experimental evidence for the clinical use of Danshen as an intervention for anemia, especially in chemotherapy-induced anemia.
Collapse
Affiliation(s)
- Shan He
- School of Pharmacology, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, PR China
| | - Tianqi Wang
- Journal Editorial Board of Science and Technology Department, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Congwei Shi
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, 250355, Shandong, PR China
| | - Zhenguo Wang
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, 250355, Shandong, PR China.
| | - Xianjun Fu
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, 250355, Shandong, PR China; Marine Traditional Chinese Medicine Research Center, Qingdao Academy of Traditional Chinese Medicine Shandong University of Traditional Chinese Medicine, Qingdao, 266114, Shandong, PR China; Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine Jinan, 250355, Shandong, PR China.
| |
Collapse
|
7
|
Thakur R, Suri CR, Kaur IP, Rishi P. Review. Crit Rev Ther Drug Carrier Syst 2022; 40:49-100. [DOI: 10.1615/critrevtherdrugcarriersyst.2022040322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
8
|
Zeng B, Zhao Q, Sun Z, Liu D, Chen H, Li X, Wang J, Xing HR. SEC23A Is an Independent Prognostic Biomarker in Bladder Cancer Correlated With MAPK Signaling. Front Genet 2021; 12:672832. [PMID: 34456965 PMCID: PMC8385657 DOI: 10.3389/fgene.2021.672832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Clinical data mining and bioinformatics analysis can be employed effectively to elucidate the function and underlying mechanisms of the gene of interest. Here, we have proposed a framework for the identification and validation of independent biomarkers in human cancer and for mechanistic profiling using gene sets enrichment analysis and pathway analysis. This is followed by validation with in vitro experiments. Using this framework to analyze the clinical relevance of SEC23A, we have discovered the prognostic potential of SEC23A in different cancers and identified SEC23A as an independent prognostic factor for poor prognosis in bladder cancer, which implicates SEC23A, for the first time, as an oncogene. Bioinformatic analyses have elucidated an association between SEC23A expression and the upregulation of the MAPK signaling pathway. Using the T24 human bladder cell line, we confirmed that knockdown of SEC23A expression could effectively impact the MAPK signaling pathway. Further, through PCR verification, we showed that MEF2A, one of the key genes of the MAPK signaling pathway, might be a downstream factor of the SEC23A gene.
Collapse
Affiliation(s)
- Bin Zeng
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Qiting Zhao
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Zhiwei Sun
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Doudou Liu
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded By Chongqing and the Ministry of Science and Technology, School of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Hao Chen
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded By Chongqing and the Ministry of Science and Technology, School of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Xiaoshuang Li
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded By Chongqing and the Ministry of Science and Technology, School of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Jianyu Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - H Rosie Xing
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded By Chongqing and the Ministry of Science and Technology, School of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Zhang X, Zhang Y, Qiao W, Zhang J, Qi Z. Baricitinib, a drug with potential effect to prevent SARS-COV-2 from entering target cells and control cytokine storm induced by COVID-19. Int Immunopharmacol 2020; 86:106749. [PMID: 32645632 PMCID: PMC7328558 DOI: 10.1016/j.intimp.2020.106749] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/06/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023]
Abstract
In December 2019, a novel coronavirus pneumonia (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) suddenly broke out in China and rapidly spread all over the world. Recently, a cell surface protein, known as angiotensin-converting enzyme II (ACE2), has been identified to be involved in receptor-mediated endocytosis for SARS-CoV-2 entry to the cells. Many studies have reported the clinical characteristics of COVID-19: sudden deterioration of disease around 1-2 weeks after onset; much lower level of lymphocytes, especially natural killer (NK) cells in peripheral blood; extremely high pro-inflammatory cytokines and C reactive protein (CRP). About 15.7% of patients develop severe pneumonia, and cytokine storm is an important factor leading to rapid disease progression. Currently, there are no specific drugs for COVID-19 and the cytokine storm it causes. Baricitinib intracellularly inhibits the proinflammatory signal of several cytokines by suppressing Janus kinase (JAK) JAK1/JAK2. It has been demonstrated clinical benefits for the patients with rheumatoid arthritis (RA), active systemic lupus erythematosus and atopic dermatitis with good efficacy and safety records. Baricitinib is expected to interrupt the passage and intracellular assembly of SARS-CoV-2 into the target cells mediated by ACE2 receptor, and treat cytokine storm caused by COVID-19. Several clinical trials are currently investigating the drug, and one of which has been completed with encouraging results. In this paper, we will elaborate the role of cytokine storm mediated by JAK-STAT pathway in severe COVID-19, the possible mechanisms of baricitinib on reducing the viral entry into the target cells and cytokine storm, the key points of pharmaceutical care based on the latest research reports, clinical trials progress and drug instruction from the US FDA, so as to provide reference for the treatment of severe COVID-19.
Collapse
Affiliation(s)
- Xiuhong Zhang
- Department of Pharmacy, Wuxi People's Hospital Attached to Nanjing Medical University, Wuxi 214023, Jiangsu, China
| | - Yan Zhang
- Research Department, Wuxi Maternal and Child Health Hospital Attached to Nanjing Medical University, Wuxi 214002, Jiangsu, China
| | - Weizhen Qiao
- Center of Clinical Research, Wuxi People's Hospital Attached to Nanjing Medical University, Wuxi 214023, Jiangsu, China
| | - Ji Zhang
- Lung Transplant Center, Wuxi People's Hospital Attached to Nanjing Medical University, Wuxi 214023, Jiangsu, China
| | - Zhigang Qi
- Department of Pharmacy, Wuxi People's Hospital Attached to Nanjing Medical University, Wuxi 214023, Jiangsu, China.
| |
Collapse
|
10
|
Xin P, Xu X, Deng C, Liu S, Wang Y, Zhou X, Ma H, Wei D, Sun S. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int Immunopharmacol 2020; 80:106210. [PMID: 31972425 DOI: 10.1016/j.intimp.2020.106210] [Citation(s) in RCA: 572] [Impact Index Per Article: 114.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/19/2019] [Accepted: 01/08/2020] [Indexed: 02/09/2023]
Abstract
The JAK/STAT signaling pathway is an universally expressed intracellular signal transduction pathway and involved in many crucial biological processes, including cell proliferation, differentiation, apoptosis, and immune regulation. It provides a direct mechanism for extracellular factors-regulated gene expression. Current researches on this pathway have been focusing on the inflammatory and neoplastic diseases and related drug. The mechanism of JAK/STAT signaling is relatively simple. However, the biological consequences of the pathway are complicated due to its crosstalk with other signaling pathways. In addition, there is increasing evidence indicates that the persistent activation of JAK/STAT signaling pathway is closely related to many immune and inflammatory diseases, yet the specific mechanism remains unclear. Therefore, it is necessary to study the detailed mechanisms of JAK/STAT signaling in disease formation to provide critical reference for clinical treatments of the diseases. In this review, we focus on the structure of JAKs and STATs, the JAK/STAT signaling pathway and its negative regulators, the associated diseases, and the JAK inhibitors for the clinical therapy.
Collapse
Affiliation(s)
- Ping Xin
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Xiaoyun Xu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Chengjie Deng
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Shuang Liu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Youzhi Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xuegang Zhou
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Hongxing Ma
- Clinical Laboratory Department, Najing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Najing 211200, China
| | - Donghua Wei
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China
| | - Shiqin Sun
- College of Pharmacy, Harbin Medical University-Daqing, Daqing 163319, China.
| |
Collapse
|
11
|
Cellular and Molecular Mechanisms of Diabetic Atherosclerosis: Herbal Medicines as a Potential Therapeutic Approach. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9080869. [PMID: 28883907 PMCID: PMC5572632 DOI: 10.1155/2017/9080869] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/30/2017] [Accepted: 07/10/2017] [Indexed: 01/09/2023]
Abstract
An increasing number of patients diagnosed with diabetes mellitus eventually develop severe coronary atherosclerosis disease. Both type 1 and type 2 diabetes mellitus increase the risk of cardiovascular disease associated with atherosclerosis. The cellular and molecular mechanisms affecting the incidence of diabetic atherosclerosis are still unclear, as are appropriate strategies for the prevention and treatment of diabetic atherosclerosis. In this review, we discuss progress in the study of herbs as potential therapeutic agents for diabetic atherosclerosis.
Collapse
|
12
|
Recio C, Maione F, Iqbal AJ, Mascolo N, De Feo V. The Potential Therapeutic Application of Peptides and Peptidomimetics in Cardiovascular Disease. Front Pharmacol 2017; 7:526. [PMID: 28111551 PMCID: PMC5216031 DOI: 10.3389/fphar.2016.00526] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/19/2016] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) remains a leading cause of mortality and morbidity worldwide. Numerous therapies are currently under investigation to improve pathological cardiovascular complications, but yet, there have been very few new medications approved for intervention/treatment. Therefore, new approaches to treat CVD are urgently required. Attempts to prevent vascular complications usually involve amelioration of contributing risk factors and underlying processes such as inflammation, obesity, hyperglycaemia, or hypercholesterolemia. Historically, the development of peptides as therapeutic agents has been avoided by the Pharmaceutical industry due to their low stability, size, rate of degradation, and poor delivery. However, more recently, resurgence has taken place in developing peptides and their mimetics for therapeutic intervention. As a result, increased attention has been placed upon using peptides that mimic the function of mediators involved in pathologic processes during vascular damage. This review will provide an overview on novel targets and experimental therapeutic approaches based on peptidomimetics for modulation in CVD. We aim to specifically examine apolipoprotein A-I (apoA-I) and apoE mimetic peptides and their role in cholesterol transport during atherosclerosis, suppressors of cytokine signaling (SOCS)1-derived peptides and annexin-A1 as potent inhibitors of inflammation, incretin mimetics and their function in glucose-insulin tolerance, among others. With improvements in technology and synthesis platforms the future looks promising for the development of novel peptides and mimetics for therapeutic use. However, within the area of CVD much more work is required to identify and improve our understanding of peptide structure, interaction, and function in order to select the best targets to take forward for treatment.
Collapse
Affiliation(s)
- Carlota Recio
- Sir William Dunn School of Pathology, University of Oxford Oxford, UK
| | - Francesco Maione
- Department of Pharmacy, University of Naples Federico II Naples, Italy
| | - Asif J Iqbal
- Sir William Dunn School of Pathology, University of Oxford Oxford, UK
| | - Nicola Mascolo
- Department of Pharmacy, University of Naples Federico II Naples, Italy
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno Salerno, Italy
| |
Collapse
|
13
|
Cai LL, Liu GY, Tzeng CM. Genome-wide DNA methylation profiling and its involved molecular pathways from one individual with thyroid malignant/benign tumor and hyperplasia: A case report. Medicine (Baltimore) 2016; 95:e4695. [PMID: 27583899 PMCID: PMC5008583 DOI: 10.1097/md.0000000000004695] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND During development, methylation permanently changes gene activity, while aberrant gene methylation is key to human tumorigenesis. Gene methylation is an epigenetic event leading to gene silencing and some tumor suppressor genes that are aberrantly methylated in both thyroid cancer and benign thyroid tumor, suggesting a role for methylation in early thyroid tumorigenesis. Specific gene methylation occurs in certain types of thyroid cancer and depends on particular signaling pathways. Most reports rely on data from varied samples that vary tremendously with respect to methylation. RESULTS We observed that hyperplastic/malignant (H/M) thyroid tissue and benign/manligant (B/M) tissue had the most profoundly methylated loci compared to hyperplastic/benign (H/B) tissue. These loci are mapped to 863 genes (|Δβ value| > 0.15) in B/M and 1082 genes (|Δβ value| > 0.15) in H/M. After bioinformatic analysis, these genes were found to be involved in T-cell receptor signaling pathway (B/M) and Jak-Stat signaling pathways (H/M). CONCLUSION Our study offers the most comprehensive DNA methylation data for thyroid disease to date, using 1 patient with 3 tissue types and high-resolution 450K arrays. Our data may lay the foundation for future identification of novel epigenetic targets or diagnosis of thyroid cancer.
Collapse
Affiliation(s)
- Liang-Liang Cai
- Translational Medicine Research Center, School of Pharmaceutical Sciences
| | - Guo-Yan Liu
- Department of Gastrointestinal Surgery, Zhongshan Hospital, Xiamen University
| | - Chi-Meng Tzeng
- Translational Medicine Research Center, School of Pharmaceutical Sciences
- Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation
- INNOVA Cell: TDx Clinics and TRANSLATE Health Group, Xiamen University, China
- Correspondence: Chi-Meng Tzeng, School of Pharmaceutical Sciences, Xiamen University, China (e-mail: )
| |
Collapse
|
14
|
Batchu SN, Xia J, Ko KA, Doyley MM, Abe JI, Morrell CN, Korshunov VA. Axl modulates immune activation of smooth muscle cells in vein graft remodeling. Am J Physiol Heart Circ Physiol 2015; 309:H1048-58. [PMID: 26276821 DOI: 10.1152/ajpheart.00495.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/06/2015] [Indexed: 12/13/2022]
Abstract
The pathophysiological mechanisms of the immune activation of smooth muscle cells are not well understood. Increased expression of Axl, a receptor tyrosine kinase, was recently found in arteries from patients after coronary bypass grafts. In the present study, we hypothesized that Axl-dependent immune activation of smooth muscle cells regulates vein graft remodeling. We observed a twofold decrease in intimal thickening after vascular and systemic depletion of Axl in vein grafts. Local depletion of Axl had the greatest effect on immune activation, whereas systemic deletion of Axl reduced intima due to an increase in apoptosis in vein grafts. Primary smooth muscle cells isolated from Axl knockout mice had reduced proinflammatory responses by prevention of the STAT1 pathway. The absence of Axl increased suppressor of cytokine signaling (SOCS)1 expression in smooth muscle cells, a major inhibitory protein for STAT1. Ultrasound imaging suggested that vascular depletion of Axl reduced vein graft stiffness. Axl expression determined the STAT1-SOCS1 balance in vein graft intima and progression of the remodeling. The results of this investigation demonstrate that Axl promotes STAT1 signaling via inhibition of SOCS1 in activated smooth muscle cells in vein graft remodeling.
Collapse
Affiliation(s)
- Sri N Batchu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Jixiang Xia
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Kyung Ae Ko
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Marvin M Doyley
- Department of Electrical and Computer Engineering, University of Rochester and Hajim School of Engineering and Applied Sciences, Rochester, New York; and
| | - Jun-Ichi Abe
- University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Craig N Morrell
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Vyacheslav A Korshunov
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, New York;
| |
Collapse
|
15
|
Recio C, Oguiza A, Mallavia B, Lazaro I, Ortiz-Muñoz G, Lopez-Franco O, Egido J, Gomez-Guerrero C. Gene delivery of suppressors of cytokine signaling (SOCS) inhibits inflammation and atherosclerosis development in mice. Basic Res Cardiol 2015; 110:8. [DOI: 10.1007/s00395-014-0458-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 01/16/2023]
|
16
|
Muendlein A, Kinz E, Gasser K, Leiherer A, Rein P, Saely CH, Grallert H, Peters A, Fraunberger P, Drexel H, Lang AH. Occurrence of the JAK2 V617F mutation in patients with peripheral arterial disease. Am J Hematol 2015; 90:E17-21. [PMID: 25345590 DOI: 10.1002/ajh.23874] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 10/21/2014] [Indexed: 12/19/2022]
Abstract
The acquired JAK2 V617F mutation is common in patients with myeloproliferative neoplasms. We previously showed that JAK2 V617F is also found in coronary patients, most of them affected by coronary atherosclerosis. Peripheral arterial disease (PAD) is another important manifestation of atherosclerosis. However, prevalence of the JAK2 V617F mutation and its effect on clinical or hematologic characteristics is unknown in PAD patients. In the present study we determined the prevalence of JAK2 V617F in a cohort of 287 patients with sonographically proven PAD and compared mutation frequency with mutational status of 997 healthy people from the KORA F4 study. JAK2 V617F screening and quantification of allele burden in both cohorts was performed with same allele-specific quantitative real-time PCR method. From a total of 287 PAD patients, 9 individuals were tested positive for the JAK2 V617F mutation. One patient showed elevated hemoglobin values, indicating polycythemia vera. Observed JAK2 V617F frequency (3.1%) in PAD patients showed a 5-fold, highly significant increase compared with healthy people (P < 0.001). Furthermore, occurrence of the mutation in PAD patients was significantly decreased in patients using aspirin (P = 0.003). We conclude that the prevalence of JAK2 V617F mutation is significantly increased in PAD patients compared to the general population. Future studies are warranted to confirm our observations and to define the underlying mechanisms behind our findings.
Collapse
Affiliation(s)
- Axel Muendlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT); Feldkirch Austria
| | - Elena Kinz
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT); Feldkirch Austria
- Private University of the Principality of Liechtenstein; Triesen Liechtenstein
| | - Klaus Gasser
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT); Feldkirch Austria
- Private University of the Principality of Liechtenstein; Triesen Liechtenstein
- Department of Medicine and Cardiology; Academic Teaching Hospital Feldkirch; Feldkirch Austria
| | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT); Feldkirch Austria
- Private University of the Principality of Liechtenstein; Triesen Liechtenstein
- Medical Central Laboratories; Feldkirch Austria
| | - Philipp Rein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT); Feldkirch Austria
- Department of Medicine and Cardiology; Academic Teaching Hospital Feldkirch; Feldkirch Austria
| | - Christoph H. Saely
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT); Feldkirch Austria
- Private University of the Principality of Liechtenstein; Triesen Liechtenstein
- Department of Medicine and Cardiology; Academic Teaching Hospital Feldkirch; Feldkirch Austria
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen; German Research Center for Environmental Health; Neuherberg Germany
| | - Annette Peters
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum Muenchen; German Research Center for Environmental Health; Neuherberg Germany
- Institute of Epidemiology II, Helmholtz Zentrum Muenchen; German Research Center for Environmental Health; Neuherberg Germany
- German Research Center for Cardiovascular Disease (DZHK-Munich partner site); Munich Germany
| | | | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT); Feldkirch Austria
- Private University of the Principality of Liechtenstein; Triesen Liechtenstein
- Department of Medicine and Cardiology; Academic Teaching Hospital Feldkirch; Feldkirch Austria
- Drexel College University of Medicine; Philadelphia Pennsylvania
| | - Alois H. Lang
- Department of Medicine and Cardiology; Academic Teaching Hospital Feldkirch; Feldkirch Austria
| |
Collapse
|
17
|
Gan AM, Butoi E, Manea A, Pirvulescu MM, Stan D, Simion V, Calin M, Simionescu M, Manduteanu I. Functional analysis of the fractalkine gene promoter in human aortic smooth muscle cells exposed to proinflammatory conditions. FEBS J 2014; 281:3869-81. [DOI: 10.1111/febs.12921] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 07/01/2014] [Accepted: 07/07/2014] [Indexed: 12/20/2022]
Affiliation(s)
- Ana-Maria Gan
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Elena Butoi
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Adrian Manea
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Monica Madalina Pirvulescu
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Daniela Stan
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Viorel Simion
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Manuela Calin
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Ileana Manduteanu
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| |
Collapse
|
18
|
Recio C, Oguiza A, Lazaro I, Mallavia B, Egido J, Gomez-Guerrero C. Suppressor of cytokine signaling 1-derived peptide inhibits Janus kinase/signal transducers and activators of transcription pathway and improves inflammation and atherosclerosis in diabetic mice. Arterioscler Thromb Vasc Biol 2014; 34:1953-60. [PMID: 25012131 DOI: 10.1161/atvbaha.114.304144] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Activation of Janus kinase/signal transducers and activators of transcription (STAT) pathway by hyperglycemia and dislypidemia contributes to the progression of diabetic complications, including atherosclerosis. Suppressor of cytokine signaling (SOCS) proteins negatively regulate Janus kinase/STAT and have emerged as promising target for anti-inflammatory therapies. We investigated whether a cell-permeable lipopeptide corresponding to the kinase inhibitory region of SOCS1 could reduce atherosclerosis in diabetic mice and identified the mechanisms involved. APPROACH AND RESULTS Streptozotocin-induced diabetic apolipoprotein E-deficient mice (aged 8 and 22 weeks) were given intraperitoneal injections of vehicle, SOCS1-derived peptide, or control mutant peptide for 6 to 10 weeks. SOCS1 therapy suppressed STAT1/STAT3 activation in atherosclerotic plaques of diabetic mice and significantly reduced lesion size at both early and advanced stages of lesion development compared with vehicle group. Plaque characterization demonstrated that SOCS1 peptide decreased the accumulation of lipids, macrophages, and T lymphocytes, whereas increasing collagen and smooth muscle cell content. This atheroprotective effect was accompanied by systemic (reduced proinflammatory Ly6C(high) monocytes and splenic cytokine expression) and local (reduced aortic expression of chemokines and cytokines) mechanisms, without impact on metabolic parameters. In vitro, SOCS1 peptide dose dependently inhibited STAT1/STAT3 activation and target gene expression in vascular smooth muscle cells and macrophages and also suppressed cytokine-induced cell migration and adhesion processes. CONCLUSIONS SOCS1-based targeting Janus kinase/STAT restrains key mechanisms of atherogenesis in diabetic mice, thereby preventing plaque formation and increasing plaque stability. Approaches to mimic native SOCS1 functions may have a therapeutic potential to retard the progression of diabetic complications.
Collapse
Affiliation(s)
- Carlota Recio
- From the Renal and Vascular Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain (C.R., A.O., I.L., B.M., J.E., C.G.-G.); and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain (C.R., A.O., J.E., C.G.-G.)
| | - Ainhoa Oguiza
- From the Renal and Vascular Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain (C.R., A.O., I.L., B.M., J.E., C.G.-G.); and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain (C.R., A.O., J.E., C.G.-G.)
| | - Iolanda Lazaro
- From the Renal and Vascular Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain (C.R., A.O., I.L., B.M., J.E., C.G.-G.); and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain (C.R., A.O., J.E., C.G.-G.)
| | - Beñat Mallavia
- From the Renal and Vascular Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain (C.R., A.O., I.L., B.M., J.E., C.G.-G.); and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain (C.R., A.O., J.E., C.G.-G.)
| | - Jesus Egido
- From the Renal and Vascular Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain (C.R., A.O., I.L., B.M., J.E., C.G.-G.); and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain (C.R., A.O., J.E., C.G.-G.)
| | - Carmen Gomez-Guerrero
- From the Renal and Vascular Research Lab, IIS-Fundacion Jimenez Diaz, Autonoma University of Madrid, Madrid, Spain (C.R., A.O., I.L., B.M., J.E., C.G.-G.); and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain (C.R., A.O., J.E., C.G.-G.).
| |
Collapse
|
19
|
Gan AM, Pirvulescu MM, Stan D, Simion V, Calin M, Manduteanu I, Butoi E. Monocytes and smooth muscle cells cross-talk activates STAT3 and induces resistin and reactive oxygen species and production. J Cell Biochem 2013; 114:2273-83. [DOI: 10.1002/jcb.24571] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 04/12/2013] [Indexed: 01/25/2023]
Affiliation(s)
- Ana Maria Gan
- Department of Biopathology and Therapy of Inflammation; Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy; Bucharest; Romania
| | - Monica Madalina Pirvulescu
- Department of Biopathology and Therapy of Inflammation; Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy; Bucharest; Romania
| | - Daniela Stan
- Department of Biopathology and Therapy of Inflammation; Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy; Bucharest; Romania
| | - Viorel Simion
- Department of Biopathology and Therapy of Inflammation; Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy; Bucharest; Romania
| | - Manuela Calin
- Department of Biopathology and Therapy of Inflammation; Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy; Bucharest; Romania
| | - Ileana Manduteanu
- Department of Biopathology and Therapy of Inflammation; Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy; Bucharest; Romania
| | - Elena Butoi
- Department of Biopathology and Therapy of Inflammation; Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy; Bucharest; Romania
| |
Collapse
|
20
|
Inflammatory effects of resistin on human smooth muscle cells: up-regulation of fractalkine and its receptor, CX3CR1 expression by TLR4 and Gi-protein pathways. Cell Tissue Res 2012; 351:161-74. [PMID: 23086480 DOI: 10.1007/s00441-012-1510-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/20/2012] [Indexed: 10/27/2022]
Abstract
In the atherosclerotic plaque, smooth muscle cells (SMC) acquire an inflammatory phenotype. Resistin and fractalkine (CX3CL1) are found in human atheroma and not in normal arteries. CX3CL1 and CX3CR1 are predominately associated with SMC. We have questioned whether resistin has a role in the expression of CX3CL1 and CX3CR1 in SMC thus contributing to the pro-inflammatory status of these cells. Cultured human aortic SMC were stimulated with 100 ng/ml resistin for 4, 6, 12, and 24 h, and then CX3CL1 and CX3CR1 expression was assessed by quantitative reverse transcription with the polymerase chain reaction and Western blot. We found that resistin up-regulated CX3CL1 and CX3CR1 in SMC and induced the phosphorylation of p38MAPK and STAT3. Inhibitors of p38MAPK, JAK-STAT, NF-kB, and AP-1 significantly reduced CX3CL1 and CX3CR1 expression. Knockdown of STAT1 and STAT3 with decoy oligodeoxinucleotides and the silencing of p65 and cjun with short interfering RNA decreased CX3CL1 and CX3CR1 expression. Anti-TLR4 antibody and pertussis toxin also reduced CX3CL1 and CX3CR1 protein expression. xCELLigence experiments revealed that resistin probably uses Gi-proteins for its effect on SMC. The CX3CL1 induced by resistin exhibited a chemotactic effect on monocyte transmigration. Thus, (1) resistin contributes to the pro-inflammatory state of SMC by the up-regulation of CX3CL1 and CX3CR1 expression via a mechanism involving NF-kB, AP-1, and STAT1/3 transcription factors, (2) resistin employs TLR4 and Gi-protein signaling for its effect on SMC, (3) CX3CL1 induced by resistin is functional in monocyte chemotaxis. The data reveal new mechanisms by which resistin promotes the inflammatory phenotype of SMC.
Collapse
|
21
|
Kraus M, Wang Y, Aleksandrowicz D, Bachman E, Szewczak AA, Walker D, Xu L, Bouthillette M, Childers KM, Dolinski B, Haidle AM, Kopinja J, Lee L, Lim J, Little KD, Ma Y, Mathur A, Mo JR, O’Hare E, Otte RD, Taoka BM, Wang W, Yin H, Zabierek AA, Zhang W, Zhao S, Zhu J, Young JR, Marshall CG. Efficacious intermittent dosing of a novel JAK2 inhibitor in mouse models of polycythemia vera. PLoS One 2012; 7:e37207. [PMID: 22623993 PMCID: PMC3356383 DOI: 10.1371/journal.pone.0037207] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 04/16/2012] [Indexed: 01/08/2023] Open
Abstract
A high percentage of patients with the myeloproliferative disorder polycythemia vera (PV) harbor a Val617→Phe activating mutation in the Janus kinase 2 (JAK2) gene, and both cell culture and mouse models have established a functional role for this mutation in the development of this disease. We describe the properties of MRLB-11055, a highly potent inhibitor of both the WT and V617F forms of JAK2, that has therapeutic efficacy in erythropoietin (EPO)-driven and JAK2V617F-driven mouse models of PV. In cultured cells, MRLB-11055 blocked proliferation and induced apoptosis in a manner consistent with JAK2 pathway inhibition. MRLB-11055 effectively prevented EPO-induced STAT5 activation in the peripheral blood of acutely dosed mice, and could prevent EPO-induced splenomegaly and erythrocytosis in chronically dosed mice. In a bone marrow reconstituted JAK2V617F-luciferase murine PV model, MRLB-11055 rapidly reduced the burden of JAK2V617F-expressing cells from both the spleen and the bone marrow. Using real-time in vivo imaging, we examined the kinetics of disease regression and resurgence, enabling the development of an intermittent dosing schedule that achieved significant reductions in both erythroid and myeloid populations with minimal impact on lymphoid cells. Our studies provide a rationale for the use of non-continuous treatment to provide optimal therapy for PV patients.
Collapse
Affiliation(s)
- Manfred Kraus
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Yuxun Wang
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Dan Aleksandrowicz
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Eric Bachman
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Alexander A. Szewczak
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Deborah Walker
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Lin Xu
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Melaney Bouthillette
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Kaleen M. Childers
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Brian Dolinski
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Andrew M. Haidle
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Johnny Kopinja
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Linda Lee
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Jongwon Lim
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Kevin D. Little
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Yanhong Ma
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Anjili Mathur
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Jan-Rung Mo
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Erin O’Hare
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Ryan D. Otte
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Brandon M. Taoka
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Wenxian Wang
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Hong Yin
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Anna A. Zabierek
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Weisheng Zhang
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Shuxia Zhao
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Joe Zhu
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
| | - Jonathan R. Young
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
- * E-mail: (CGM); (JRY)
| | - C. Gary Marshall
- Departments of DMPK, in vitro Sciences, in vivo Sciences, Medicinal Chemistry, Basic Pharmaceutical Sciences and Oncology, Merck Research Laboratories, Boston, Massachusetts, United States of America
- * E-mail: (CGM); (JRY)
| |
Collapse
|
22
|
Alves ES, Haidar AA, Quadros CD, Carvalho DS, Morgan D, Rocha MS, Curi R, Carpinelli AR, Hirata AE. Angiotensin II-induced JNK activation is mediated by NAD(P)H oxidase in isolated rat pancreatic islets. ACTA ACUST UNITED AC 2012; 175:1-6. [PMID: 22280799 DOI: 10.1016/j.regpep.2012.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 12/14/2011] [Accepted: 01/10/2012] [Indexed: 10/14/2022]
Abstract
Angiotensin II (AII), the active component of the renin angiotensin system (RAS), plays a vital role in the regulation of physiological processes of the cardiovascular system, but also has autocrine and paracrine actions in various tissues and organs. Many studies have shown the existence of RAS in the pancreas of humans and rodents. The aim of this study was to evaluate potential signaling pathways mediated by AII in isolated pancreatic islets of rats. Phosphorylation of MAPKs (ERK1/2, JNK and p38MAPK), and the interaction between proteins JAK/STAT were evaluated. AII increased JAK2/STAT1 (42%) and JAK2/STAT3 (100%) interaction without altering the total content of JAK2. Analyzing the activation of MAPKs (ERK1/2, JNK and p38MAPK) in isolated pancreatic islets from rats we observed that AII rapidly (3 min) promoted a significant increase in the phosphorylation degree of these proteins after incubation with the hormone. Curiously JNK protein phosphorylation was inhibited by DPI, suggesting the involvement of NAD(P)H oxidase in the activation of protein.
Collapse
Affiliation(s)
- E S Alves
- Department of Physiology, Federal University of Sao Paulo-UNIFESP, São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Fenyo IM, Florea IC, Raicu M, Manea A. Tyrphostin AG490 reduces NAPDH oxidase activity and expression in the aorta of hypercholesterolemic apolipoprotein E-deficient mice. Vascul Pharmacol 2011; 54:100-6. [DOI: 10.1016/j.vph.2011.03.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 03/01/2011] [Accepted: 03/24/2011] [Indexed: 11/28/2022]
|
24
|
NADPH oxidase-derived reactive oxygen species: involvement in vascular physiology and pathology. Cell Tissue Res 2010; 342:325-39. [PMID: 21052718 DOI: 10.1007/s00441-010-1060-y] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 09/13/2010] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species (ROS) are essential mediators of normal cell physiology. However, in the last few decades, it has become evident that ROS overproduction and/or alterations of the antioxidant system associated with inflammation and metabolic dysfunction are key pathological triggers of cardiovascular disorders. NADPH oxidases (Nox) represent a class of hetero-oligomeric enzymes whose primary function is the generation of ROS. In the vasculature, Nox-derived ROS contribute to the maintenance of vascular tone and regulate important processes such as cell growth, proliferation, differentiation, apoptosis, cytoskeletal organization, and cell migration. Under pathological conditions, excessive Nox-dependent ROS formation, which is generally associated with the up-regulation of different Nox subtypes, induces dysregulation of the redox control systems and promotes oxidative injury of the cardiovascular cells. The molecular mechanism of Nox-derived ROS generation and the means by which this class of molecule contributes to vascular damage remain debatable issues. This review focuses on the processes of ROS formation, molecular targets, and neutralization in the vasculature and provides an overview of the novel concepts regarding Nox functions, expression, and regulation in vascular health and disease. Because Nox enzymes are the most important sources of ROS in the vasculature, therapeutic perspectives to counteract Nox-dependent oxidative stress in the cardiovascular system are discussed.
Collapse
|
25
|
Manea A, Tanase LI, Raicu M, Simionescu M. Jak/STAT signaling pathway regulates nox1 and nox4-based NADPH oxidase in human aortic smooth muscle cells. Arterioscler Thromb Vasc Biol 2009; 30:105-12. [PMID: 19834108 DOI: 10.1161/atvbaha.109.193896] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Oxidative stress mediated by Nox1- and Nox4-based NADPH oxidase (Nox) plays a key role in vascular diseases. The molecular mechanisms involved in the regulation of Nox are not entirely elucidated. Because JAK/STAT regulates many genes linked to inflammation, cell proliferation, and differentiation, we questioned whether this pathway is involved in the regulation of Nox1 and Nox4 in human aortic smooth muscle cells (SMCs). METHODS AND RESULTS Cultured SMCs were exposed to interferon gamma (IFNgamma) for 24 hours. Using lucigenin-enhanced chemiluminescence and dihydroethidium assays, real-time polymerase chain reaction, and Western blot analysis, we found that JAK/STAT inhibitors significantly diminished the IFNgamma-dependent upregulation of Nox activity, Nox1 and Nox4 expression. In silico analysis revealed the presence of highly conserved GAS elements within human Nox1, Nox4, p22phox, p47phox, and p67phox promoters. Transient overexpression of STAT1/STAT3 augmented the promoter activities of each subunit. JAK/STAT blockade reduced the Nox subunits transcription. Chromatin immunoprecipitation demonstrated the physical interaction of STAT1/STAT3 proteins with the predicted GAS elements from Nox1 and Nox4 promoters. CONCLUSIONS JAK/STAT is a key regulator of Nox1 and Nox4 in human vascular SMCs. Inhibition of JAK/STAT pathway and the consequent Nox-dependent oxidative stress may be an efficient therapeutic strategy to reduce atherogenesis.
Collapse
Affiliation(s)
- Adrian Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu," 8, BP Hasdeu St, Bucharest, PO Box 35-14, Romania
| | | | | | | |
Collapse
|