1
|
Fang Y, Guo Z, Zhou L, Zhang J, Li H, Hao J. Pim1 inactivating induces RUNX3 upregulation that improves/alleviates airway inflammation and mucus hypersecretion in vitro and in vivo. BMJ Open Respir Res 2024; 11:e002066. [PMID: 39521608 PMCID: PMC11552021 DOI: 10.1136/bmjresp-2023-002066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/11/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE Our research aimed to evaluate whether proto-oncogene serine/threonine-protein kinase Pim-1 (Pim1) inactivation could attenuate asthma by promoting runt-related transcription factor 3 (Runx3) expression and explore the underlying molecular mechanism. METHOD Phorbol 12-myristate 13-acetate (PMA, 50 nM) was used to induce inflammation in BEAS-2B human airway epithelial cells. ELISA and immunofluorescence double staining confirmed inflammation modelling and differential expression of Pim1 and Runx3. Pim1 inhibitor (SGI-1776) and Runx3 siRNA (siRunx3) were used in this study. Apoptosis, inflammation, MUC5AC protein expression, Pim1 kinase and Runx3 protein expression, and PI3K/AKT/nuclear factor-κB (NF-κB) pathway-associated protein expression were also assessed by flow cytometry, immunofluorescence and western blot. The effects of Pim1 inactivation on airway inflammation, pathological injury and mucus secretion in wild-type and Runx3 knockout mice were observed by in vivo experiments. RESULTS The results of the in vitro experiments showed that PMA stimulation causes BEAS-2B cell apoptosis and promotes the MUC5AC expression. In addition, PMA stimulation activated the PI3K/AKT/NF-κB pathway. SGI-1776 treatment partially reversed these effects, whereas siRunx3 attenuated the effects of SGI-1776 on PMA-stimulated BEAS-2B cells. In vivo experiments showed that in Runx3-KO asthmatic mice, inhibition of Pim1 kinase had less effect on airway inflammation, pathological injury and mucus secretion. Meanwhile, Pim1 kinase expression was higher in Runx3-KO asthmatic mice than in wild-type asthmatic mice. Furthermore, inhibition of Pim1 kinase inhibited activation of the PI3K/AKT/NF-κB pathway, whereas these effects were attenuated in Runx3-KO mice. CONCLUSION Our results suggest that Pim1 inactivation can ameliorate airway inflammation and mucus hypersecretion through upregulation of Runx3 and the effect could be mediated through modulation of the PI3K/AKT/NF-κB pathway.
Collapse
Affiliation(s)
- Yanni Fang
- Department of Pediatrics, Yantaishan Hospital, Yantai, Shandong, China
| | - Zhen Guo
- Department of Pediatrics, Yantaishan Hospital, Yantai, Shandong, China
| | - Lanzhi Zhou
- Department of Pediatrics, Yantaishan Hospital, Yantai, Shandong, China
| | - Juan Zhang
- Department of Pediatrics, Yantaishan Hospital, Yantai, Shandong, China
| | - Haiyan Li
- Department of Pediatrics, Yantaishan Hospital, Yantai, Shandong, China
| | - Jumei Hao
- Department of Pediatrics, Yantaishan Hospital, Yantai, Shandong, China
| |
Collapse
|
2
|
Baek HS, Kim N, Park JW, Kwon TK, Kim S. The role of Pim-1 kinases in inflammatory signaling pathways. Inflamm Res 2024; 73:1671-1685. [PMID: 39079978 PMCID: PMC11457682 DOI: 10.1007/s00011-024-01924-2] [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: 04/26/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 10/02/2024] Open
Abstract
OBJECTIVE AND DESIGN This observational study investigated the regulatory mechanism of Pim-1 in inflammatory signaling pathways. MATERIALS THP-1, RAW 264.7, BV2, and Jurkat human T cell lines were used. TREATMENT None. METHODS Lipopolysaccharide (LPS) was used to induce inflammation, followed by PIM1 knockdown. Western blot, immunoprecipitation, immunofluorescence, and RT-PCR assays were used to assess the effect of PIM1 knockdown on LPS-induced inflammation. RESULTS PIM1 knockdown in macrophage-like THP-1 cells suppressed LPS-induced upregulation of pro-inflammatory cytokines, inducible nitric oxide synthase, cyclooxygenase-2, phosphorylated Janus kinase, signal transducer and activator of transcription 3, extracellular signal-regulated kinase, c-Jun N-terminal kinase, p38, and nuclear factor kappa B p65 (NF-κB p65). It also suppressed upregulation of inhibitor of NF-κB kinase α/β and enhanced the nuclear translocation of NF-κB p65. Moreover, it inhibited the upregulation of Nod-like receptor family pyrin domain-containing 3 (NLRP3) and cleavage of caspase-1 induced by co-treatment of LPS with adenosine triphosphate. Additionally, p-transforming growth factor-β-activated kinase 1 (TAK1) interacted with Pim-1. All three members of Pim kinases (Pim-1, Pim-2, and Pim-3) were required for LPS-mediated inflammation in macrophages; however, unlike Pim-1 and Pim-3, Pim-2 functioned as a negative regulator of T cell activity. CONCLUSIONS Pim-1 interacts with TAK1 in LPS-induced inflammatory responses and is involved in MAPK/NF-κB/NLRP3 signaling pathways. Additionally, considering the negative regulatory role of Pim-2 in T cells, further in-depth studies on their respective functions are needed.
Collapse
Affiliation(s)
- Hye Suk Baek
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam University, Gwangju, 61469, Republic of Korea
| | - Jong Wook Park
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
- Institute of Medical Science, Keimyung University, Daegu, 42601, Republic of Korea
- Institute for Cancer Research, Keimyung University Dongsan Medical Center, Dalseo-gu, Daegu, 42601, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
- Institute of Medical Science, Keimyung University, Daegu, 42601, Republic of Korea
- Institute for Cancer Research, Keimyung University Dongsan Medical Center, Dalseo-gu, Daegu, 42601, Republic of Korea
- Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, 42601, Republic of Korea
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea.
- Institute of Medical Science, Keimyung University, Daegu, 42601, Republic of Korea.
- Center for Forensic Pharmaceutical Science, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
3
|
Yang X, Liu C, Lei Y, Liu Z, Zhu B, Zhao D. PIM1 signaling in immunoinflammatory diseases: an emerging therapeutic target. Front Immunol 2024; 15:1443784. [PMID: 39372407 PMCID: PMC11449710 DOI: 10.3389/fimmu.2024.1443784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
PIM1, the proviral integration site for Moloney murine leukemia virus, is a member of the serine/threonine protein kinase family. It is involved in many biological events, such as cell survival, cell cycle progression, cell proliferation, and cell migration, and has been widely studied in malignant diseases. However, recent studies have shown that PIM1 plays a prominent role in immunoinflammatory diseases, including autoimmune uveitis, inflammatory bowel disease, asthma, and rheumatoid arthritis. PIM1 can function in inflammatory signal transduction by phosphorylating multiple inflammatory protein substrates and mediating macrophage activation and T lymphocyte cell specification, thus participating in the development of multiple immunoinflammatory diseases. Moreover, the inhibition of PIM1 has been demonstrated to ameliorate certain immunoinflammatory disorders. Based on these studies, we suggest PIM1 as a potential therapeutic target for immunoinflammatory diseases and a valid candidate for future research. Herein, for the first time, we provide a detailed review that focuses on the roles of PIM1 in the pathogenesis of immunoinflammatory diseases.
Collapse
Affiliation(s)
- Xue Yang
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunming Liu
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuxi Lei
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhi Liu
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Bin Zhu
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Dongchi Zhao
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
4
|
Green JR, Mahalingaiah PKS, Gopalakrishnan SM, Liguori MJ, Mittelstadt SW, Blomme EAG, Van Vleet TR. Off-target pharmacological activity at various kinases: Potential functional and pathological side effects. J Pharmacol Toxicol Methods 2023; 123:107468. [PMID: 37553032 DOI: 10.1016/j.vascn.2023.107468] [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: 03/23/2023] [Revised: 06/16/2023] [Accepted: 08/01/2023] [Indexed: 08/10/2023]
Abstract
In drug discovery, during the lead optimization and candidate characterization stages, novel small molecules are frequently evaluated in a battery of in vitro pharmacology assays to identify potential unintended, off-target interactions with various receptors, transporters, ion channels, and enzymes, including kinases. Furthermore, these screening panels may also provide utility at later stages of development to provide a mechanistic understanding of unexpected safety findings. Here, we present a compendium of the most likely functional and pathological outcomes associated with interaction(s) to a panel of 95 kinases based on an extensive curation of the scientific literature. This panel of kinases was designed by AbbVie based on safety-related data extracted from the literature, as well as from over 20 years of institutional knowledge generated from discovery efforts. For each kinase, the scientific literature was reviewed using online databases and the most often reported functional and pathological effects were summarized. This work should serve as a practical guide for small molecule drug discovery scientists and clinical investigators to predict and/or interpret adverse effects related to pharmacological interactions with these kinases.
Collapse
Affiliation(s)
- Jonathon R Green
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States.
| | | | - Sujatha M Gopalakrishnan
- Drug Discovery Science and Technology, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Michael J Liguori
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Scott W Mittelstadt
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Eric A G Blomme
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| | - Terry R Van Vleet
- Departments of Preclinical Safety, AbbVie, 1 North Waukegan Road, North Chicago, IL 60064, United States
| |
Collapse
|
5
|
Koshman YE, Bielinski AL, Bird BM, Green JR, Kowalkowski KL, Lai-Zhang J, Mahalingaiah PK, Sawicki JW, Talaty NN, Wilsey AS, Zafiratos MT, Van Vleet TR. Disconnect between COX-2 selective inhibition and cardiovascular risk in preclinical models. J Pharmacol Toxicol Methods 2023; 120:107251. [PMID: 36792039 DOI: 10.1016/j.vascn.2023.107251] [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: 07/19/2022] [Revised: 11/20/2022] [Accepted: 01/17/2023] [Indexed: 02/15/2023]
Abstract
INTRODUCTION Secondary pharmacology profiling is routinely applied in pharmaceutical drug discovery to investigate the pharmaceutical effects of a drug at molecular targets distinct from (off-target) the intended therapeutic molecular target (on-target). Data from a randomized, placebo-controlled clinical trial, the APPROVe (Adenomatous Polyp Prevention on VIOXX, rofecoxib) trial, raised significant concerns about COX-2 inhibition as a primary or secondary target, shaping the screening and decision-making processes of some pharmaceutical companies. COX-2 is often included in off-target screens due to cardiovascular (CV) safety concerns about secondary interactions with this target. Several potential mechanisms of COX-2-mediated myocardial infarctions have been considered including, effects on platelet stickiness/aggregation, vasal tone and blood pressure, and endothelial cell activation. In the present study, we focused on each of these mechanisms as potential effects of COX-2 inhibitors, to find evidence of mechanism using various in vitro and in vivo preclinical models. METHODS Compounds tested in the study, with a range of COX-2 selectivity, included rofecoxib, celecoxib, etodolac, and meloxicam. Compounds were screened for inhibition of COX-2 vs COX-1 enzymatic activity, ex vivo platelet aggregation (using whole blood from multiple species), ex vivo canine femoral vascular ring model, in vitro human endothelial cell activation (with and without COX-2 induction), and in vivo cardiovascular assessment (anesthetized dog). RESULTS The COX-2 binding assessment generally confirmed the COX-2 selectivity previously reported. COX-2 inhibitors did not have effects on platelet function (spontaneous aggregation or inhibition of aggregation), cardiovascular parameters (mean arterial pressure, heart rate, and left ventricular contractility), or endothelial cell activation. However, rofecoxib uniquely produced an endothelial mediated constriction response in canine femoral arteries. CONCLUSION Our data suggest that rofecoxib-related cardiovascular events in humans are not predicted by COX-2 potency or selectivity. In addition, the vascular ring model suggested possible adverse cardiovascular effects by COX-2 inhibitors, although these effects were not seen in vivo studies. These results may also suggest that COX-2 inhibition alone is not responsible for rofecoxib-mediated adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Yevgeniya E Koshman
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America.
| | - Aimee L Bielinski
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Brandan M Bird
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Jonathon R Green
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Kenneth L Kowalkowski
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Jie Lai-Zhang
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | | | - James W Sawicki
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Nari N Talaty
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Amanda S Wilsey
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Mark T Zafiratos
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Terry R Van Vleet
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| |
Collapse
|
6
|
The Role of NLRP3 Inflammasome in Lupus Nephritis. Int J Mol Sci 2021; 22:ijms222212476. [PMID: 34830358 PMCID: PMC8625721 DOI: 10.3390/ijms222212476] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/15/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023] Open
Abstract
Lupus nephritis (LN) is the most frequent and severe of systemic lupus erythematosus (SLE) clinical manifestations and contributes to the increase of morbidity and mortality of patients due to chronic kidney disease. The NLRP3 (NLR pyrin domain containing 3) is a member of the NLR (NOD-like receptors), and its activation results in the production of pro-inflammatory cytokines, which can contribute to the pathogenesis of LN. In this review manuscript, we approach the relation between the NLRP3 inflammasome, SLE, and LN, highlighting the influence of genetic susceptibility of NLRP3 polymorphisms in the disease; the main functional studies using cellular and animal models of NLRP3 activation; and finally, some mechanisms of NLRP3 inhibition for the development of possible therapeutic drugs for LN.
Collapse
|
7
|
Pritzl CJ, Daniels MA, Teixeiro E. Interplay of Inflammatory, Antigen and Tissue-Derived Signals in the Development of Resident CD8 Memory T Cells. Front Immunol 2021; 12:636240. [PMID: 34234771 PMCID: PMC8255970 DOI: 10.3389/fimmu.2021.636240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/29/2021] [Indexed: 12/21/2022] Open
Abstract
CD8 positive, tissue resident memory T cells (TRM) are a specialized subset of CD8 memory T cells that surveil tissues and provide critical first-line protection against tumors and pathogen re-infection. Recently, much effort has been dedicated to understanding the function, phenotype and development of TRM. A myriad of signals is involved in the development and maintenance of resident memory T cells in tissue. Much of the initial research focused on the roles tissue-derived signals play in the development of TRM, including TGFß and IL-33 which are critical for the upregulation of CD69 and CD103. However, more recent data suggest further roles for antigenic and pro-inflammatory cytokines. This review will focus on the interplay of pro-inflammatory, tissue and antigenic signals in the establishment of resident memory T cells.
Collapse
Affiliation(s)
| | | | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, United States
| |
Collapse
|
8
|
Barberis C, Erdman P, Czekaj M, Fire L, Pribish J, Tserlin E, Maniar S, Batchelor JD, Liu J, Patel VF, Hebert A, Levit M, Wang A, Sun F, Huang SMA. Discovery of SARxxxx92, a pan-PIM kinase inhibitor, efficacious in a KG1 tumor model. Bioorg Med Chem Lett 2020; 30:127625. [PMID: 33096160 DOI: 10.1016/j.bmcl.2020.127625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/10/2020] [Accepted: 10/14/2020] [Indexed: 11/25/2022]
Abstract
N-substituted azaindoles were discovered as potent pan-PIM inhibitors. Lead optimization, guided by structure and focused on physico-chemical properties allowed us to solve inherent hERG and permeability liabilities, and provided compound 27, which subsequently impacted KG-1 tumor growth in a mouse model.
Collapse
Affiliation(s)
- Claude Barberis
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States.
| | - Paul Erdman
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States; Present address: AbbVie, 100 Abbott Park Road, Abbott Park, IL 60064-3500, United States
| | - Mark Czekaj
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Luke Fire
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States; Present address: Rakuten Medical, 11080 Roselle St, San Diego, CA 92121, United States
| | - James Pribish
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Elina Tserlin
- Present address: Qiagen, 561 Virginia Road, Concord, MA 01742, United States
| | - Sachin Maniar
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Joseph D Batchelor
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Jinyu Liu
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Vinod F Patel
- Present address: TME Therapeutics, 3 Mossy Lane, Acton, MA 01720, United States
| | - Andrew Hebert
- Oncology Biochemistry, Sanofi, 270 Albany Street, Cambridge MA 02139, United States
| | - Mikhail Levit
- Oncology Biochemistry, Sanofi, 270 Albany Street, Cambridge MA 02139, United States
| | - Anlai Wang
- Oncology Biology, Sanofi, 270 Albany Street, Cambridge MA 02139, United States
| | - Frank Sun
- Oncology Pharmacology, Sanofi, 640 Memorial Drive, Cambridge MA 02139, United States
| | - Shih-Min A Huang
- Oncology Biology, Sanofi, 270 Albany Street, Cambridge MA 02139, United States; Present address: Bristol-Myers Squibb, 3551 Lawrenceville Princeton, Lawrence Township, NJ 08648, United States
| |
Collapse
|
9
|
Tang H, Gao Y, Li Z, Miao Y, Huang Z, Liu X, Xie L, Li H, Wen W, Zheng Y, Su W. The noncoding and coding transcriptional landscape of the peripheral immune response in patients with COVID-19. Clin Transl Med 2020; 10:e200. [PMID: 33135345 PMCID: PMC7548099 DOI: 10.1002/ctm2.200] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND COVID-19 is currently a global pandemic, but the response of human immune system to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection remains unclear. Noncoding RNAs serve as immune regulators and thus may play a critical role in disease progression. METHODS We performed multi-transcriptome sequencing of both noncoding RNAs and mRNAs isolated from the red blood cell depleted whole blood of moderate and severe COVID-19 patients. The functions of noncoding RNAs were validated by analyses of the expression of downstream mRNAs. We further utilized the single-cell RNA-seq data of COVID-19 patients from Wilk et al. and Chua et al. to characterize noncoding RNA functions in different cell types. RESULTS We defined four types of microRNAs with different expression tendencies that could serve as biomarkers for COVID-19 progress. We also identified miR-146a-5p, miR-21-5p, miR-142-3p, and miR-15b-5p as potential contributors to the disease pathogenesis, possibly serving as biomarkers of severe COVID-19 and as candidate therapeutic targets. In addition, the transcriptome profiles consistently suggested hyperactivation of the immune response, loss of T-cell function, and immune dysregulation in severe patients. CONCLUSIONS Collectively, these findings provide a comprehensive view of the noncoding and coding transcriptional landscape of peripheral immune cells during COVID-19, furthering our understanding and offering novel insights into COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Hao Tang
- Department of Respiratory and Critical Care Medicine Changzheng HospitalSecond Military Medical UniversityShanghaiChina
- Department of Critical CareWuhan Huo Shen Shan HospitalHubeiChina
| | - Yuehan Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Yushan Miao
- Department of Respiratory and Critical Care Medicine Changzheng HospitalSecond Military Medical UniversityShanghaiChina
| | - Zhaohao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Xiuxing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Lihui Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - He Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Wen Wen
- National Center for Liver CancerSecond Military Medical UniversityShanghaiChina
| | - Yingfeng Zheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
10
|
Lan F, Zhang N, Bachert C, Zhang L. Stability of regulatory T cells in T helper 2-biased allergic airway diseases. Allergy 2020; 75:1918-1926. [PMID: 32124987 DOI: 10.1111/all.14257] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/11/2022]
Abstract
Regulatory T (Treg) cells potentially suppress the deleterious activities of effector T cells and maintain a state of tolerance against antigens in the airway mucosa. A decrease in the number and function of Treg cells is observed in T helper 2 (Th2)-biased allergic airway diseases. However, adoptive transfer of naturally occurring Treg (tTreg) cells or peripherally derived Treg (pTreg) cells in asthmatic mouse models did not yield satisfactory results in any previous studies. Here, we review the recent progress in the identification and plasticity of tTreg and pTreg cells in Th2-biased airway diseases and summarize the factors affecting the stability and function of Treg cells. This review may serve as foundation for understanding the molecular mechanisms underlying the stability of tTreg and pTreg cells and development of effective strategies for treating allergic airway diseases.
Collapse
Affiliation(s)
- Feng Lan
- Department of Otolaryngology Head and Neck Surgery Beijing TongRen HospitalCapital Medical University Beijing China
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
| | - Nan Zhang
- Upper Airways Research Laboratory ENT Department Ghent University Ghent Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory ENT Department Ghent University Ghent Belgium
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery Beijing TongRen HospitalCapital Medical University Beijing China
- Beijing Key Laboratory of Nasal Disease Beijing Institute of Otolaryngology Beijing China
| |
Collapse
|
11
|
Baek HS, Min HJ, Hong VS, Kwon TK, Park JW, Lee J, Kim S. Anti-Inflammatory Effects of the Novel PIM Kinase Inhibitor KMU-470 in RAW 264.7 Cells through the TLR4-NF-κB-NLRP3 Pathway. Int J Mol Sci 2020; 21:ijms21145138. [PMID: 32698512 PMCID: PMC7403980 DOI: 10.3390/ijms21145138] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
PIM kinases, a small family of serine/threonine kinases, are important intermediates in the cytokine signaling pathway of inflammatory disease. In this study, we investigated whether the novel PIM kinase inhibitor KMU-470, a derivative of indolin-2-one, inhibits lipopolysaccharide (LPS)-induced inflammatory responses in RAW 264.7 cells. We demonstrated that KMU-470 suppressed the production of nitric oxide and inducible nitric oxide synthases that are induced by LPS in RAW 264.7 cells. Furthermore, KMU-470 inhibited LPS-induced up-regulation of TLR4 and MyD88, as well as the phosphorylation of IκB kinase and NF-κB in RAW 264.7 cells. Additionally, KMU-470 suppressed LPS-induced up-regulation at the transcriptional level of various pro-inflammatory cytokines such as IL-1β, TNF-α, and IL-6. Notably, KMU-470 inhibited LPS-induced up-regulation of a major component of the inflammasome complex, NLRP3, in RAW 264.7 cells. Importantly, PIM-1 siRNA transfection attenuated up-regulation of NLRP3 and pro-IL-1β in LPS-treated RAW 264.7 cells. Taken together, these findings indicate that PIM-1 plays a key role in inflammatory signaling and that KMU-470 is a potential anti-inflammatory agent.
Collapse
Affiliation(s)
- Hye Suk Baek
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
| | - Hyeon Ji Min
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
| | | | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
| | - Jong Wook Park
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
| | - Jinho Lee
- Department of Chemistry, Keimyung University, Daegu 42601, Korea;
- Correspondence: (J.L.); (S.K.); Tel.: +82-53-580-5183 (J.L.); +82-53-258-7359 (S.K.); Fax: +82-050-4154-2213 (J.L.); +82-53-258-7355 (S.K.)
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Korea; (H.S.B.); (H.J.M.); (T.K.K.); (J.W.P.)
- Institute of Medical Science, Keimyung University, Daegu 42601, Korea
- Correspondence: (J.L.); (S.K.); Tel.: +82-53-580-5183 (J.L.); +82-53-258-7359 (S.K.); Fax: +82-050-4154-2213 (J.L.); +82-53-258-7355 (S.K.)
| |
Collapse
|
12
|
Wang J, Cao Y, Liu Y, Zhang X, Ji F, Li J, Zou Y. PIM1 inhibitor SMI-4a attenuated lipopolysaccharide-induced acute lung injury through suppressing macrophage inflammatory responses via modulating p65 phosphorylation. Int Immunopharmacol 2019; 73:568-574. [PMID: 31203114 DOI: 10.1016/j.intimp.2019.05.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 05/01/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022]
Abstract
PIM kinase is involved in the cellular processes of growth, differentiation and apoptosis. However, the role of PIM1 in lipopolysaccharide (LPS)-induced acute lung injury (ALI) remains largely unknown. A trend of PIM1 in the lung tissue of LPS-induced ALI at different time points was detected. Histology, wet/dry (W/D) ratio, inflammatory cells in the bronchoalveolar lavage fluid (BALF) and survival rate analyses were performed when mice received the PIM1 inhibitor SMI-4a intratracheally 3 h before LPS administration. Cytokine production in vivo and in vitro was measured after SMI-4a pretreatment. NF-κB subunit p65 expression in nuclei and phosphorylation at Ser276 in lung tissues or cells were detected by Western blot analysis. The results showed that PIM1 mRNA and protein were upregulated in the lung tissue of LPS-induced ALI. The PIM1 inhibitor SMI-4a markedly improved the survival rate after lethal LPS administration, reduced the severity of lung edema, attenuated the histologic injuries of the lung tissue and reduced the counts of infiltrated inflammatory cells in the BALF. The PIM1 inhibitor SMI-4a suppressed the production of cytokines in LPS-treated RAW264.7 cell supernatants and BALF. Furthermore, LPS administration upregulated the levels of nuclear p65 and phosphorylated p65 (p-p65) at Ser276, whereas pretreatment with the PIM1 inhibitor SMI-4a reduced p65 upregulation in the nucleus and p-p65 at Ser276. Taken together, these data indicate that the PIM1 inhibitor SMI-4a may serve as a promising therapeutic strategy for LPS-induced ALI by suppressing macrophage production of cytokines via a reduction of p65 activities.
Collapse
Affiliation(s)
- Jinxuan Wang
- Department of Anesthesiology, Weifang Medical University, Weifang, China; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yumeng Cao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqi Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyi Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fanceng Ji
- Department of Anesthesiology, Weifang People's Hospital, Weifang, China
| | - Jinbao Li
- Department of Anesthesiology, Weifang Medical University, Weifang, China; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Yun Zou
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
13
|
Barberis C, Pribish J, Tserlin E, Gross A, Czekaj M, Barragué M, Erdman P, Maniar S, Jiang J, Fire L, Patel V, Hebert A, Levit M, Wang A, Sun F, Huang SMA. Discovery of N-substituted 7-azaindoles as Pan-PIM kinases inhibitors - Lead optimization - Part III. Bioorg Med Chem Lett 2019; 29:491-495. [PMID: 30553737 DOI: 10.1016/j.bmcl.2018.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/04/2018] [Accepted: 12/08/2018] [Indexed: 12/17/2022]
Abstract
N-substituted azaindoles were discovered as promising pan-PIM inhibitors. Lead optimization is described en route toward the identification of a clinical candidate. Modulation of physico-chemical properties allowed to solve inherent hERG and permeability liabilities. Compound 17 showed tumor growth inhibition in a KG1 tumor-bearing mouse model.
Collapse
Affiliation(s)
- Claude Barberis
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States.
| | - James Pribish
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Elina Tserlin
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Alexandre Gross
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Mark Czekaj
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Matthieu Barragué
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Paul Erdman
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Sachin Maniar
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - John Jiang
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Luke Fire
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Vinod Patel
- IDD Medicinal Chemistry, Sanofi, 153 Second Avenue, Waltham MA 02451, United States
| | - Andrew Hebert
- Oncology Biochemistry, Sanofi, 270 Albany Street, Cambridge MA 02139, United States
| | - Mikhail Levit
- Oncology Biochemistry, Sanofi, 270 Albany Street, Cambridge MA 02139, United States
| | - Anlai Wang
- Oncology Biology, Sanofi, 270 Albany Street, Cambridge MA 02139, United States
| | - Frank Sun
- Oncology Pharmacology, Sanofi, 640 Memorial Drive, Cambridge MA 02139, United States
| | - Shih-Min A Huang
- Oncology Biology, Sanofi, 270 Albany Street, Cambridge MA 02139, United States
| |
Collapse
|
14
|
Trinh HKT, Suh DH, Nguyen TVT, Choi Y, Park HS, Shin YS. Characterization of cysteinyl leukotriene-related receptors and their interactions in a mouse model of asthma. Prostaglandins Leukot Essent Fatty Acids 2019; 141:17-23. [PMID: 30661601 DOI: 10.1016/j.plefa.2018.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 09/01/2018] [Accepted: 12/11/2018] [Indexed: 12/22/2022]
Abstract
Identification of the characterization of cysteinyl leukotrienes receptor (CysLTRs) could facilitate our understanding of these receptors' role in asthma. We aimed to investigate the localization and interactions of CysLTRs using a mouse model of asthma. BALB/c mice were administered ovalbumin (OVA) to induce allergic asthma. Some mice were administered the antagonists of CysLTR1, CysLTR2, and purinergic receptor P2Y12 (P2Y12R) (montelukast, HAMI 3379 and clopidogrel, respectively). The expression levels of CysLTR1, CysLTR2, and P2Y12R on lung tissues and inflammatory cells were evaluated by western blot, flow cytometry, and immunochemistry. CysLTR1 and P2Y12R were significantly up-regulated in lung tissues (P < 0.05 for each) from mouse after being sensitized and challenged with OVA (OVA/OVA). The ratio of CysLTR1: CysLTR2: P2Y12R in lungs of negative control (NC) mice was shifted from 1:0.43:0.35 to 1:0.65:1.34 in OVA/OVA mice. Montelukast significantly diminished the up-regulation of CysLTR1, CysLTR2, and P2Y12R (P < 0.05 for each), while the effects of HAMI 3379 and clopidogrel were predominant on the expression of CysLTR2 and P2Y12R, respectively. Montelukast predominantly diminished the cell count, while clopidogrel potently inhibited the release of interleukin (IL)-4, IL-5, and IL-13. Our study demonstrated the interactions between CysLTRs, thereby highlighting the potential synergistic effects of CysLTR antagonists in asthma treatment.
Collapse
Affiliation(s)
- Hoang Kim Tu Trinh
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Dong-Hyeon Suh
- Department of Pharmacology, CKD Research institute, Yong-in, South Korea
| | - Thuy Van Thao Nguyen
- Pediatric Department, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh city, Vietnam
| | - Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Science, Ajou University School of Medicine, Suwon, South Korea
| | - Yoo Seob Shin
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea.
| |
Collapse
|
15
|
Zhao M, Chen Y, Wang C, Xiao W, Chen S, Zhang S, Yang L, Li Y. Systems Pharmacology Dissection of Multi-Scale Mechanisms of Action of Huo-Xiang-Zheng-Qi Formula for the Treatment of Gastrointestinal Diseases. Front Pharmacol 2019; 9:1448. [PMID: 30687082 PMCID: PMC6336928 DOI: 10.3389/fphar.2018.01448] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/26/2018] [Indexed: 12/19/2022] Open
Abstract
Multi-components Traditional Chinese Medicine (TCM) treats various complex diseases (multi-etiologies and multi-symptoms) via herbs interactions to exert curative efficacy with less adverse effects. However, the ancient Chinese compatibility theory of herbs formula still remains ambiguous. Presently, this combination principle is dissected through a systems pharmacology study on the mechanism of action of a representative TCM formula, Huo-xiang-zheng-qi (HXZQ) prescription, on the treatment of functional dyspepsia (FD), a chronic or recurrent clinical disorder of digestive system, as typical gastrointestinal (GI) diseases which burden human physical and mental health heavily and widely. In approach, a systems pharmacology platform which incorporates the pharmacokinetic and pharmaco-dynamics evaluation, target fishing and network pharmacological analyses is employed. As a result, 132 chemicals and 48 proteins are identified as active compounds and FD-related targets, and the mechanism of HXZQ formula for the treatment of GI diseases is based on its three function modules of anti-inflammation, immune protection and gastrointestinal motility regulation mainly through four, i.e., PIK-AKT, JAK-STAT, Toll-like as well as Calcium signaling pathways. In addition, HXZQ formula conforms to the ancient compatibility rule of "Jun-Chen-Zuo-Shi" due to the different, while cooperative roles that herbs possess, specifically, the direct FD curative effects of GHX (serving as Jun drug), the anti-bacterial efficacy and major accompanying symptoms-reliving bioactivities of ZS and BZ (as Chen), the detoxication and ADME regulation capacities of GC (as Shi), as well as the minor symptoms-treating efficacy of the rest 7 herbs (as Zuo). This work not only provides an insight of the therapeutic mechanism of TCMs on treating GI diseases from a multi-scale perspective, but also may offer an efficient way for drug discovery and development from herbal medicine as complementary drugs.
Collapse
Affiliation(s)
- Miaoqing Zhao
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian, China.,Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Shihezi, China
| | - Yangyang Chen
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, China
| | - Chao Wang
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian, China
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Shusheng Chen
- Systems Biology Laboratory, Department of Computer & Information Science & Engineering, University of Florida, Gainesville, FL, United States
| | - Shuwei Zhang
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian, China
| | - Ling Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Li
- Key Laboratory of Industrial Ecology and Environmental Engineering, Faculty of Chemical, Environmental and Biological Science and Technology, Dalian University of Technology, Dalian, China.,Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Shihezi, China
| |
Collapse
|
16
|
Lim R, Barker G, Lappas M. Inhibition of PIM1 kinase attenuates inflammation-induced pro-labour mediators in human foetal membranes in vitro. Mol Hum Reprod 2018; 23:428-440. [PMID: 28333279 DOI: 10.1093/molehr/gax013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/06/2017] [Indexed: 01/29/2023] Open
Abstract
STUDY QUESTION Does proviral integration site for Moloney murine leukaemic virus (PIM)1 kinase play a role in regulating the inflammatory processes of human labour and delivery? SUMMARY ANSWER PIM1 kinase plays a critical role in foetal membranes in regulating pro-inflammatory and pro-labour mediators. WHAT IS KNOWN ALREADY Infection and inflammation have strong causal links to preterm delivery by stimulating pro-inflammatory cytokines and collagen degrading enzymes, which can lead to rupture of membranes. PIM1 has been shown to have a role in immune regulation and inflammation in non-gestational tissues; however, its role has not been explored in the field of human labour. STUDY DESIGN, SIZE, DURATION PIM1 expression was analysed in myometrium and/or foetal membranes obtained at term and preterm (n = 8-9 patients per group). Foetal membranes, freshly isolated amnion cells and primary myometrial cells were used to investigate the effect of PIM1 inhibition on pro-labour mediators (n = 5 patients per treatment group). PARTICIPANTS/MATERIALS, SETTING AND METHODS Foetal membranes, from term and preterm, were obtained from non-labouring and labouring women, and from preterm pre-labour rupture of membranes (PPROM) (n = 9 per group). Amnion was collected from women with and without preterm chorioamnionitis (n = 8 per group). Expression of PIM1 kinase was determined by qRT-PCR and western blotting. To determine the effect of PIM1 kinase inhibition on the expression of pro-inflammatory and pro-labour mediators induced by bacterial products lipopolysaccharide (LPS) (10 μg/ml) and flagellin (1 μg/ml) and pro-inflammatory cytokine tumour necrosis factor (TNF) (10 ng/ml), chemical inhibitors SMI-4a (20 μM) and AZD1208 (50 μM) were used in foetal membrane explants and siRNA against PIM1 was used in primary amnion cells. Statistical significance was set at P < 0.05. MAIN RESULTS AND THE ROLE OF CHANCE PIM1 expression was significantly increased in foetal membranes after spontaneous term labour compared to no labour at term and in amnion with preterm chorioamnionitis compared to preterm with no chorioamnionitis. There was no change in PIM1 expression with preterm labour or PPROM compared to preterm with no labour or PPROM. In human foetal membranes, PIM1 inhibitors SMI-4a and AZD1208 significantly decreased the expression of pro-inflammatory cytokine interleukin-6 (IL6) and chemokines CXCL8 and CCL2 mRNA and release, prostaglandin prostaglandin F2α (PGF2α) release, adhesion molecule intercellular adhesion molecule 1 mRNA expression and release, and oxidative stress marker 8-isoprostane release after stimulation with either LPS or flagellin. Primary amnion cells transfected with PIM1 siRNA also showed decreased expression of IL6, CXCL8 and CCL2, PTGS2 mRNA and PGF2α release, and matrix metalloproteinase-9 (MMP9) expression, when stimulated with TNF. LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION The conclusions were drawn from in vitro experiments using foetal membrane explants and primary cells isolated from amnion. Animal models are necessary to determine whether PIM1 kinase inhibitors can prevent spontaneous preterm birth in vivo. WIDER IMPLICATIONS OF THE FINDINGS PIM1 kinase inhibitors may provide a novel therapeutic approach for preventing spontaneous preterm birth. STUDY FUNDING/COMPETING INTEREST(S) Associate Professor Martha Lappas is supported by a Career Development Fellowship from the National Health and Medical Research Council (NHMRC; grant no. 1047025). Funding for this study was provided by the NHMRC (grant no. 1058786), Norman Beischer Medical Research Foundation and the Mercy Research Foundation. The authors have no conflict of interest.
Collapse
Affiliation(s)
- Ratana Lim
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Level 4/163 Studley Road, Heidelberg, Victoria 3084, Australia.,Mercy Perinatal Research Centre, Mercy Hospital for Women, Level 4/163 Studley Road, Heidelberg, Victoria 3084, Australia
| | - Gillian Barker
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Level 4/163 Studley Road, Heidelberg, Victoria 3084, Australia.,Mercy Perinatal Research Centre, Mercy Hospital for Women, Level 4/163 Studley Road, Heidelberg, Victoria 3084, Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Level 4/163 Studley Road, Heidelberg, Victoria 3084, Australia.,Mercy Perinatal Research Centre, Mercy Hospital for Women, Level 4/163 Studley Road, Heidelberg, Victoria 3084, Australia
| |
Collapse
|
17
|
Aweya JJ, Wang W, Zhang Y, Yao D, Li S, Wang F. Identification and molecular characterization of the Pim1 serine/threonine kinase homolog in Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2018; 74:491-500. [PMID: 29355758 DOI: 10.1016/j.fsi.2018.01.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/08/2018] [Accepted: 01/11/2018] [Indexed: 06/07/2023]
Abstract
The Pim1 serine/threonine kinase is associated with multiple cellular functions including proliferation, survival, differentiation, apoptosis, tumorigenesis, immune regulation and inflammation in vertebrates. However, little is known about the role of Pim1 in invertebrate immunity. In this study, we identified and characterized for the first time, a Pim1 (LvPim1) gene in Litopenaeus vannamei, with a full-length cDNA of 2352 bp and a 1119 bp open reading frame (ORF) encoding a putative protein of 372 amino acids, which contains a typical serine/threonine kinase domain. Sequence and phylogenetic analysis revealed that LvPim1 shared a close evolutionary relationship with Pim1 from vertebrates. Real-time qPCR analysis showed that LvPim1 was widely expressed in all tissues tested; with its transcript level induced in hepatopancreas and hemocytes upon challenge with Vibrio parahaemolyticus, Streptoccocus iniae, lipopolysaccharide (LPS), and white spot syndrome virus (WSSV), thus, suggesting its probable involvement in shrimp immune response. Moreover, knockdown of LvPim1 resulted in increased hemocytes apoptosis; shown by high caspase3/7 activity, coupled with increase in pro-apoptotic LvCaspase3 and LvCytochrome C, and decrease in pro-survival LvBcl2, LvIAP1, and LvIAP2 mRNA expression in hemocytes. Finally, LvPim1 knockdown renders shrimps more susceptible to V. parahaemolyticus infection. Taken together, our present data strongly suggest that LvPim1 is involved in modulating shrimp resistance to pathogen infection, promote hemocytes survival, and therefore plays a role in shrimp immune response.
Collapse
Affiliation(s)
- Jude Juventus Aweya
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Wei Wang
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Yueling Zhang
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Defu Yao
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Shengkang Li
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China
| | - Fan Wang
- Department of Biology and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China.
| |
Collapse
|
18
|
Du W, Chen T, Ni Y, Hou X, Yu Y, Zhou Q, Wu F, Tang W, Shi G. Role of PIM2 in allergic asthma. Mol Med Rep 2017; 16:7504-7512. [PMID: 28944837 PMCID: PMC5865883 DOI: 10.3892/mmr.2017.7499] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 07/21/2017] [Indexed: 01/11/2023] Open
Abstract
T cell-associated inflammation, particularly type 2 inflammation, has an important role in asthma pathogenesis, which is suppressed by regulatory T cells (Tregs). Proviral integration site for Moloney murine leukemia virus 2 (PIM2), a member off the serine/threonine kinase family, promotes the growth and survival of T cells and influences the function of Treg cells. However, whether PIM2 affects asthma pathogenesis remains unclear. Peripheral blood mononuclear cells and Treg cells from asthmatic and healthy subjects were obtained, and the expression level of PIM2 was measured by reverse transcription-quantitative polymerase chain reaction and immunocytochemistry. In addition, BALB/c female mice sensitized and challenged by ovalbumin were used as an asthma model, and PIM2 inhibitor was injected during the challenge period to observe the effect of PIM2 on asthma. The asthma symptoms were recorded, and airway hyper-responsiveness (AHR), expression levels of cytokines in the serum or bronchoalveolar lavage fluid (BALF), and the number of BALF leukocytes were evaluated. In addition, hematoxylin and eosin staining and immunohistochemistry of lung tissues was performed. The results demonstrated that PIM2 was overexpressed in patients with asthma in natural Treg cells. Inhibition of PIM2 attenuated asthma symptoms, and improved AHR and airway inflammation compared with asthmatic mice without inhibition of PIM2. In addition, expression levels of interleukin (IL)-10 and forkhead box protein 3 (FOXP3) in BALF were increased following PIM2 inhibition (IL-10, 470.3±21.78 vs. 533.7±25.55 pg/ml, P<0.05; FOXP3, 259±4.68 vs. 279.3±3.68 pg/ml; asthma and PIM2 inhibition groups, respectively; P<0.05). In conclusion, PIM2 may exhibit an important role in asthma pathogenesis and exacerbate AHR, airway inflammation and asthma symptoms. These effects of PIM2 may be dependent on Treg cells and the secretion of IL-10 by Tregs. The results of the present study suggest that PIM2 may be a potential target molecule for asthma treatment.
Collapse
Affiliation(s)
- Wei Du
- Department of Pulmonary Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Tiantian Chen
- Department of Pulmonary Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Yingmeng Ni
- Department of Pulmonary Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Xiaoxia Hou
- Department of Pulmonary Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Youchao Yu
- Department of Pulmonary Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Qi Zhou
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Orthopedics and Traumatology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Fang Wu
- Department of Geratology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Wei Tang
- Department of Pulmonary Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| | - Guochao Shi
- Department of Pulmonary Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, P.R. China
| |
Collapse
|
19
|
Barberis C, Moorcroft N, Pribish J, Tserlin E, Gross A, Czekaj M, Barrague M, Erdman P, Majid T, Batchelor J, Levit M, Hebert A, Shen L, Moreno-Mazza S, Wang A. Discovery of N-substituted 7-azaindoles as Pan-PIM kinase inhibitors - Lead series identification - Part II. Bioorg Med Chem Lett 2017; 27:4735-4740. [PMID: 28927793 DOI: 10.1016/j.bmcl.2017.08.068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/25/2017] [Accepted: 08/31/2017] [Indexed: 02/08/2023]
Abstract
N-Substituted azaindoles have been discovered as pan-PIM kinase inhibitors. Initial SAR, early ADME and PK/PD data of a series of compounds is described and led to the identification of promising pan-PIM inhibitors which validated our interest in the 7-azaindole scaffold and led us to pursue the identification of a clinical candidate.
Collapse
Affiliation(s)
- Claude Barberis
- IDD Medicinal Chemistry, Sanofi Genzyme, 153 Second Avenue, Waltham, MA 02451, USA.
| | - Neil Moorcroft
- IDD Medicinal Chemistry, Sanofi Genzyme, 153 Second Avenue, Waltham, MA 02451, USA
| | - James Pribish
- IDD Medicinal Chemistry, Sanofi Genzyme, 153 Second Avenue, Waltham, MA 02451, USA
| | - Elina Tserlin
- IDD Medicinal Chemistry, Sanofi Genzyme, 153 Second Avenue, Waltham, MA 02451, USA
| | - Alexandre Gross
- IDD Medicinal Chemistry, Sanofi Genzyme, 153 Second Avenue, Waltham, MA 02451, USA
| | - Mark Czekaj
- IDD Medicinal Chemistry, Sanofi Genzyme, 153 Second Avenue, Waltham, MA 02451, USA
| | - Matthieu Barrague
- IDD Medicinal Chemistry, Sanofi Genzyme, 153 Second Avenue, Waltham, MA 02451, USA
| | - Paul Erdman
- IDD Medicinal Chemistry, Sanofi Genzyme, 153 Second Avenue, Waltham, MA 02451, USA
| | - Tahir Majid
- IDD Medicinal Chemistry, Sanofi Genzyme, 153 Second Avenue, Waltham, MA 02451, USA
| | - Joseph Batchelor
- IDD In Vitro Biology, Sanofi, 153 Second Avenue, Waltham, MA 02451, USA
| | - Mikhail Levit
- Oncology Biology, Sanofi, 270 Albany Street, Cambridge, MA 02139, USA
| | - Andrew Hebert
- Oncology Biology, Sanofi, 270 Albany Street, Cambridge, MA 02139, USA
| | - Liduo Shen
- DSAR, Sanofi Genzyme, 211 Second Avenue, Waltham, MA 02451, USA
| | | | - Anlai Wang
- Oncology Biology, Sanofi, 270 Albany Street, Cambridge, MA 02139, USA
| |
Collapse
|
20
|
Gelfand EW, Joetham A, Wang M, Takeda K, Schedel M. Spectrum of T-lymphocyte activities regulating allergic lung inflammation. Immunol Rev 2017; 278:63-86. [PMID: 28658551 PMCID: PMC5501488 DOI: 10.1111/imr.12561] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite advances in the treatment of asthma, optimization of symptom control remains an unmet need in many patients. These patients, labeled severe asthma, are responsible for a substantial fraction of the disease burden. In these patients, research is needed to define the cellular and molecular pathways contributing to disease which in large part are refractory to corticosteroid treatment. The causes of steroid-resistant asthma are multifactorial and result from complex interactions of genetics, environmental factors, and innate and adaptive immunity. Adaptive immunity, addressed here, integrates the activities of distinct T-cell subsets and by definition is dynamic and responsive to an ever-changing environment and the influences of epigenetic modifications. These T-cell subsets exhibit different susceptibilities to the actions of corticosteroids and, in some, corticosteroids enhance their functional activation. Moreover, these subsets are not fixed in lineage differentiation but can undergo transcriptional reprogramming in a bidirectional manner between protective and pathogenic effector states. Together, these factors contribute to asthma heterogeneity between patients but also in the same patient at different stages of their disease. Only by carefully defining mechanistic pathways, delineating their sensitivity to corticosteroids, and determining the balance between regulatory and effector pathways will precision medicine become a reality with selective and effective application of targeted therapies.
Collapse
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Anthony Joetham
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Meiqin Wang
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Katsuyuki Takeda
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| |
Collapse
|
21
|
Placental Pim-1 expression is increased in obesity and regulates cytokine- and toll-like receptor-mediated inflammation. Placenta 2017; 53:101-112. [PMID: 28487013 DOI: 10.1016/j.placenta.2017.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/06/2017] [Accepted: 04/09/2017] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Obesity is a growing epidemic, and as a consequence the number of obese pregnancies has also increased. Pregnancy is characterised by maternal and placental inflammation which is intensified with maternal obesity. The proviral integration site for Moloney murine leukemic virus (Pim)-1 protein is a serine/threonine kinase involved in a wide range of inflammatory diseases. In relation to obesity, however, its role has not been elucidated in human placenta. The aims were to determine the placental expression of Pim-1 with pre-existing maternal obesity and its role in regulating placental inflammation associated with obesity. METHODS Human placenta was obtained at the time of term Caesarean section from lean and pre-existing obese pregnant women to determine the effect of maternal obesity on Pim-1 expression. To determine the effect of Pim-1 on the inflammatory response induced by bacterial endotoxin LPS and pro-inflammatory cytokines TNF-α or IL-1β, the chemical inhibitor SMI-4a and siRNA were used. RESULTS Pim-1 protein and mRNA expression was significantly increased in placenta of obese women. SMI-4a significantly suppressed the expression and release of pro-inflammatory cytokine IL-6 and chemokines GRO-α and MCP-1 when stimulated with LPS or TNF-α in placenta. Primary trophoblast cells transfected with Pim-1 siRNA had decreased expression and release of pro-inflammatory cytokines IL-1β, IL-6, chemokines GRO-α and MCP-1, when stimulated with LPS, TNF-α or IL-1β. DISCUSSION The findings from this study implicate Pim-1 may contribute to placental inflammation in pregnancies complicated by maternal obesity. Thus, therapeutic targets for Pim-1 may improve fetal outcomes complicated by obese pregnancies.
Collapse
|
22
|
Bataille CJR, Brennan MB, Byrne S, Davies SG, Durbin M, Fedorov O, Huber KVM, Jones AM, Knapp S, Liu G, Nadali A, Quevedo CE, Russell AJ, Walker RG, Westwood R, Wynne GM. Thiazolidine derivatives as potent and selective inhibitors of the PIM kinase family. Bioorg Med Chem 2017; 25:2657-2665. [PMID: 28341403 DOI: 10.1016/j.bmc.2017.02.056] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/23/2017] [Accepted: 02/25/2017] [Indexed: 12/31/2022]
Abstract
The PIM family of serine/threonine kinases have become an attractive target for anti-cancer drug development, particularly for certain hematological malignancies. Here, we describe the discovery of a series of inhibitors of the PIM kinase family using a high throughput screening strategy. Through a combination of molecular modeling and optimization studies, the intrinsic potencies and molecular properties of this series of compounds was significantly improved. An excellent pan-PIM isoform inhibition profile was observed across the series, while optimized examples show good selectivity over other kinases. Two PIM-expressing leukemic cancer cell lines, MV4-11 and K562, were employed to evaluate the in vitro anti-proliferative effects of selected inhibitors. Encouraging activities were observed for many examples, with the best example (44) giving an IC50 of 0.75μM against the K562 cell line. These data provide a promising starting point for further development of this series as a new cancer therapy through PIM kinase inhibition.
Collapse
Affiliation(s)
- Carole J R Bataille
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Méabh B Brennan
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Simon Byrne
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Stephen G Davies
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK.
| | - Matthew Durbin
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Oleg Fedorov
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, UK
| | - Kilian V M Huber
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Alan M Jones
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Stefan Knapp
- Structural Genomics Consortium, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, UK
| | - Gu Liu
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Anna Nadali
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Camilo E Quevedo
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Angela J Russell
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| | - Roderick G Walker
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Robert Westwood
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| | - Graham M Wynne
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, UK
| |
Collapse
|
23
|
de Vries M, Hesse L, Jonker MR, van den Berge M, van Oosterhout AJM, Heijink IH, Nawijn MC. Pim1 kinase activity preserves airway epithelial integrity upon house dust mite exposure. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1344-53. [PMID: 26453516 DOI: 10.1152/ajplung.00043.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 10/04/2015] [Indexed: 12/31/2022] Open
Abstract
Most patients with allergic asthma are sensitized to house dust mite (HDM). The allergenicity of HDM largely depends on disruption of the integrity and proinflammatory activation of the airway epithelium. In this study, we hypothesized that Pim1 kinase activity attenuates HDM-induced asthma by preserving airway epithelial integrity. The effects of Pim1 kinase activity on barrier function and release of the proinflammatory mediators IL-1α and CCL20 were studied in vitro in 16HBE and primary bronchial epithelial cells (PBECs). Pim1-proficient and -deficient mice were exposed to a HDM-driven model of allergic asthma, and airway hyperresponsiveness (AHR) was measured upon methacholine challenge. Airway inflammation and proinflammatory mediators in lung tissue and BAL fluid were determined. We observed that inhibition of Pim1 kinase prolongs the HDM-induced loss of barrier function in 16HBE cells and sensitizes PBECs to HDM-induced barrier dysfunction. Additionally, inhibition of Pim1 kinase increased the HDM-induced proinflammatory activity of 16HBE cells as measured by IL-1α secretion. In line herewith, HDM exposure induced an enhanced production of the proinflammatory chemokines CCL17 and CCL20 in Pim1-deficient mice compared with wild-type controls. While we observed a marked increase in eosinophilic and neutrophilic granulocytes as well as mucus cell metaplasia and AHR to methacholine in mice exposed to HDM, these parameters were independent of Pim1 kinase activity. In contrast, levels of the Th2-cytokines IL-5 and IL-10 were significantly augmented in HDM-treated Pim1-deficient mice. Taken together, our study shows that Pim1 kinase activity maintains airway epithelial integrity and protects against HDM-induced proinflammatory activation of the airway epithelium.
Collapse
Affiliation(s)
- M de Vries
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - L Hesse
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M R Jonker
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M van den Berge
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; and GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - A J M van Oosterhout
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - I H Heijink
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M C Nawijn
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
24
|
Vargas JE, Puga R, Poloni JDF, Saraiva Macedo Timmers LF, Porto BN, Norberto de Souza O, Bonatto D, Condessa Pitrez PM, Tetelbom Stein R. A network flow approach to predict protein targets and flavonoid backbones to treat respiratory syncytial virus infection. BIOMED RESEARCH INTERNATIONAL 2015; 2015:301635. [PMID: 25879022 PMCID: PMC4386546 DOI: 10.1155/2015/301635] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 09/11/2014] [Indexed: 01/19/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection is the major cause of respiratory disease in lower respiratory tract in infants and young children. Attempts to develop effective vaccines or pharmacological treatments to inhibit RSV infection without undesired effects on human health have been unsuccessful. However, RSV infection has been reported to be affected by flavonoids. The mechanisms underlying viral inhibition induced by these compounds are largely unknown, making the development of new drugs difficult. METHODS To understand the mechanisms induced by flavonoids to inhibit RSV infection, a systems pharmacology-based study was performed using microarray data from primary culture of human bronchial cells infected by RSV, together with compound-proteomic interaction data available for Homo sapiens. RESULTS After an initial evaluation of 26 flavonoids, 5 compounds (resveratrol, quercetin, myricetin, apigenin, and tricetin) were identified through topological analysis of a major chemical-protein (CP) and protein-protein interacting (PPI) network. In a nonclustered form, these flavonoids regulate directly the activity of two protein bottlenecks involved in inflammation and apoptosis. CONCLUSIONS Our findings may potentially help uncovering mechanisms of action of early RSV infection and provide chemical backbones and their protein targets in the difficult quest to develop new effective drugs.
Collapse
Affiliation(s)
- José Eduardo Vargas
- Centro Infant, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenue Ipiranga 6681, 90619-900 Porto Alegre, RS, Brazil
| | - Renato Puga
- Clinical Research Center, Hospital Israelita Albert Einstein (HIAE), São Paulo, Brazil
| | - Joice de Faria Poloni
- Department of Molecular Biology and Biotechnology, Federal University of Rio Grande do Sul (UFRGS), 90619-900 Porto Alegre, RS, Brazil
| | - Luis Fernando Saraiva Macedo Timmers
- Faculty of Informatics, Laboratory for Bioinformatics, Modelling & Simulation of Biosystems, Pontifical Catholic University of Rio Grande do Sul (PUCRS), 90619-900 Porto Alegre, RS, Brazil
| | - Barbara Nery Porto
- Centro Infant, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenue Ipiranga 6681, 90619-900 Porto Alegre, RS, Brazil
| | - Osmar Norberto de Souza
- Faculty of Informatics, Laboratory for Bioinformatics, Modelling & Simulation of Biosystems, Pontifical Catholic University of Rio Grande do Sul (PUCRS), 90619-900 Porto Alegre, RS, Brazil
| | - Diego Bonatto
- Department of Molecular Biology and Biotechnology, Federal University of Rio Grande do Sul (UFRGS), 90619-900 Porto Alegre, RS, Brazil
| | - Paulo Márcio Condessa Pitrez
- Centro Infant, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenue Ipiranga 6681, 90619-900 Porto Alegre, RS, Brazil
| | - Renato Tetelbom Stein
- Centro Infant, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenue Ipiranga 6681, 90619-900 Porto Alegre, RS, Brazil
| |
Collapse
|
25
|
Chen Y, Zhou X, Rong L. Analysis of mechanical ventilation and lipopolysaccharide‑induced acute lung injury using DNA microarray analysis. Mol Med Rep 2015; 11:4239-45. [PMID: 25672411 PMCID: PMC4394949 DOI: 10.3892/mmr.2015.3335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 01/15/2015] [Indexed: 01/25/2023] Open
Abstract
Gene expression profiles of samples taken from patients with acute lung injury (ALI) induced by mechanical ventilation (MV) and lipopolysaccharide (LPS) were analyzed in order to identify key genes, and explore the underlying mechanisms. The GSE2411 microarray data set was downloaded from the Gene Expression Omnibus. This data set contained microarray data from 24 mouse lung samples, which were equally divided into four groups: Control group, MV group, LPS group and MV+LPS group. Differentially expressed genes (DEGs) were identified in the MV, LPS and MV+LPS groups, as compared with the control group, using packages of R software. Hierarchical clustering and between‑group comparisons were performed for each group of DEGs. Overrepresented biological processes were revealed by functional enrichment analysis using the Database for Annotation, Visualization and Integrated Discovery. Unique DEGs in the LPS and MV+LPS groups were selected, and pathway enrichment analyses were performed using the Kyoto Encyclopedia of Genes and Genomes Orthology Based Annotation system. A total of 32, 264 and 685 DEGs were identified in the MV, LPS and MV+LPS groups, respectively. The MV+LPS group had more DEGs, as compared with the other two treatment groups. Genes associated with the immune and inflammatory responses were significantly overrepresented in both the LPS and MV+LPS groups, suggesting that LPS dominated the progression of ALI. Unique DEGs in the LPS and MV+LPS groups were associated with cytokine‑cytokine receptor interaction. The Janus kinase‑signal transducer and activator of transcription signaling pathway was shown to be enriched in the LPS+MV‑unique DEGs. The results of the present study demonstrated that MV could exaggerate the transcriptional response of the lungs to LPS. Numerous key genes were identified, which may advance knowledge regarding the pathogenesis of ALI.
Collapse
Affiliation(s)
- Yuqing Chen
- Department of Respiratory Medicine, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Xin Zhou
- Department of Respiratory Medicine, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, P.R. China
| | - Ling Rong
- Department of Respiratory Medicine, The People's Hospital of Bozhou, Bozhou, Anhui 236804, P.R. China
| |
Collapse
|
26
|
Liu JN, Suh DH, Yang EM, Lee SI, Park HS, Shin YS. Attenuation of airway inflammation by simvastatin and the implications for asthma treatment: is the jury still out? Exp Mol Med 2014; 46:e113. [PMID: 25213768 PMCID: PMC4183942 DOI: 10.1038/emm.2014.55] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 06/27/2014] [Accepted: 07/07/2014] [Indexed: 11/20/2022] Open
Abstract
Although some studies have explained the immunomodulatory effects of statins, the exact mechanisms and the therapeutic significance of these molecules remain to be elucidated. This study not only evaluated the therapeutic potential and inhibitory mechanism of simvastatin in an ovalbumin (OVA)-specific asthma model in mice but also sought to clarify the future directions indicated by previous studies through a thorough review of the literature. BALB/c mice were sensitized to OVA and then administered three OVA challenges. On each challenge day, 40 mg kg−1 simvastatin was injected before the challenge. The airway responsiveness, inflammatory cell composition, and cytokine levels in bronchoalveolar lavage (BAL) fluid were assessed after the final challenge, and the T cell composition and adhesion molecule expression in lung homogenates were determined. The administration of simvastatin decreased the airway responsiveness, the number of airway inflammatory cells, and the interleukin (IL)-4, IL-5 and IL-13 concentrations in BAL fluid compared with vehicle-treated mice (P<0.05). Histologically, the number of inflammatory cells and mucus-containing goblet cells in lung tissues also decreased in the simvastatin-treated mice. Flow cytometry showed that simvastatin treatment significantly reduced the percentage of pulmonary CD4+ cells and the CD4+/CD8+ T-cell ratio (P<0.05). Simvastatin treatment also decreased the expression of the vascular cell adhesion molecule 1 and intercellular adhesion molecule 1 proteins, as measured in homogenized lung tissues (P<0.05) and human epithelial cells. The reduction in the T cell influx as a result of the decreased expression of cell adhesion molecules is one of the mechanisms by which simvastatin attenuates airway responsiveness and allergic inflammation. Rigorous review of the literature together with our findings suggested that simvastatin should be further developed as a potential therapeutic strategy for allergic asthma.
Collapse
Affiliation(s)
- Jing-Nan Liu
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Dong-Hyeon Suh
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Eun-Mi Yang
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Seung-Ihm Lee
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Yoo Seob Shin
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
27
|
A network-based systematic study for the mechanism of the treatment of zhengs related to cough variant asthma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:595924. [PMID: 24348708 PMCID: PMC3855943 DOI: 10.1155/2013/595924] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/10/2013] [Accepted: 09/12/2013] [Indexed: 02/06/2023]
Abstract
Traditional Chinese medicine (TCM) has shown significant efficacy in the treatment of cough variant asthma (CVA), a special type of asthma. However, there is shortage of explanations for relevant mechanism of treatment. As Zhengs differentiation is a critical concept in TCM, it is necessary to explain the mechanism of treatment of Zhengs. Based on TCM clinical cases, this study illustrated the mechanism of the treatment of three remarkably relevant Zhengs for CVA: “FengXieFanFei,” “FeiQiShiXuan”, and “QiDaoLuanJi.” To achieve this goal, five steps were carried out: (1) determining feature Zhengs and corresponding key herbs of CVA by analyses of clinical cases; (2) finding out potential targets of the key herbs and clustering them based on their functional annotations; (3) constructing an ingredient-herb network and an ingredient network; (4) identifying modules of the ingredient network; (5) illustrating the mechanism of the treatment by further mining the latent biological implications within each module. The systematic study reveals that the treatment of “FengXieFanFei,” “FeiQiShiXuan,” and “QiDaoLuanJi” has effects on the regulation of multiple bioprocesses by herbs containing different ingredients with functions of steroid metabolism regulation, airway inflammation, and ion conduction and transportation. This network-based systematic study will be a good way to boost the scientific understanding of mechanism of the treatment of Zhengs.
Collapse
|
28
|
Wang M, Gelfand EW. Targeting Pim1 kinase in the treatment of peanut allergy. Expert Opin Ther Targets 2013; 18:177-83. [DOI: 10.1517/14728222.2014.855721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
29
|
Systematic identification of novel SLE related autoantibodies responsible for type I IFN production in human plasmacytoid dendritic cells. Cell Immunol 2013; 284:119-28. [DOI: 10.1016/j.cellimm.2013.07.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 07/11/2013] [Accepted: 07/29/2013] [Indexed: 12/25/2022]
|
30
|
Drygin D, Haddach M, Pierre F, Ryckman DM. Potential Use of Selective and Nonselective Pim Kinase Inhibitors for Cancer Therapy. J Med Chem 2012; 55:8199-208. [DOI: 10.1021/jm3009234] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Denis Drygin
- Cylene Pharmaceuticals, 5820 Nancy Ridge Drive, Suite 200, San Diego, California 92121,
United States
| | - Mustapha Haddach
- HTK Corporation, 5218 Rivergrade Road, Irwindale, California
91706, United States
| | - Fabrice Pierre
- 3244
Caminito Eastbluff, Apt 40, La Jolla, California 92037, United States
| | - David M. Ryckman
- Cylene Pharmaceuticals, 5820 Nancy Ridge Drive, Suite 200, San Diego, California 92121,
United States
| |
Collapse
|
31
|
Inhibition of Pim1 kinase prevents peanut allergy by enhancing Runx3 expression and suppressing T(H)2 and T(H)17 T-cell differentiation. J Allergy Clin Immunol 2012; 130:932-44.e12. [PMID: 22944483 DOI: 10.1016/j.jaci.2012.07.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 06/28/2012] [Accepted: 07/03/2012] [Indexed: 12/31/2022]
Abstract
BACKGROUND The provirus integration site for Moloney murine leukemia virus (Pim) 1 kinase is an oncogenic serine/threonine kinase implicated in cytokine-induced cell signaling, whereas Runt-related transcription factor (Runx) has been implicated in the regulation of T-cell differentiation. The interaction of Pim1 kinase and Runx3 in the pathogenesis of peanut allergy has not been defined. OBJECTIVES We sought to determine the effects of Pim1 kinase modulation on Runx3 expression and T(H)2 and T(H)17 cell function in an experimental model of peanut allergy. METHODS A Pim1 kinase inhibitor was administered to peanut-sensitized and challenged wild-type and Runx3(+/-) mice. Symptoms, intestinal inflammation, and Pim1 kinase and Runx3 mRNA expression and protein levels were assessed. The effects of Pim1 kinase inhibition on T(H)1, T(H)2, and T(H)17 differentiation in vivo and in vitro were also determined. RESULTS Peanut sensitization and challenge resulted in accumulation of inflammatory cells and goblet cell metaplasia and increased levels of Pim1 kinase and T(H)2 and T(H)17 cytokine production but decreased levels of Runx3 mRNA and protein in the small intestines of wild-type mice. All of these findings were normalized with Pim1 kinase inhibition. In sensitized and challenged Runx3(+/-) mice, inhibition of Pim1 kinase had less effect on the development of the full spectrum of intestinal allergic responses. In vitro inhibition of Pim1 kinase attenuated T(H)2 and T(H)17 cell differentiation and expansion while maintaining Runx3 expression in T-cell cultures from wild-type mice; these effects were reduced in T-cell cultures from Runx3(+/-) mice. CONCLUSION These data support a novel regulatory axis involving Pim1 kinase and Runx3 in the control of food-induced allergic reactions through the regulation of T(H)2 and T(H)17 differentiation.
Collapse
|