1
|
Christopoulou ME, Aletras AJ, Papakonstantinou E, Stolz D, Skandalis SS. WISP1 and Macrophage Migration Inhibitory Factor in Respiratory Inflammation: Novel Insights and Therapeutic Potentials for Asthma and COPD. Int J Mol Sci 2024; 25:10049. [PMID: 39337534 PMCID: PMC11432718 DOI: 10.3390/ijms251810049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Recent advancements highlight the intricate interplay between the extracellular matrix (ECM) and immune responses, notably in respiratory diseases such as asthma and Chronic Obstructive Pulmonary Disease (COPD). The ECM, a dynamic structural framework within tissues, orches-trates a plethora of cellular processes, including immune cell behavior and tissue repair mecha-nisms. WNT1-inducible-signaling pathway protein 1 (WISP1), a key ECM regulator, controls immune cell behavior, cytokine production, and tissue repair by modulating integrins, PI3K, Akt, β-catenin, and mTOR signaling pathways. WISP1 also induces macrophage migration inhibitory factor (MIF) expression via Src kinases and epidermal growth factor receptor (EGFR) activation. MIF, through its wide range of activities, enhances inflammation and tissue restructuring. Rec-ognized for its versatile roles in regulating the immune system, MIF interacts with multiple immune components, such as the NLRP3 inflammasome, thereby sustaining inflammatory pro-cesses. The WISP1-MIF axis potentially unveils complex molecular mechanisms governing im-mune responses and inflammation. Understanding the intricate roles of WISP1 and MIF in the pathogenesis of chronic respiratory diseases such as asthma and COPD could lead to the identi-fication of novel targets for therapeutic intervention to alleviate disease severity and enhance patient outcomes.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexios J Aletras
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Eleni Papakonstantinou
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daiana Stolz
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Spyros S Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
2
|
Christopoulou ME, Skandalis SS, Papakonstantinou E, Stolz D, Aletras AJ. WISP1 induces the expression of macrophage migration inhibitory factor in human lung fibroblasts through Src kinases and EGFR-activated signaling pathways. Am J Physiol Cell Physiol 2024; 326:C850-C865. [PMID: 38145300 PMCID: PMC11193488 DOI: 10.1152/ajpcell.00410.2023] [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: 08/27/2023] [Revised: 12/18/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Wnt1-inducible signaling protein 1 (WISP1/CCN4) is a secreted matricellular protein that is implicated in lung and airway remodeling. The macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that has been associated with chronic lung diseases. In this study, we aimed to investigate the WISP1 signaling pathway and its ability to induce the expression of MIF in primary cultures of fibroblasts from normal human lungs (HLFs). Our results showed that WISP1 significantly stimulated the expression of MIF in a concentration- and time-dependent fashion. In WISP1-induced expression of MIF, αvβ5-integrin and chondroitin sulfate proteoglycans as well as Src tyrosine kinases, MAP kinases, phosphatidylinositol 3-kinase/Akt, PKC, and NF-κB were involved. WISP1-induced expression of MIF was attenuated in the presence of the Src kinase inhibitor PP2 or the MIF tautomerase activity inhibitor ISO-1. Moreover, WISP1 significantly increased the phosphorylation and activation of EGF receptor (EGFR) through transactivation by Src kinases. WISP1 also induced the expression of MIF receptor CD74 and coreceptor CD44, through which MIF exerts its effects on HLFs. In addition, it was found that MIF induced its own expression, as well as its receptors CD74/CD44, acting in an autocrine manner. Finally, WISP1-induced MIF promoted the expression of cyclooxygenase 2, prostaglandin E2, IL-6, and matrix metalloproteinase-2 demonstrating the regulatory role of WISP1-MIF axis in lung inflammation and remodeling involving mainly integrin αvβ5, Src kinases, PKC, NF-κB, and EGFR. The specific signaling pathways involved in WISP1-induced expression of MIF may prove to be excellent candidates for novel targets to control inflammation in chronic lung diseases.NEW & NOTEWORTHY The present study demonstrates for the first time that Wnt1-inducible signaling protein 1 (WISP1) regulates migration inhibitory factor (MIF) expression and activity and identifies the main signaling pathways involved. The newly discovered WISP1-MIF axis may drive lung inflammation and could result in the design of novel targeted therapies in inflammatory lung diseases.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Spyros S Skandalis
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Eleni Papakonstantinou
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daiana Stolz
- Clinic of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexios J Aletras
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
3
|
Chang KS, Chen ST, Sung HC, Hsu SY, Lin WY, Hou CP, Lin YH, Feng TH, Tsui KH, Juang HH. WNT1 Inducible Signaling Pathway Protein 1 Is a Stroma-Specific Secreting Protein Inducing a Fibroblast Contraction and Carcinoma Cell Growth in the Human Prostate. Int J Mol Sci 2022; 23:ijms231911437. [PMID: 36232736 PMCID: PMC9570503 DOI: 10.3390/ijms231911437] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
The WNT1 inducible signaling pathway protein 1 (WISP1), a member of the connective tissue growth factor family, plays a crucial role in several important cellular functions in a highly tissue-specific manner. Results of a RT-qPCR indicated that WISP1 expressed only in cells of the human prostate fibroblasts, HPrF and WPMY-1, but not the prostate carcinoma cells in vitro. Two major isoforms (WISP1v1 and WISP1v2) were identified in the HPrF cells determined by RT-PCR and immunoblot assays. The knock-down of a WISP1 blocked cell proliferation and contraction, while treating respectively with the conditioned medium from the ectopic WISP1v1- and WISPv2-overexpressed 293T cells enhanced the migration of HPrF cells. The TNFα induced WISP1 secretion and cell contraction while the knock-down of WISP1 attenuated these effects, although TNFα did not affect the proliferation of the HPrF cells. The ectopic overexpression of WISP1v1 but not WISP1v2 downregulated the N-myc downstream regulated 1 (NDRG1) while upregulating N-cadherin, slug, snail, and vimentin gene expressions which induced not only the cell proliferation and invasion in vitro but also tumor growth of prostate carcinoma cells in vivo. The results confirmed that WISP1 is a stroma-specific secreting protein, enhancing the cell migration and contraction of prostate fibroblasts, as well as the proliferation, invasion, and tumor growth of prostate carcinoma cells.
Collapse
Affiliation(s)
- Kang-Shuo Chang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Syue-Ting Chen
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Shu-Yuan Hsu
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Wei-Yin Lin
- Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Chen-Pang Hou
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Shuang Ho Hospital, New Taipei City 235041, Taiwan
- TMU Research Center of Urology and Kidney, Department of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (K.-H.T.); (H.-H.J.); Tel.: +886-3-2118800 (ext. 5071) (H.-H.J.); Fax: +886-3-2118112 (H.-H.J.)
| | - Horng-Heng Juang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan
- Correspondence: (K.-H.T.); (H.-H.J.); Tel.: +886-3-2118800 (ext. 5071) (H.-H.J.); Fax: +886-3-2118112 (H.-H.J.)
| |
Collapse
|
4
|
Zhang QN, Xiao H, Fang LT, Sun QX, Li LD, Xu SY, Li CQ. Aerosol inhalation of Mycobacterium vaccae ameliorates airway structural remodeling in chronic asthma mouse model. Exp Lung Res 2022; 48:239-250. [PMID: 36001552 DOI: 10.1080/01902148.2022.2115166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background: Airway remodeling is accepted to be a determining component within the natural history of asthma. Nebulized inhalation of Mycobacterium vaccae (M. vaccae) has a protective effect on asthmatic mice. However, little is known regarding the effect of M. vaccae on airway structural remodeling in asthmatic mice. The purpose of this study was to explore the effect and the underlying mechanism of M. vaccae aerosol inhalation on airway structural remodeling in an asthma mouse model. Methods: Chronic asthma mouse models were established by ovalbumin induction. The number of inflammatory cells in bronchoalveolar lavage fluid (BALF), pathological alterations in lung tissue, and levels of associated cytokines (IL-5, IL-13, TNF-α, and ovalbumin-specific immunoglobulin E [OVA-sIgE]) were all assessed after M. vaccae therapy. The relative expression of interleukin (IL)-1β, tumor necrosis factor-alpha (TNF-α), nuclear factor kappa B (NF-κB), and Wnt1-induced signaling protein 1 (WISP1) mRNA were detected. Western blotting and immunohistochemistry detected the expression of Wnt/β-catenin pathway-related proteins in lung tissue. Results: M. vaccae aerosol inhalation relieved airway inflammation, airway hyper-responsiveness, and airway remodeling. M. vaccae reduced the levels of IL-5, IL-13, TNF-α, and OVA-sIgE in and downregulated the expression of IL-1β, TNF-α, NF-κB, and WISP1 mRNA in the pulmonary. In addition, M. vaccae inhibited the expression of β-catenin, WISP1, and Wnt1 protein and upregulated the expression of glycogen synthase kinase-3beta (GSK-3β). Conclusion: Nebulized inhalation of M. vaccae can reduce airway remodeling during asthma.
Collapse
Affiliation(s)
- Qian-Nan Zhang
- Departments of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huan Xiao
- Departments of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li-Ting Fang
- Departments of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qi-Xiang Sun
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lao-Dong Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Si-Yue Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chao-Qian Li
- Departments of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
5
|
Li L, Lv S, Li X, Liu J. Wnt-induced secreted proteins-1 play an important role in paraquat-induced pulmonary fibrosis. BMC Pharmacol Toxicol 2022; 23:21. [PMID: 35387687 PMCID: PMC8988378 DOI: 10.1186/s40360-022-00560-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 03/08/2022] [Indexed: 11/10/2022] Open
Abstract
Background The objective of this article is to observe the expression of Wnt-induced secreted proteins-1 (WISP1) in paraquat (PQ)-induced pulmonary fibrosis (PF) to explore the role of WISP1. Methods Healthy individuals were included in the control group. Patients who had acute lung injury or PF were included in the PF group. Venous blood samples were collected from the patients on days 1 and 3 following PQ poisoning to detect the expression levels of the WISP1 gene and protein concentration. Any changes in the patients’ blood gas analysis index were reviewed. In addition, chest computed tomography (CT) and x-ray images were observed to evaluate the relationship between WISP1 expression and disease severity. Results The expression of the WISP1 gene and the serum WISP1 protein concentration were higher in patients with PQ poisoning combined with PF than in patients without PF (P < 0.01). Serum PQ concentration was positively correlated with WISP1 gene expression (r = 0.621, P < 0.01), and serum WISP1 protein concentration (r = 0.596, P < 0.01) was considered a risk factor [odds ratio (OR) = 4.356, P < 0.05] for PQ-induced PF. Concurrently, the results of the adjusted and non-adjusted OR value for WISP1 gene expression and WISP1 protein concentration on day 1 was, respectively, as follows: OR = 12.797, 95% confidence interval (CI) (2.478–66.076), P = 0.002, OR’ = 11.353, P = 0.005; and OR = 1.545, 95% CI (1.197–1.995), P = 0.001, OR’ = 1.487, P = 0.003. The CT scan of a 20-year-old male with PQ-induced PF (20 ml) was observed, and it showed a typical hyaline-like lesion in the lungs on day 22 after poisoning; on day 33 after poisoning, the lungs showed localised consolidation combined with air bronchography. Conclusion The expression of WISP1 was higher in the patients with PQ-induced PF compared with the patients without PF. Accordingly, WISP1 plays an important role in PQ-induced PF.
Collapse
Affiliation(s)
- Lanrong Li
- Emergency Department, Linyi People's Hospital, Linyi, China
| | - Shengnan Lv
- Outpatient Department, Linyi People's Hospital, Linyi, China
| | - Xin Li
- Outpatient Department, Linyi People's Hospital, Linyi, China
| | - Jingyan Liu
- Emergency Department, Longgang District People's Hospital of Shenzhen, No. 53 of Aixin Road, Longgang District, Shenzhen, 518115, Guangdong Province, China.
| |
Collapse
|
6
|
Chetty A, Nielsen HC. Targeting Airway Smooth Muscle Hypertrophy in Asthma: An Approach Whose Time Has Come. J Asthma Allergy 2021; 14:539-556. [PMID: 34079293 PMCID: PMC8164696 DOI: 10.2147/jaa.s280247] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 04/20/2021] [Indexed: 01/13/2023] Open
Abstract
Airway smooth muscle (ASM) cell dysfunction is an important component of several obstructive pulmonary diseases, particularly asthma. External stimuli such as allergens, dust, air pollutants, and change in environmental temperatures provoke ASM cell hypertrophy, proliferation, and migration without adequate mechanistic controls. ASM cells can switch between quiescent, migratory, and proliferative phenotypes in response to extracellular matrix proteins, growth factors, and other soluble mediators. While some aspects of airway hypertrophy and remodeling could have beneficial effects, in many cases these contribute to a clinical phenotype of difficult to control asthma. In this review, we discuss the factors responsible for ASM hypertrophy and proliferation in asthma, focusing on cytokines, growth factors, and ion transporters, and discuss existing and potential approaches that specifically target ASM hypertrophy to reduce the ASM mass and improve asthma symptoms. The goal of this review is to highlight strategies that appear ready for translational investigations to improve asthma therapy.
Collapse
Affiliation(s)
- Anne Chetty
- Tufts Medical Center, Tufts University, Boston, MA, USA
| | | |
Collapse
|
7
|
Cheng C, Tian J, Zhang F, Deng Z, Tu M, Li L, Yang H, Xiao K, Guo W, Yang R, Gao S, Zhou Z. WISP1 Protects Against Chondrocyte Senescence and Apoptosis by Regulating αvβ3 and PI3K/Akt Pathway in Osteoarthritis. DNA Cell Biol 2021; 40:629-637. [PMID: 33646053 DOI: 10.1089/dna.2020.5926] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Our study aimed at validating the effect of WISP1 on osteoarthritis (OA) and the pathway involved in the WISP1-induced protection against OA. The expression of WISP1 was measured by immunohistochemical analyses. We found that WISP1 expression was shown to be upregulated within human OA cartilage compared with controls. WISP1 expression was related to knee OA severity. rhWISP1 inhibited OA chondrocyte senescence and apoptosis in vitro, which was reversed by the αvβ3 antibody and PI3K/Akt inhibitor LY294002. WISP1 overexpression induced by knee injection of LiCI could also prevent the senescence and apoptosis of rat chondrocytes. Safranin-O staining and Mankin score revealed that WISP1 overexpression can protect rat chondrocytes from degeneration. Nearly opposite results were obtained in the treatment of ICG-001 and siRNA-WISP1 in vivo. These data strongly suggest that WISP1 can protect against the senescence and apoptosis of chondrocytes via modulating the αvβ3 receptor and PI3K/Akt signaling pathway within OA. Therefore, the development of specific activators of WISP1 may present the value of an underlying OA treatment.
Collapse
Affiliation(s)
- Chao Cheng
- Department of Orthopaedics, Yiyang Central Hospital, Yiyang, China.,Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, China
| | - Jian Tian
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Fangjie Zhang
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenhan Deng
- Department of Sports Medicine, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Min Tu
- Department of Orthopedics, Second People's Hospital of Jingmen, Jingmen, China
| | - Liangjun Li
- Department of Orthopedic, Changsha Central Hospital, Changsha, China
| | - Hua Yang
- Department of Orthopaedics, Yiyang Central Hospital, Yiyang, China.,Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, China
| | - Kai Xiao
- Department of Orthopaedics, Yiyang Central Hospital, Yiyang, China.,Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, China
| | - Wei Guo
- Department of Orthopaedics, Yiyang Central Hospital, Yiyang, China.,Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, China
| | - Ruiqi Yang
- Department of Orthopaedics, Yiyang Central Hospital, Yiyang, China.,Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, China
| | - Shuguang Gao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhihong Zhou
- Department of Orthopaedics, Yiyang Central Hospital, Yiyang, China.,Clinical Medical Technology Demonstration Base for Minimally Invasive and Digital Orthopaedics in Hunan Province, Yiyang, China
| |
Collapse
|
8
|
Bai J, Zhong JY, Liao W, Hu R, Chen L, Wu XJ, Liu SP. iTRAQ‑based proteomic analysis reveals potential regulatory networks in dust mite‑related asthma treated with subcutaneous allergen immunotherapy. Mol Med Rep 2020; 22:3607-3620. [PMID: 32901873 PMCID: PMC7533450 DOI: 10.3892/mmr.2020.11472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 02/24/2020] [Indexed: 12/29/2022] Open
Abstract
Asthma is one of the most common childhood chronic diseases worldwide. Subcutaneous immunotherapy (SCIT) is commonly used in the treatment of house dust mite (HDM)‑related asthma in children. However, the therapeutic mechanism of SCIT in asthma remains unclear. The present study aimed to investigate the molecular biomarkers associated with HDM‑related asthma in asthmatic children prior and subsequent to SCIT treatment compared with those in healthy children via proteomic analysis. The study included a control group (30 healthy children), ‑Treatment group (30 children with HDM‑related allergic asthma) and +Treatment group (30 children with HDM‑related allergic asthma treated with SCIT). An isobaric labeling with relative and absolute quantification‑based method was used to analyze serum proteome changes to detect differentially expressed proteins, while functional enrichment and protein‑protein interaction network analysis were used to select candidate biomarkers. A total of 72 differentially expressed proteins were detected in the ‑Treatment, +Treatment and control groups. A total of 33 and 57 differentially expressed proteins were observed in the ‑Treatment vs. control and +Treatment vs. control groups, respectively. Through bioinformatics analysis, 5 candidate proteins [keratin 1 (KRT1), apolipoprotein B (APOB), fibronectin 1, antithrombin III (SERPINC1) and α‑1‑antitrypsin (SERPINA1)] were selected for validation by western blotting; among them, 4 proteins (KRT1, APOB, SERPINC1 and SERPINA1) showed robust reproducibility in asthma and control samples. This study illustrated the changes in proteome regulation following SCIT treatment for asthma. The 4 identified proteins may serve as potential biomarkers prior and subsequent to SCIT treatment, and help elucidate the molecular regulation mechanisms of SCIT to treat HDM‑related asthma.
Collapse
Affiliation(s)
- Jun Bai
- Department of Pediatrics, Foshan Maternal and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
| | - Jia-Yong Zhong
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Wang Liao
- Department of Pediatrics, Foshan Maternal and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
| | - Ruo Hu
- School of Computer Science, Guangdong Polytechnic Normal University, Guangzhou, Guangdong 510000, P.R. China
| | - Liang Chen
- Department of Pediatrics, Foshan Maternal and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
| | - Xian-Jin Wu
- Key Laboratory of Research and Utilization of Ethnomedicinal Plant Resources of Hunan Province, Key Laboratory of Hunan Higher Education for Western Hunan Medicinal Plant and Ethnobotany, College of Biological and Food Engineering, Huaihua University, Huaihua, Hunan 418008, P.R. China
| | - Shuang-Ping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| |
Collapse
|
9
|
Guguen C, Launois C, Dormoy V, Dewolf M, Dumazet A, Dury S, Lebargy F, Deslee G, Perotin JM. [Obesity and asthma: Mechanisms and therapeutic options]. Rev Mal Respir 2019; 37:134-143. [PMID: 31864881 DOI: 10.1016/j.rmr.2019.03.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/06/2019] [Indexed: 12/22/2022]
Abstract
Asthma and obesity are both common conditions, which lead to a substantial public health burden. The obese-asthma phenotype is characterized by poor asthma control, impaired lung function and decreased efficacy of inhaled treatment. However, this phenotype is highly heterogeneous and involves numerous mechanisms, including systemic inflammation and adipokines. A role for microbiota modifications and genetics has been suggested. Obese-asthma patient management currently consists in weight loss and usual anti-asthmatic treatment. New therapeutic options are being evaluated.
Collapse
Affiliation(s)
- C Guguen
- Service des maladies respiratoires, Inserm UMR-S 1250, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - C Launois
- Service des maladies respiratoires, Inserm UMR-S 1250, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - V Dormoy
- Service des maladies respiratoires, Inserm UMR-S 1250, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - M Dewolf
- Service des maladies respiratoires, Inserm UMR-S 1250, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - A Dumazet
- Service des maladies respiratoires, Inserm UMR-S 1250, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - S Dury
- Service des maladies respiratoires, Inserm UMR-S 1250, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - F Lebargy
- Service des maladies respiratoires, Inserm UMR-S 1250, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - G Deslee
- Service des maladies respiratoires, Inserm UMR-S 1250, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France
| | - J-M Perotin
- Service des maladies respiratoires, Inserm UMR-S 1250, CHU de Reims, 45, rue Cognacq-Jay, 51100 Reims, France.
| |
Collapse
|
10
|
Li Y, Qu T, Tian L, Han T, Jin Y, Wang Y. Human placenta mesenchymal stem cells suppress airway inflammation in asthmatic rats by modulating Notch signaling. Mol Med Rep 2018; 17:5336-5343. [PMID: 29363718 DOI: 10.3892/mmr.2018.8462] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 11/02/2017] [Indexed: 11/05/2022] Open
Abstract
Neurogenic locus notch homolog protein (Notch) signaling mediates intracellular communication and may regulate cell fate decisions, including cell proliferation, differentiation, and apoptosis. Mesenchymal stem cells (MSCs) possess immunomodulatory properties and the potential for use in stem cell replacement treatments. The aim of the present study was to evaluate the therapeutic effects of human placenta‑deviated MSCs (hPMSCs) in asthma and to investigate the mechanisms of Notch signaling mediated by transplanted MSCs. A Sprague‑Dawley rat ovalbumin (OVA)‑sensitized acute asthma model was established and challenged. MSCs derived from human placenta (hPMSCs) were transplanted into the asthmatic rats. Transplantation resulted in reduced Notch‑1, Notch‑2 and jagged‑1, and increased Notch‑3, Notch‑4 and delta‑like ligand (delta)‑4 expression in lung, blood, and lymph samples. Notch‑1, Notch‑2, and jagged‑1 expression in OVA‑treated rats was significantly decreased compared with controls and hPMSC‑treated rats; however, Notch‑3, Notch‑4 and delta‑4 expression was significantly increased. Serum interferon‑γ significantly increased after hPMSCs transplantation, whereas interleukin‑4 and immunoglobulin E decreased. In OVA‑treated rats, Notch‑1, Notch‑2 and jagged‑1 levels were increased in the lymph compared with the blood, although Notch‑4 and delta‑4 levels were decreased. Peribronchial infiltration of cells and goblet cell hyperplasia were markedly decreased in the OVA + hPMSCs group compared with those in the OVA‑treated and control groups. Alterations in Notch signaling pathway expression were accompanied by decreased inflammatory cell infiltration, goblet cell hyperplasia and mucus production in lung tissues. The results of the present study are consistent with hPMSC suppression of asthma symptoms and inflammation by regulating the Notch signaling pathway in the rat asthma model.
Collapse
Affiliation(s)
- Yuanyuan Li
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Tiantian Qu
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Lijun Tian
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Tingting Han
- Department of Respiratory, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Yongjun Jin
- Department of Endocrine Metabolism, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yuesi Wang
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| |
Collapse
|
11
|
Li Y, Li H, Cao Y, Wu F, Ma W, Wang Y, Sun S. Placenta‑derived mesenchymal stem cells improve airway hyperresponsiveness and inflammation in asthmatic rats by modulating the Th17/Treg balance. Mol Med Rep 2017; 16:8137-8145. [PMID: 28944907 PMCID: PMC5779899 DOI: 10.3892/mmr.2017.7605] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 08/04/2017] [Indexed: 01/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) possess reparative and immunoregulatory properties, representing a hope for stem cell-based treatments. However, the mechanisms by which transplanted MSCs affect T helper (Th)17/regulatory T cell (Treg) balance in asthma patients remain unclear. The aim of the present study was to assess the therapeutic effects of human placenta MSCs (hPMSCs) in asthma, and explore the underlying mechanisms; in addition, the impact of hPMSCs transplantation on Th17/Treg balance in lymph and serum samples from asthmatic animals was evaluated. Sprague-Dawley rats were sensitized and challenged with ovalbumin (OVA). Administration of hPMSCs from human placenta resulted in increased Th17 and Treg in lymph samples compared with peripheral blood specimens. Enhanced pause values in OVA-treated animals were significantly higher than those in the control and hPMSCs treatment groups. The numbers of total cells, macrophages, neutrophils, and eosinophils were markedly increased in the OVA group compared with those of control + hPMSCs and control groups. In addition, interleukin 10, forkhead box P3 (Foxp3) and Treg levels in lymph, peripheral blood and lung tissue samples from asthma rats were increased significantly following hPMSC transplantation. Furthermore, Foxp3 protein levels increased, while those of RAR-related orphan receptor γ (RORγt) decreased after hPMSCs transplantation compared with the asthma group. Reduced IL-17, RORγt and Th17 levels were accompanied by reduced inflammatory cell infiltration, sub-epithelial smooth layer attenuation and mucus production in lung tissues. These results suggest that hPMSCs may improve airway hyperresponsiveness and inflammation by regulating the Th17/Treg balance in rats with asthma.
Collapse
Affiliation(s)
- Yingying Li
- Department of Pediatrics, Nephrology, Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hongbo Li
- Department of Respiratory, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Yinyin Cao
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Fuling Wu
- Department of Pediatrics, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Wenbin Ma
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Yuesi Wang
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Shuzhen Sun
- Department of Pediatrics, Nephrology, Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
12
|
Hussain M, Xu C, Lu M, Wu X, Tang L, Wu X. Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3226-3242. [PMID: 28866134 DOI: 10.1016/j.bbadis.2017.08.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/10/2017] [Accepted: 08/29/2017] [Indexed: 12/23/2022]
Abstract
Embryonic lung development requires reciprocal endodermal-mesodermal interactions; mediated by various signaling proteins. Wnt/β-catenin is a signaling protein that exhibits the pivotal role in lung development, injury and repair while aberrant expression of Wnt/β-catenin signaling leads to asthmatic airway remodeling: characterized by hyperplasia and hypertrophy of airway smooth muscle cells, alveolar and vascular damage goblet cells metaplasia, and deposition of extracellular matrix; resulting in decreased lung compliance and increased airway resistance. The substantial evidence suggests that Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Here, we summarized the recent advances related to the mechanistic role of Wnt/β-catenin signaling in lung development, consequences of aberrant expression or deletion of Wnt/β-catenin signaling in expansion and progression of asthmatic airway remodeling, and linking early-impaired pulmonary development and airway remodeling later in life. Finally, we emphasized all possible recent potential therapeutic significance and future prospectives, that are adaptable for therapeutic intervention to treat asthmatic airway remodeling.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| | - Chengyun Xu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China
| | - Meiping Lu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Xiling Wu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China.
| | - Lanfang Tang
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| |
Collapse
|
13
|
Prakash YS, Halayko AJ, Gosens R, Panettieri RA, Camoretti-Mercado B, Penn RB. An Official American Thoracic Society Research Statement: Current Challenges Facing Research and Therapeutic Advances in Airway Remodeling. Am J Respir Crit Care Med 2017; 195:e4-e19. [PMID: 28084822 DOI: 10.1164/rccm.201611-2248st] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Airway remodeling (AR) is a prominent feature of asthma and other obstructive lung diseases that is minimally affected by current treatments. The goals of this Official American Thoracic Society (ATS) Research Statement are to discuss the scientific, technological, economic, and regulatory issues that deter progress of AR research and development of therapeutics targeting AR and to propose approaches and solutions to these specific problems. This Statement is not intended to provide clinical practice recommendations on any disease in which AR is observed and/or plays a role. METHODS An international multidisciplinary group from within academia, industry, and the National Institutes of Health, with expertise in multimodal approaches to the study of airway structure and function, pulmonary research and clinical practice in obstructive lung disease, and drug discovery platforms was invited to participate in one internet-based and one face-to-face meeting to address the above-stated goals. Although the majority of the analysis related to AR was in asthma, AR in other diseases was also discussed and considered in the recommendations. A literature search of PubMed was performed to support conclusions. The search was not a systematic review of the evidence. RESULTS Multiple conceptual, logistical, economic, and regulatory deterrents were identified that limit the performance of AR research and impede accelerated, intensive development of AR-focused therapeutics. Complementary solutions that leverage expertise of academia and industry were proposed to address them. CONCLUSIONS To date, numerous factors related to the intrinsic difficulty in performing AR research, and economic forces that are disincentives for the pursuit of AR treatments, have thwarted the ability to understand AR pathology and mechanisms and to address it clinically. This ATS Research Statement identifies potential solutions for each of these factors and emphasizes the importance of educating the global research community as to the extent of the problem as a critical first step in developing effective strategies for: (1) increasing the extent and impact of AR research and (2) developing, testing, and ultimately improving drugs targeting AR.
Collapse
|
14
|
Functional Effects of WNT1-Inducible Signaling Pathway Protein-1 on Bronchial Smooth Muscle Cell Migration and Proliferation in OVA-Induced Airway Remodeling. Inflammation 2016; 39:16-29. [PMID: 26242865 DOI: 10.1007/s10753-015-0218-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Upregulation of WISP1 has been demonstrated in lung remodeling. Moreover, it has been recently found that some signaling components of WNT pathway can activate GSK3β signaling to mediate remodeling of airway smooth muscle (ASM) in asthma. Therefore, we hypothesized that WISP1, a signaling molecule downstream of the WNT signaling pathway, is involved in PI3K/GSK3β signaling to mediate ASM remodeling in asthma. Our results showed that WISP1 depletion partly suppressed OVA-induced ASM hypertrophy in vivo. In vitro, WISP1 could induce hBSMC hypertrophy and proliferation, accompanied by upregulation of levels of PI3K, p-Akt, p-GSK3β, and its own expression. TGF-β treatment could increase expression of PI3K, p-Akt, p-GSK3β, and WISP1. SH-5 treatment could partly suppress TGF-β-induced hypertrophy and proliferation of hBSMC, and depress expression of p-GSK3β and WISP1. In conclusion, WISP1 may be a potential inducer of ASM proliferation and hypertrophy in asthma. The pro-remodeling effect of WISP1 is likely due to be involved in PI3K-GSK3β-dependent noncanonical TGF-β signaling.
Collapse
|
15
|
Gueugnon F, Thibault VC, Kearley J, Petit-Courty A, Vallet A, Guillon A, Si-Tahar M, Humbles AA, Courty Y. Altered expression of the CCN genes in the lungs of mice in response to cigarette smoke exposure and viral and bacterial infections. Gene 2016; 586:176-83. [PMID: 27080955 DOI: 10.1016/j.gene.2016.04.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/25/2016] [Accepted: 04/07/2016] [Indexed: 01/29/2023]
Abstract
The CCN proteins are key signaling and regulatory molecules involved in many biological functions and contribute to malignant and non-malignant lung diseases. Despite the high morbidity and mortality of the lung respiratory infectious diseases, there is very little data related to the expression of the CCNs during infection. We investigated in mice the pulmonary mRNA expression levels of five CCNs (1 to 5) in response to influenza A virus (IAV) and bacterial agents (Nontypeable Haemophilus influenzae (NTHi), lipopolysaccharide (LPS) and lipoteichoic acid (LTA)). IAV, NTHi, LPS or LTA were instilled intranasally into mice. Mice were also exposed for 4days or 8weeks to cigarette smoke alone or prior infection to IAV in order to determine if CS modifies the CCN response to a viral infection. All challenges induced a robust inflammation. The mRNA expression of CCN1, CCN2 and CCN3 was decreased after short exposure to CS whereas prolonged exposure altered the expression of CCN1, CCN3 and CCN4. Influenza A virus infection increased CCN1, 2, 4 and 5 mRNA levels but expression of CCN3 was significantly decreased. Acute CS exposure prior infection had little effect on the expression of CCN genes but prolonged exposure abolished the IAV-dependent induction. Treatment with LPS or LTA and infection with NTHi revealed that both Gram-positive and Gram-negative bacteria rapidly modulate the expression of the CCN genes. Our findings reveal that several triggers of lung inflammation influence differently the CCN genes. CCN3 deserves special attention since its mRNA expression is decreased by all the triggers studied.
Collapse
Affiliation(s)
- Fabien Gueugnon
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France; Université François Rabelais, Tours, France
| | - Virginie C Thibault
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France; Université François Rabelais, Tours, France
| | | | - Agnès Petit-Courty
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France; Université François Rabelais, Tours, France
| | - Amandine Vallet
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France; Université François Rabelais, Tours, France
| | - Antoine Guillon
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France; Université François Rabelais, Tours, France
| | - Mustapha Si-Tahar
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France; Université François Rabelais, Tours, France
| | | | - Yves Courty
- INSERM U1100, Centre d'Etude des Pathologies Respiratoires, Faculté de Médecine, Tours, France; Université François Rabelais, Tours, France.
| |
Collapse
|
16
|
WISP1 mediates IL-6-dependent proliferation in primary human lung fibroblasts. Sci Rep 2016; 6:20547. [PMID: 26867691 PMCID: PMC4751539 DOI: 10.1038/srep20547] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/20/2015] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal interstitial lung disease. IPF is characterized by epithelial cell injury and reprogramming, increases in (myo)fibroblasts, and altered deposition of extracellular matrix. The Wnt1-inducible signaling protein 1 (WISP1) is involved in impaired epithelial-mesenchymal crosstalk in pulmonary fibrosis. Here, we aimed to further investigate WISP1 regulation and function in primary human lung fibroblasts (phLFs). We demonstrate that WISP1 is directly upregulated by Transforming growth factor β1 (TGFβ1) and Tumor necrosis factor α (TNFα) in phLFs, using a luciferase-based reporter system. WISP1 mRNA and protein secretion increased in a time- and concentration-dependent manner by TGFβ1 and TNFα in phLFs, as analysed by qPCR and ELISA, respectively. Notably, WISP1 is required for TGFβ1- and TNFα-dependent induction of interleukin 6 (IL-6), a mechanism that is conserved in IPF phLFs. The siRNA-mediated WISP1 knockdown led to a significant IL-6 reduction after TGFβ1 or TNFα stimulation. Furthermore, siRNA-mediated downregulation or antibody-mediated neutralization of WISP1 reduced phLFs proliferation, a process that was in part rescued by IL-6. Taken together, these results strongly indicate that WISP1-induced IL-6 expression contributes to the pro-proliferative effect on fibroblasts, which is likely orchestrated by a variety of profibrotic mediators, including Wnts, TGFβ1 and TNFα.
Collapse
|
17
|
Wnt pathway in Dupuytren disease: connecting profibrotic signals. Transl Res 2015; 166:762-771.e3. [PMID: 26470681 DOI: 10.1016/j.trsl.2015.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 09/15/2015] [Accepted: 09/17/2015] [Indexed: 12/28/2022]
Abstract
A role of Wnt signaling in Dupuytren disease, a fibroproliferative disease of the hand and fingers, has not been fully elucidated. We examined a large set of Wnt pathway components and signaling targets and found significant dysregulation of 41 Wnt-related genes in tissue from the Dupuytren nodules compared with patient-matched control tissue. A large proportion of genes coding for Wnt proteins themselves was downregulated. However, both canonical Wnt targets and components of the noncanonical signaling pathway were upregulated. Immunohistochemical analysis revealed that protein expression of Wnt1-inducible secreted protein 1 (WISP1), a known Wnt target, was increased in nodules compared with control tissue, but knockdown of WISP1 using small interfering RNA (siRNA) in the Dupuytren myofibroblasts did not confirm a functional role. The protein expression of noncanonical pathway components Wnt5A and VANGL2 as well as noncanonical coreceptors Ror2 and Ryk was increased in nodules. On the contrary, the strongest downregulated genes in this study were 4 antagonists of Wnt signaling (DKK1, FRZB, SFRP1, and WIF1). Downregulation of these genes in the Dupuytren tissue was mimicked in vitro by treating normal fibroblasts with transforming growth factor β1 (TGF-β1), suggesting cross talk between different profibrotic pathways. Furthermore, siRNA-mediated knockdown of these antagonists in normal fibroblasts led to increased nuclear translocation of Wnt target β-catenin in response to TGF-β1 treatment. In conclusion, we have shown extensive dysregulation of Wnt signaling in affected tissue from Dupuytren disease patients. Components of both the canonical and the noncanonical pathways are upregulated, whereas endogenous antagonists are downregulated, possibly via interaction with other profibrotic pathways.
Collapse
|
18
|
Maiese K. Erythropoietin and diabetes mellitus. World J Diabetes 2015; 6:1259-1273. [PMID: 26516410 PMCID: PMC4620106 DOI: 10.4239/wjd.v6.i14.1259] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/25/2015] [Accepted: 09/28/2015] [Indexed: 02/05/2023] Open
Abstract
Erythropoietin (EPO) is a 30.4 kDa growth factor and cytokine that governs cell proliferation, immune modulation, metabolic homeostasis, vascular function, and cytoprotection. EPO is under investigation for the treatment of variety of diseases, but appears especially suited for the treatment of disorders of metabolism that include diabetes mellitus (DM). DM and the complications of this disease impact a significant portion of the global population leading to disability and death with currently limited therapeutic options. In addition to its utility for the treatment of anemia, EPO can improve cardiac function, reduce fatigue, and improve cognition in patients with DM as well as regulate cellular energy metabolism, obesity, tissue repair and regeneration, apoptosis, and autophagy in experimental models of DM. Yet, EPO can have adverse effects that involve the vasculature system and unchecked cellular proliferation. Critical to the cytoprotective capacity and the potential for a positive clinical outcome with EPO are the control of signal transduction pathways that include protein kinase B, the mechanistic target of rapamycin, Wnt signaling, mammalian forkhead transcription factors of the O class, silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae), and AMP activated protein kinase. Therapeutic strategies that can specifically target and control EPO and its signaling pathways hold great promise for the development of new and effective clinical treatments for DM and the complications of this disorder.
Collapse
|
19
|
New Insights for Oxidative Stress and Diabetes Mellitus. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:875961. [PMID: 26064426 PMCID: PMC4443788 DOI: 10.1155/2015/875961] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 04/15/2015] [Indexed: 12/12/2022]
Abstract
The release of reactive oxygen species (ROS) and the generation of oxidative stress are considered critical factors for the pathogenesis of diabetes mellitus (DM), a disorder that is growing in prevalence and results in significant economic loss. New therapeutic directions that address the detrimental effects of oxidative stress may be especially warranted to develop effective care for the millions of individuals that currently suffer from DM. The mechanistic target of rapamycin (mTOR), silent mating type information regulation 2 homolog 1 (S. cerevisiae) (SIRT1), and Wnt1 inducible signaling pathway protein 1 (WISP1) are especially justified to be considered treatment targets for DM since these pathways can address the complex relationship between stem cells, trophic factors, impaired glucose tolerance, programmed cell death pathways of apoptosis and autophagy, tissue remodeling, cellular energy homeostasis, and vascular biology that greatly impact the biology and disease progression of DM. The translation and development of these pathways into viable therapies will require detailed understanding of their proliferative nature to maximize clinical efficacy and limit adverse effects that have the potential to lead to unintended consequences.
Collapse
|
20
|
Maiese K. Programming apoptosis and autophagy with novel approaches for diabetes mellitus. Curr Neurovasc Res 2015; 12:173-88. [PMID: 25742566 PMCID: PMC4380829 DOI: 10.2174/1567202612666150305110929] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/16/2015] [Accepted: 02/19/2015] [Indexed: 12/13/2022]
Abstract
According to the World Health Organization, diabetes mellitus (DM) in the year 2030 will be ranked the seventh leading cause of death in the world. DM impacts all systems of the body with oxidant stress controlling cell fate through endoplasmic reticulum stress, mitochondrial dysfunction, alterations in uncoupling proteins, and the induction of apoptosis and autophagy. Multiple treatment approaches are being entertained for DM with Wnt1 inducible signaling pathway protein 1 (WISP1), mechanistic target of rapamycin (mTOR), and silent mating type information regulation 2 homolog) 1 (S. cerevisiae) (SIRT1) generating significant interest as target pathways that can address maintenance of glucose homeostasis as well as prevention of cellular pathology by controlling insulin resistance, stem cell proliferation, and the programmed cell death pathways of apoptosis and autophagy. WISP1, mTOR, and SIRT1 can rely upon similar pathways such as AMP activated protein kinase as well as govern cellular metabolism through cytokines such as EPO and oral hypoglycemics such as metformin. Yet, these pathways require precise biological control to exclude potentially detrimental clinical outcomes. Further elucidation of the ability to translate the roles of WISP1, mTOR, and SIRT1 into effective clinical avenues offers compelling prospects for new therapies against DM that can benefit hundreds of millions of individuals throughout the globe.
Collapse
Affiliation(s)
- Kenneth Maiese
- MD, Cellular and Molecular Signaling, Newark, New Jersey 07101, USA.
| |
Collapse
|
21
|
Chen J, Yin JY, Li XP, Wang Y, Zheng Y, Qian CY, He H, Fang C, Wang Z, Zhang Y, Xiao L, Wang SY, Zhang W, Zhou HH, Liu ZQ. Association of Wnt-Inducible Signaling Pathway Protein 1 Genetic Polymorphisms With Lung Cancer Susceptibility and Platinum-Based Chemotherapy Response. Clin Lung Cancer 2014; 16:298-304.e1-2. [PMID: 25656821 DOI: 10.1016/j.cllc.2014.12.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/24/2014] [Accepted: 12/23/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Platinum-based chemotherapy is the main treatment method for lung cancer patients. The genetic polymorphisms of Wnt-inducible signaling pathway protein 1 (WISP1) were reported to be associated with the development of diverse lung diseases. In this study, we aimed to investigate the relationship of WISP1 genetic polymorphisms with lung cancer susceptibility and platinum-based chemotherapy response in Chinese lung cancer patients. MATERIALS AND METHODS A total of 556 lung cancer patients and 254 healthy controls were enrolled onto this study. The 28 polymorphisms of the WISP1 gene were genotyped by the Sequenom MassARRAY system. RESULTS We found that WISP1 rs16893344, rs2977530, rs2977537, and rs62514004 (P = .009, .033, .049, and .036, respectively) polymorphisms were related to susceptibility of lung cancer; and WISP1 rs11778573 (P = .023, nonsmokers), rs16893344 (P = .013, ≥ 50 years old), rs2977536 (P = .039, ≥ 50 years old; P = .044, nonsmokers; P = .047, non-small-cell lung cancer, respectively), rs2977549 (P = .013, smokers), and rs62514004 (P = .033, ≥ 50 years old) polymorphisms were significantly associated with platinum-based chemotherapy response in lung cancer patients. CONCLUSION Genotypes of WISP1 may be novel and useful biomarkers for diagnosis of lung cancer and evaluation of platinum-based chemotherapy response in lung cancer patients.
Collapse
Affiliation(s)
- Juan Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China; Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China; Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| | - Xiang-Ping Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Ying Wang
- Affiliated Cancer Hospital of XiangYa School of Medicine, Central South University, Changsha, P.R. China
| | - Yi Zheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Chen-Yue Qian
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Hui He
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Chao Fang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Zhan Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Yu Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Ling Xiao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China; Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| | - Sai-Ying Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China; Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, P.R. China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, P.R. China; Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, P. R. China.
| |
Collapse
|
22
|
Boorsma CE, Dekkers BGJ, van Dijk EM, Kumawat K, Richardson J, Burgess JK, John AE. Beyond TGFβ--novel ways to target airway and parenchymal fibrosis. Pulm Pharmacol Ther 2014; 29:166-80. [PMID: 25197006 DOI: 10.1016/j.pupt.2014.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/18/2014] [Accepted: 08/26/2014] [Indexed: 01/18/2023]
Abstract
Within the lungs, fibrosis can affect both the parenchyma and the airways. Fibrosis is a hallmark pathological change in the parenchyma in patients with idiopathic pulmonary fibrosis (IPF), whilst in asthma or chronic obstructive pulmonary disease (COPD) fibrosis is a component of the remodelling of the airways. In the past decade, significant advances have been made in understanding the disease behaviour and pathogenesis of parenchymal and airway fibrosis and as a result a variety of novel therapeutic targets for slowing or preventing progression of these fibrotic changes have been identified. This review highlights a number of these targets and discusses the potential for treating parenchymal or airway fibrosis through these mediators/pathways in the future.
Collapse
Affiliation(s)
- C E Boorsma
- Department of Pharmacokinetics, Toxicology, and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - B G J Dekkers
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - E M van Dijk
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - K Kumawat
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - J Richardson
- Division of Respiratory Medicine, Nottingham University Hospitals, QMC Campus, Nottingham NG7 2UH, United Kingdom
| | - J K Burgess
- Woolcock Institute of Medical Research, Glebe 2037, Australia; Discipline of Pharmacology, The University of Sydney, Sydney 2006, Australia
| | - A E John
- Division of Respiratory Medicine, Nottingham University Hospitals, City Campus, Nottingham NG5 1PB, United Kingdom.
| |
Collapse
|