1
|
El-kady AM, Abdel-Rahman IAM, Sayed E, Wakid MH, Alobaid HM, Mohamed K, Alshehri EA, Elshazly H, Al-Megrin WAI, Iqbal F, Elshabrawy HA, Timsah AG. A potential herbal therapeutic for trichinellosis. Front Vet Sci 2022; 9:970327. [PMID: 36082215 PMCID: PMC9445247 DOI: 10.3389/fvets.2022.970327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTrichinellosis is a helminthic disease caused by Trichinella spiralis via the ingestion of raw or undercooked meat of infected animals. Current estimates indicate that 11 million humans have trichinellosis, worldwide. The effective use of anti-trichinella medications is limited by side effects and resistance which highlight the critical need for safe and effective drugs, particularly those derived from medicinal plants. Therefore, in the present study, we aimed to evaluate the efficacy of the ethanolic extract of Artemisia annua (A. annua) in treatment of experimentally induced trichinellosis.Materials and methodsTrichinellosis was induced experimentally in male 6–8 weeks BALB/c mice. BALB/c mice were divided into four groups, 10 mice each. One group was left uninfected and untreated, whereas three groups were infected with T. spiralis. One infected group of mice was left untreated (negative control) while the remaining two infected groups received either 300 mg/kg of the ethanolic extract of A. annua or 50 mg/kg of albendazole (positive control). All treatments started from the third day post-infection (dpi) for 3 successive days. All animals were sacrificed on the 7th dpi for evaluation of treatment efficacy.ResultsOur findings showed that A. annua treatment reduced the T. spiralis adult-worm count in the intestine of infected animals. Moreover, treatment with A. annua restored the normal intestinal architecture, reduced edema, alleviated inflammation as demonstrated by reduced inflammatory infiltrate and expression of TGF-β in intestinal tissues of A. annua-treated animals compared to infected untreated animals.ConclusionsOur findings show that A. annua extract is effective in treating experimentally induced trichinellosis which highlight the therapeutic potential of A. annua for intestinal trichinellosis.
Collapse
Affiliation(s)
- Asmaa M. El-kady
- Department of Medical Parasitology, Faculty of Medicine, South Valley University, Qena, Egypt
- *Correspondence: Asmaa M. El-kady
| | | | - Eman Sayed
- Department of Parasitology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Majed H. Wakid
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, Jeddah, Saudi Arabia
| | - Hussah M. Alobaid
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Khalil Mohamed
- Department of Epidemiology, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Mecca, Saudi Arabia
| | | | - Hayam Elshazly
- Department of Biology, Faculty of Sciences-Scientific Departments, Qassim University, Buraidah, Saudi Arabia
- Department of Zoology, Faculty of Science, Beni-Suef University, Beni Suef, Egypt
| | - Wafa Abdullah I. Al-Megrin
- Department of Biology, College of Science, Princess Nourah bint Adbulrahman University, Riyadh, Saudi Arabia
| | - Furhan Iqbal
- Zoology Division, Institute of Pure and Applied Biology, Bahauddin Zakariya University, Multan, Pakistan
| | - Hatem A. Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX, United States
- Hatem A. Elshabrawy
| | - Ashraf G. Timsah
- Department of Microbiology, Faculty of Medicine, Al-Baha University, Al Baha, Saudi Arabia
- Department of Parasitology, Faculty of Medicine, Al-Azhar University, New Damietta City, Egypt
| |
Collapse
|
2
|
Álvarez-Santos MD, Álvarez-González M, Eslava-De-Jesus E, González-López A, Pacheco-Alba I, Pérez-Del-Valle Y, Rojas-Madrid R, Bazán-Perkins B. Role of airway smooth muscle cell phenotypes in airway tone and obstruction in guinea pig asthma model. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2022; 18:3. [PMID: 35016714 PMCID: PMC8753847 DOI: 10.1186/s13223-022-00645-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/03/2022] [Indexed: 12/24/2022]
Abstract
Background Airway obstruction (AO) in asthma is driven by airway smooth muscle (ASM) contraction. AO can be induced extrinsically by direct stimulation of ASM with contractile agonists as histamine, or by indirect provocation with antigens as ovalbumin, while the airway tone is dependent on intrinsic mechanisms. The association of the ASM phenotypes involved in different types of AO and airway tone in guinea pigs was evaluated. Methods Guinea pigs were sensitized to ovalbumin and challenged with antigen. In each challenge, the maximum OA response to ovalbumin was determined, and before the challenges, the tone of the airways. At third challenge, airway responsiveness (AR) to histamine was evaluated and ASM cells from trachea were disaggregated to determinate: (a) by flow cytometry, the percentage of cells that express transforming growth factor-β1 (TGF-β1), interleukin-13 (IL-13) and sarco-endoplasmic Ca2+ ATPase-2b (SERCA2b), (b) by RT-PCR, the SERCA2B gene expression, (c) by ELISA, reduced glutathione (GSH) and, (d) Ca2+ sarcoplasmic reticulum refilling rate by microfluorometry. Control guinea pig group received saline instead ovalbumin. Results Antigenic challenges in sensitized guinea pigs induced indirect AO, AR to histamine and increment in airway tone at third challenge. No relationship was observed between AO induced by antigen and AR to histamine with changes in airway tone. The extent of antigen-induced AO was associated with both, TGF-β1 expression in ASM and AR degree. The magnitude of AR and antigen-induced AO showed an inverse correlation with GSH levels in ASM. The airway tone showed an inverse association with SERCA2b expression. Conclusions Our data suggest that each type of AO and airway tone depends on different ASM phenotypes: direct and indirect AO seems to be sensitive to the level of oxidative stress; indirect obstruction induced by antigen appears to be influenced by the expression of TGF-β1 and the SERCA2b expression level plays a role in the airway tone.
Collapse
Affiliation(s)
- Mayra D Álvarez-Santos
- Biology Area, Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Marisol Álvarez-González
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Mexico City, Mexico
| | - Elizabeth Eslava-De-Jesus
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Mexico City, Mexico
| | - Angel González-López
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Mexico City, Mexico
| | - Ivonne Pacheco-Alba
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Mexico City, Mexico
| | - Yazmín Pérez-Del-Valle
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Mexico City, Mexico
| | - Rodrigo Rojas-Madrid
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Mexico City, Mexico
| | - Blanca Bazán-Perkins
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Mexico City, Mexico. .,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 14380, Mexico City, Mexico.
| |
Collapse
|
3
|
D'Annunzio G, Gobbo F, Avallone G, Bacci B, Sabattini S, Sarli G. Airway Remodeling in Feline Lungs. Top Companion Anim Med 2021; 46:100587. [PMID: 34624551 DOI: 10.1016/j.tcam.2021.100587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/18/2021] [Accepted: 09/30/2021] [Indexed: 12/22/2022]
Abstract
Airway remodeling encompass structural changes that occur as the result of chronic injury and lead to persistently altered airway structure and function. Although this process is known in several human respiratory conditions such as asthma and chronic obstructive pulmonary disease (COPD), airway remodeling is poorly characterized in the feline counterpart. In this study, we describe the spontaneous pulmonary changes in 3 cats paralleling the airway remodeling reported in humans. We observed airway smooth muscle cells (ASMCs) hyperplasia (peribronchial and interstitial), airway subepithelial and interstitial fibrosis, and vascular remodeling by increased number of vessels in the bronchial submucosa. The hyperplastic ASMCs co-expressed α-SMA, vimentin and desmin suggesting that vimentin, which is not normally expressed by ASMCs, may play a role in airway thickening, and remodeling. ASMCs had strong cytoplasmic expression of TGFβ-1, which is known to contribute to tissue remodeling in asthma and in various bronchial and interstitial lung diseases, suggesting its involvement in the pathogenesis of ASMCs hyperplasia. Our findings provide histologic evidence of airway remodeling in cats. Further studies on larger caseloads are needed to support our conclusions on the value of this feline condition as an animal model for nonspecific airway remodeling in humans.
Collapse
Affiliation(s)
- Giulia D'Annunzio
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy.
| | - Francesca Gobbo
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| | - Giancarlo Avallone
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| | - Barbara Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| | - Silvia Sabattini
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, 40064 Bologna, Italy
| |
Collapse
|
4
|
Moreno-Manzano V, Zaytseva-Zotova D, López-Mocholí E, Briz-Redón Á, Løkensgard Strand B, Serrano-Aroca Á. Injectable Gel Form of a Decellularized Bladder Induces Adipose-Derived Stem Cell Differentiation into Smooth Muscle Cells In Vitro. Int J Mol Sci 2020; 21:E8608. [PMID: 33203120 PMCID: PMC7696281 DOI: 10.3390/ijms21228608] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 01/03/2023] Open
Abstract
Biologic scaffolds composed of extracellular matrix components have been proposed to repair and reconstruct a variety of tissues in clinical and pre-clinical studies. Injectable gels can fill and conform any three-dimensional shape and can be delivered to sites of interest by minimally invasive techniques. In this study, a biological gel was produced from a decellularized porcine urinary bladder by enzymatic digestion with pepsin. The enzymatic digestion was confirmed by visual inspection after dissolution in phosphate-buffered saline solution and Fourier-transform infrared spectroscopy. The rheological and biological properties of the gel were characterized and compared to those of the MatrigelTM chosen as a reference material. The storage modulus G' reached 19.4 ± 3.7 Pa for the 30 mg/mL digested decellularized bladder gels after ca. 3 h at 37 °C. The results show that the gel formed of the porcine urinary bladder favored the spontaneous differentiation of human and rabbit adipose-derived stem cells in vitro into smooth muscle cells to the detriment of cell proliferation. The results support the potential of the developed injectable gel for tissue engineering applications to reconstruct for instance the detrusor muscle part of the human urinary bladder.
Collapse
Affiliation(s)
- Victoria Moreno-Manzano
- Neuronal and Tissue Regeneration Lab, Centro de Investigación Príncipe Felipe, c/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain;
| | - Daria Zaytseva-Zotova
- NOBIPOL, Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Sem Sælands vei 6-8, N-7491 Trondheim, Norway; (D.Z.-Z.); (B.L.S.)
| | - Eric López-Mocholí
- Neuronal and Tissue Regeneration Lab, Centro de Investigación Príncipe Felipe, c/Eduardo Primo Yúfera, 3, 46012 Valencia, Spain;
| | - Álvaro Briz-Redón
- Statistics Office, City Council of Valencia, Plaza Ayuntamiento 1, 46002 Valencia, Spain;
| | - Berit Løkensgard Strand
- NOBIPOL, Department of Biotechnology and Food Science, NTNU Norwegian University of Science and Technology, Sem Sælands vei 6-8, N-7491 Trondheim, Norway; (D.Z.-Z.); (B.L.S.)
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain
| |
Collapse
|
5
|
Melatonin modulates airway smooth muscle cell phenotype by targeting the STAT3/Akt/GSK-3β pathway in experimental asthma. Cell Tissue Res 2019; 380:129-142. [PMID: 31867684 DOI: 10.1007/s00441-019-03148-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022]
Abstract
Among the troika of clinicopathologic features of asthma, airway remodelling has gained sufficient attention for its contribution to progressive airway narrowing. Much effort has been directed at the management of airway smooth muscle cells (ASMCs), but few attempts have proven to prevent the progression of remodelling. Recently, accumulating data have shown the anti-inflammatory/anti-proliferative potency of melatonin (a crucial neurohormone involved in many physiological and pathological processes) in diverse cells. However, no evidence has confirmed its effect on ASMCs. The present study investigates the benefits of melatonin in asthma, with an emphasis on airway remodelling. The results indicated that melatonin significantly attenuated airway hyperresponsiveness (AHR), inflammation and remodelling in a house dust mite (HDM) model. Melatonin markedly alleviated goblet cell hyperplasia/metaplasia, collagen deposition and airway smooth muscle hyperplasia/hypertrophy, implying the achievement of remodelling remission. The data obtained in vitro further revealed that melatonin notably inhibited ASMCs proliferation, VEGF synthesis and cell migration induced by PDGF, which might depend on STAT3 signalling. Moreover, melatonin remarkably relieved ASMCs contraction and reversed ASMCs phenotype switching induced by TGF-β, probably via the Akt/GSK-3β pathway. Altogether, our findings illustrated for the first time that melatonin improves asthmatic airway remodelling by balancing the phenotypic proportions of ASMCs, thus highlighting a novel purpose for melatonin as a potent option for the management of asthma.
Collapse
|
6
|
Novel phosphodiesterases inhibitors from the group of purine-2,6-dione derivatives as potent modulators of airway smooth muscle cell remodelling. Eur J Pharmacol 2019; 865:172779. [PMID: 31705904 DOI: 10.1016/j.ejphar.2019.172779] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/24/2019] [Accepted: 11/04/2019] [Indexed: 12/20/2022]
Abstract
Airway remodelling (AR) is an important pathological feature of chronic asthma and chronic obstructive pulmonary disease. The etiology of AR is complex and involves both lung structural and immune cells. One of the main contributors to airway remodelling is the airway smooth muscle (ASM), which is thickened by asthma, becomes more contractile and produces more extracellular matrix. As a second messenger, adenosine 3',5'-cyclic monophosphate (cAMP) has been shown to contribute to ASM cell (ASMC) relaxation as well as to anti-remodelling effects in ASMC. Phosphodiesterase (PDE) inhibitors have drawn attention as an interesting new group of potential anti-inflammatory and anti-remodelling drugs. Recently, new hydrazide and amide purine-2,6-dione derivatives with anti-inflammatory properties have been synthesized by our team (compounds 1 and 2). We expanded our study of their PDE selectivity profile, ability to increase intracellular cAMP levels, metabolic stability and, above all, their capacity to modulate cell responses associated with ASMC remodelling. The results show that both compounds have subtype specificity for several PDE isoforms (including inhibition of PDE1, PDE3, PDE4 and PDE7). Interestingly, such combined PDE subtype inhibition exerts improved anti-remodelling efficacies against several ASMC-induced responses such as proliferation, contractility, extracellular matrix (ECM) protein expression and migration when compared to other non-selective and selective PDE inhibitors. Our findings open novel perspectives in the search for new chemical entities with dual anti-inflammatory and anti-remodelling profiles in the group of purine-2,6-dione derivatives as broad-spectrum PDE inhibitors.
Collapse
|
7
|
Tetrandrine Ameliorates Airway Remodeling of Chronic Asthma by Interfering TGF- β1/Nrf-2/HO-1 Signaling Pathway-Mediated Oxidative Stress. Can Respir J 2019; 2019:7930396. [PMID: 31781316 PMCID: PMC6875008 DOI: 10.1155/2019/7930396] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/24/2019] [Accepted: 09/11/2019] [Indexed: 01/10/2023] Open
Abstract
Background Imbalanced oxidative stress and antioxidant defense are involved in airway remodeling in asthma. It has been demonstrated that Tetrandrine has a potent role in antioxidant defense in rheumatoid arthritis and hypertension. However, the correlation between Tetrandrine and oxidative stress in asthma is utterly blurry. This study aimed to investigate the role of Tetrandrine on oxidative stress-mediated airway remolding. Materials and Methods Chronic asthma was established by ovalbumin (OVA) administration in male Wistar rats. Histopathology was determined by HE staining. Immunofluorescence was employed to detect the expression of α-SMA and Nrf-2. Level of oxidative stress and matrix metalloproteinases were examined by ELISA kits. Cell viability and cell cycle of primary airway smooth muscle cells (ASMCs) were evaluated by CCK8 and flow cytometry, respectively. Signal molecules were detected using western blot. Results Tetrandrine effectively impairs OVA-induced airway inflammatory and airway remodeling by inhibiting the expression of CysLT1 and CysLTR1. The increase of oxidative stress and subsequent enhancement of MMP9 and TGF-β1 expression were rescued by the administration of Tetrandrine in the rat model of asthma. In in vitro experiments, Tetrandrine markedly suppressed TGF-β1-evoked cell viability and cell cycle promotion of ASMCs in a dose-dependent manner. Furthermore, Tetrandrine promoted Nrf-2 nuclear transcription and activated its downstream HO-1 in vivo and in vitro. Conclusion Tetrandrine attenuates airway inflammatory and airway remodeling in rat model of asthma and TGF-β1-induced cell proliferation of ASMCs by regulating oxidative stress in primary ASMCs, suggesting that Tetrandrine possibly is an effective candidate therapy for asthma.
Collapse
|
8
|
Park YH, Oh EY, Han H, Yang M, Park HJ, Park KH, Lee JH, Park JW. Insulin resistance mediates high-fat diet-induced pulmonary fibrosis and airway hyperresponsiveness through the TGF-β1 pathway. Exp Mol Med 2019; 51:1-12. [PMID: 31133649 PMCID: PMC6536500 DOI: 10.1038/s12276-019-0258-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/31/2018] [Accepted: 01/23/2019] [Indexed: 12/23/2022] Open
Abstract
Prior studies have reported the presence of lung fibrosis and enhanced airway hyperresponsiveness (AHR) in mice with high-fat-diet (HFD)-induced obesity. This study evaluated the role of TGF-β1 in HFD-induced AHR and lung fibrosis in a murine model. We generated HFD-induced obesity mice and performed glucose and insulin tolerance tests. HFD mice with or without ovalbumin sensitization and challenge were also treated with an anti-TGF-β1 neutralizing antibody. AHR to methacholine, inflammatory cells in the bronchoalveolar lavage fluid (BALF), and histological features were evaluated. Insulin was intranasally administered to normal diet (ND) mice, and in vitro insulin stimulation of BEAS-2b cells was performed. HFD-induced obesity mice had increased insulin resistance, enhanced AHR, peribronchial and perivascular fibrosis, and increased numbers of macrophages in the BALF. However, they did not have meaningful eosinophilic or neutrophilic inflammation in the lungs compared with ND mice. The HFD enhanced TGF-β1 expression in the bronchial epithelium, but we found no differences in the expression of interleukin (IL)-4 or IL-5 in lung homogenates. Administration of the anti-TGF-β1 antibody attenuated HFD-induced AHR and lung fibrosis. It also attenuated goblet cell hyperplasia, but did not affect the AHR and inflammatory cell infiltration induced by OVA challenge. The intranasal administration of insulin enhanced TGF-β1 expression in the bronchial epithelium and lung fibrosis. Stimulating BEAS-2b cells with insulin also increased TGF-β1 production by 24 h. We concluded that HFD-induced obesity-associated insulin resistance enhances TGF-β1 expression in the bronchial epithelium, which may play an important role in the development of lung fibrosis and AHR in obesity.
Collapse
Affiliation(s)
- Yoon Hee Park
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Yi Oh
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Heejae Han
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Misuk Yang
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Jung Park
- Department of Internal Medicine and Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Hee Park
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Hyun Lee
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Won Park
- Institute for Allergy, Yonsei University College of Medicine, Seoul, Korea.
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Yu Q, Yu X, Zhao W, Zhu M, Wang Z, Zhang J, Huang M, Zeng X. Inhibition of H3K27me3 demethylases attenuates asthma by reversing the shift in airway smooth muscle phenotype. Clin Exp Allergy 2018; 48:1439-1452. [PMID: 30084510 DOI: 10.1111/cea.13244] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 07/11/2018] [Accepted: 07/25/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The shift in airway smooth muscle cells (ASMCs) phenotype between proliferation and contraction during asthma has been reported recently, highlighting a role of ASMCs plasticity in the pathophysiology of asthma. As an event involved in epigenetic post-translational modification, histone H3 lysine27 (H3K27) demethylation has attracted significant attention with respect to the epigenetic changes in diverse cells; however, little is known about its contribution to the switching of ASMCs phenotype in asthma. OBJECTIVE To investigate the role of trimethylated H3K27 (H3k27me3) demethylation in ASM remodelling as well as the underling mechanism. METHODS Mice were exposed five times a week to house dust mite (HDM) extract for 5 weeks. Lung function was measured following the final HDM challenge. Airway inflammation and remodelling were then assessed in lungs of individual mice. Human ASMCs were purchased from Sciencell Research Laboratories. Proliferation, synthesis, migration and contraction of ASMCs were analysed, respectively. RESULTS We observed demethylation at H3k27me3 sites in lungs harvested from mice exposed to HDM extract. Administration of a selective inhibitor of H3K27 demethylase (GSK-J4) could ameliorate the classical hallmarks of asthma, such as airway hyperresponsiveness, airway inflammation and remodelling. We established a proliferative as well as a contractive model of human ASMCs to explore the impacts of H3K27 demethylase inhibition on ASMCs phenotype. Our results indicated that GSK-J4 decreased ASMCs proliferation and migration elicited by PDGF through the Akt/JNK signalling; GSK-J4 also prevented the upregulation of contractile proteins in ASMCs induced by TGF-β through the Smad3 pathway. CONCLUSIONS Inhibition of H3K27me3 demethylation alleviated the development of asthmatic airway disease in vivo and modulated ASMCs phenotype in vitro. Collectively, our findings highlight a role of H3K27me3 demethylation in experimental asthma and ASMCs phenotype switch.
Collapse
Affiliation(s)
- Qijun Yu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaowei Yu
- Department of Respiratory Medicine, Changzhou Second People's Hospital, Nanjing Medical University, Changzhou, Jiangsu, China
| | - Wenxue Zhao
- Department of Medicine, Lung Biology Center, University of California San Francisco, San Francisco, California
| | - Manni Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhengxia Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaxiang Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mao Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoning Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
Chen F, Shao F, Hinds A, Yao S, Ram-Mohan S, Norman TA, Krishnan R, Fine A. Retinoic acid signaling is essential for airway smooth muscle homeostasis. JCI Insight 2018; 3:120398. [PMID: 30135301 DOI: 10.1172/jci.insight.120398] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022] Open
Abstract
Airway smooth muscle (ASM) is a dynamic and complex tissue involved in regulation of bronchomotor tone, but the molecular events essential for the maintenance of ASM homeostasis are not well understood. Observational and genome-wide association studies in humans have linked airway function to the nutritional status of vitamin A and its bioactive metabolite retinoic acid (RA). Here, we provide evidence that ongoing RA signaling is critical for the regulation of adult ASM phenotype. By using dietary, pharmacologic, and genetic models in mice and humans, we show that (a) RA signaling is active in adult ASM in the normal lung, (b) RA-deficient ASM cells are hypertrophic, hypercontractile, profibrotic, but not hyperproliferative, (c) TGF-β signaling, known to cause ASM hypertrophy and airway fibrosis in human obstructive lung diseases, is hyperactivated in RA-deficient ASM, (d) pharmacologic and genetic inhibition of the TGF-β activity in ASM prevents the development of the aberrant phenotype induced by RA deficiency, and (e) the consequences of transient RA deficiency in ASM are long-lasting. These results indicate that RA signaling actively maintains adult ASM homeostasis, and disruption of RA signaling leads to aberrant ASM phenotypes similar to those seen in human chronic airway diseases such as asthma.
Collapse
Affiliation(s)
- Felicia Chen
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Fengzhi Shao
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Anne Hinds
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sean Yao
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Sumati Ram-Mohan
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Timothy A Norman
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Alan Fine
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.,Division of Pulmonary, Critical Care, and Allergy, West Roxbury Veterans Hospital, West Roxbury, Massachusetts, USA
| |
Collapse
|
11
|
Ojiaku CA, Yoo EJ, Panettieri RA. Transforming Growth Factor β1 Function in Airway Remodeling and Hyperresponsiveness. The Missing Link? Am J Respir Cell Mol Biol 2017; 56:432-442. [PMID: 27854509 DOI: 10.1165/rcmb.2016-0307tr] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of asthma includes a complex interplay among airway inflammation, hyperresponsiveness, and remodeling. Current evidence suggests that airway structural cells, including bronchial smooth muscle cells, myofibroblasts, fibroblasts, and epithelial cells, mediate all three aspects of asthma pathogenesis. Although studies show a connection between airway remodeling and changes in bronchomotor tone, the relationship between the two remains unclear. Transforming growth factor β1 (TGF-β1), a growth factor elevated in the airway of patients with asthma, plays a role in airway remodeling and in the shortening of various airway structural cells. However, the role of TGF-β1 in mediating airway hyperresponsiveness remains unclear. In this review, we summarize the literature addressing the role of TGF-β1 in airway remodeling and shortening. Through our review, we aim to further elucidate the role of TGF-β1 in asthma pathogenesis and the link between airway remodeling and airway hyperresponsiveness in asthma and to define TGF-β1 as a potential therapeutic target for reducing asthma morbidity and mortality.
Collapse
Affiliation(s)
- Christie A Ojiaku
- 1 Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Edwin J Yoo
- 1 Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and.,2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| | - Reynold A Panettieri
- 2 Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
12
|
Moore-Olufemi SD, Olsen AB, Hook-Dufresne DM, Bandla V, Cox CS. Transforming growth factor-beta 3 alters intestinal smooth muscle function: implications for gastroschisis-related intestinal dysfunction. Dig Dis Sci 2015; 60:1206-14. [PMID: 25431043 PMCID: PMC4427617 DOI: 10.1007/s10620-014-3439-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 11/11/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND Gastroschisis (GS) is a congenital abdominal wall defect that results in the development of GS-related intestinal dysfunction (GRID). Transforming growth factor-β, a pro-inflammatory cytokine, has been shown to cause organ dysfunction through alterations in vascular and airway smooth muscle. The purpose of this study was to evaluate the effects of TGF-β3 on intestinal smooth muscle function and contractile gene expression. METHODS Archived human intestinal tissue was analyzed using immunohistochemistry and RT-PCR for TGF-β isoforms and markers of smooth muscle gene and micro-RNA contractile phenotype. Intestinal motility was measured in neonatal rats ± TGF-β3 (0.2 and 1 mg/kg). Human intestinal smooth muscle cells (hiSMCs) were incubated with fetal bovine serum ± 100 ng/ml of TGF-β 3 isoforms for 6, 24 and 72 h. The effects of TGF-β3 on motility, hiSMC contractility and hiSMC contractile phenotype gene and micro-RNA expression were measured using transit, collagen gel contraction assay and RT-PCR analysis. Data are expressed as mean ± SEM, ANOVA (n = 6-7/group). RESULTS GS infants had increased immunostaining of TGF-β3 and elevated levels of micro-RNA 143 & 145 in the intestinal smooth muscle. Rats had significantly decreased intestinal transit when exposed to TGF-β3 in a dose-dependent manner compared with Sham animals. TGF-β3 significantly increased hiSMC gel contraction and contractile protein gene and micro-RNA expression. CONCLUSION TGF-β3 contributed to intestinal dysfunction at the organ level, increased contraction at the cellular level and elevated contractile gene expression at the molecular level. A hyper-contractile response may play a role in the persistent intestinal dysfunction seen in GRID.
Collapse
Affiliation(s)
- S. D. Moore-Olufemi
- Department of Pediatric Surgery, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 5.222, Houston, TX 77030 USA
| | - A. B. Olsen
- Department of Pediatric Surgery, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 5.222, Houston, TX 77030 USA
| | - D. M. Hook-Dufresne
- Department of Surgery, The University of Texas Medical School at Houston, Houston, TX USA
| | - V. Bandla
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX USA
| | - C. S. Cox
- Department of Pediatric Surgery, The University of Texas Medical School at Houston, 6431 Fannin Street, MSB 5.222, Houston, TX 77030 USA
| |
Collapse
|
13
|
Oenema TA, Mensink G, Smedinga L, Halayko AJ, Zaagsma J, Meurs H, Gosens R, Dekkers BGJ. Cross-talk between transforming growth factor-β₁ and muscarinic M₂ receptors augments airway smooth muscle proliferation. Am J Respir Cell Mol Biol 2013; 49:18-27. [PMID: 23449734 DOI: 10.1165/rcmb.2012-0261oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Transforming growth factor-β₁ (TGF-β₁) is a central mediator in tissue remodeling processes, including fibrosis and airway smooth muscle (ASM) hyperplasia, as observed in asthma. The mechanisms underlying this response, however, remain unclear because TGF-β₁ exerts only weak mitogenic effects on ASM cells. In this study, we hypothesized that the mitogenic effect of TGF-β₁ on ASM is indirect and requires prolonged exposure to allow for extracellular matrix (ECM) deposition. To address this hypothesis, we investigated the effects of acute and prolonged treatment with TGF-β₁, alone and in combination with the muscarinic receptor agonist methacholine, on human ASM cell proliferation. Acutely, TGF-β₁ exerted no mitogenic effect. However, prolonged treatment (for 7 d) with TGF-β₁ increased ASM cell proliferation and potentiated the platelet-derived growth factor-induced mitogenic response. Muscarinic receptor stimulation with methacholine synergistically enhanced the effect of TGF-β₁. Interestingly, the integrin-blocking peptide Arg-Gly-Asp-Ser, as well as integrin α5β1 function-blocking antibodies, inhibited the effects of TGF-β₁ and its combination with methacholine on cell proliferation. Accordingly, prolonged treatment with TGF-β₁ increased fibronectin expression, which was also synergistically enhanced by methacholine. The synergistic effects of methacholine on TGF-β₁-induced proliferation were reduced by the long-acting muscarinic receptor antagonist tiotropium and the M₂ receptor subtype-selective antagonist gallamine, but not the M₃-selective antagonist DAU5884. In line with these findings, the irreversible Gi protein inhibitor pertussis toxin also prevented the potentiation of TGF-β₁-induced proliferation by methacholine. We conclude that prolonged exposure to TGF-β₁ enhances ASM cell proliferation, which is mediated by extracellular matrix-integrin interactions, and which can be enhanced by muscarinic M₂ receptor stimulation.
Collapse
Affiliation(s)
- Tjitske A Oenema
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Oenema TA, Smit M, Smedinga L, Racké K, Halayko AJ, Meurs H, Gosens R. Muscarinic receptor stimulation augments TGF-β1-induced contractile protein expression by airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2012; 303:L589-97. [PMID: 22865549 DOI: 10.1152/ajplung.00400.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Acetylcholine (ACh) is the primary parasympathetic neurotransmitter in the airways. Recently, it was established that ACh, via muscarinic receptors, regulates airway remodeling in animal models of asthma and chronic obstructive pulmonary disease (COPD). The mechanisms involved are not well understood. Here, we investigated the functional interaction between muscarinic receptor stimulation and transforming growth factor (TGF)-β(1) on the expression of contractile proteins in human airway smooth muscle (ASM) cells. ASM cells expressing functional muscarinic M(2) and M(3) receptors were stimulated with methacholine (MCh), TGF-β(1), or their combination for up to 7 days. Western blot analysis revealed a strong induction of sm-α-actin and calponin by TGF-β(1), which was increased by MCh in ASM cells. Immunocytochemistry confirmed these results and revealed that the presence of MCh augmented the formation of sm-α-actin stress fibers by TGF-β(1). MCh did not augment TGF-β(1)-induced gene transcription of contractile phenotype markers. Rather, translational processes were involved in the augmentation of TGF-β(1)-induced contractile protein expression by muscarinic receptor stimulation, including phosphorylation of glycogen synthase kinase-3β and 4E-binding protein 1, which was enhanced by MCh. In conclusion, muscarinic receptor stimulation augments functional effects of TGF-β(1) in human ASM cells on cellular processes that underpin ASM remodeling in asthma and COPD.
Collapse
Affiliation(s)
- Tjitske A Oenema
- Department of Molecular Pharmacology, University of Groningen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
15
|
Gosens R, Stelmack GL, Bos ST, Dueck G, Mutawe MM, Schaafsma D, Unruh H, Gerthoffer WT, Zaagsma J, Meurs H, Halayko AJ. Caveolin-1 is required for contractile phenotype expression by airway smooth muscle cells. J Cell Mol Med 2011; 15:2430-42. [PMID: 21199324 PMCID: PMC3822954 DOI: 10.1111/j.1582-4934.2010.01246.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 12/14/2010] [Indexed: 12/18/2022] Open
Abstract
Airway smooth muscle cells exhibit phenotype plasticity that underpins their ability to contribute both to acute bronchospasm and to the features of airway remodelling in chronic asthma. A feature of mature, contractile smooth muscle cells is the presence of abundant caveolae, plasma membrane invaginations that develop from the association of lipid rafts with caveolin-1, but the functional role of caveolae and caveolin-1 in smooth muscle phenotype plasticity is unknown. Here, we report a key role for caveolin-1 in promoting phenotype maturation of differentiated airway smooth muscle induced by transforming growth factor (TGF)-β(1). As assessed by Western analysis and laser scanning cytometry, caveolin-1 protein expression was selectively enriched in contractile phenotype airway myocytes. Treatment with TGF-β(1) induced profound increases in the contractile phenotype markers sm-α-actin and calponin in cells that also accumulated abundant caveolin-1; however, siRNA or shRNAi inhibition of caveolin-1 expression largely prevented the induction of these contractile phenotype marker proteins by TGF-β(1). The failure by TGF-β(1) to adequately induce the expression of these smooth muscle specific proteins was accompanied by a strongly impaired induction of eukaryotic initiation factor-4E binding protein(4E-BP)1 phosphorylation with caveolin-1 knockdown, indicating that caveolin-1 expression promotes TGF-β(1) signalling associated with myocyte maturation and hypertrophy. Furthermore, we observed increased expression of caveolin-1 within the airway smooth muscle bundle of guinea pigs repeatedly challenged with allergen, which was associated with increased contractile protein expression, thus providing in vivo evidence linking caveolin-1 expression with accumulation of contractile phenotype myocytes. Collectively, we identify a new function for caveolin-1 in controlling smooth muscle phenotype; this mechanism could contribute to allergic asthma.
Collapse
Affiliation(s)
- Reinoud Gosens
- Departments of Physiology & Internal Medicine, University of ManitobaWinnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child HealthWinnipeg, Manitoba, Canada
- Department of Molecular Pharmacology, University of GroningenGroningen, The Netherlands
| | - Gerald L Stelmack
- Departments of Physiology & Internal Medicine, University of ManitobaWinnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child HealthWinnipeg, Manitoba, Canada
| | - Sophie T Bos
- Department of Molecular Pharmacology, University of GroningenGroningen, The Netherlands
| | - Gordon Dueck
- Departments of Physiology & Internal Medicine, University of ManitobaWinnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child HealthWinnipeg, Manitoba, Canada
| | - Mark M Mutawe
- Departments of Physiology & Internal Medicine, University of ManitobaWinnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child HealthWinnipeg, Manitoba, Canada
| | - Dedmer Schaafsma
- Departments of Physiology & Internal Medicine, University of ManitobaWinnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child HealthWinnipeg, Manitoba, Canada
| | - Helmut Unruh
- Section of Thoracic Surgery, University of ManitobaWinnipeg, Manitoba, Canada
| | - William T Gerthoffer
- Department of Pharmacology, University of Nevada School of MedicineReno, NV, USA
| | - Johan Zaagsma
- Department of Molecular Pharmacology, University of GroningenGroningen, The Netherlands
| | - Herman Meurs
- Department of Molecular Pharmacology, University of GroningenGroningen, The Netherlands
| | - Andrew J Halayko
- Departments of Physiology & Internal Medicine, University of ManitobaWinnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child HealthWinnipeg, Manitoba, Canada
| |
Collapse
|
16
|
Chou MT, Chang SN, Ke C, Chang HI, Sung ML, Kuo HC, Chen CN. The proliferation and differentiation of placental-derived multipotent cells into smooth muscle cells on fibrillar collagen. Biomaterials 2010; 31:4367-75. [PMID: 20199810 DOI: 10.1016/j.biomaterials.2010.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 02/05/2010] [Indexed: 01/10/2023]
Abstract
Type I collagen constitutes a major portion of the extracellular matrix (ECM) in arterial wall and it is the major substrate for cell growth and differentiation. The goal of this study was to evaluate the differentiation and proliferation of placenta-derived multipotent cells (PDMCs) on polymerized type I collagen fibrils and monomer collagen. PDMCs grown on both polymerized collagen and monomer collagen with transforming growth factor (TGF)-beta treatment increases the expression of smooth muscle cell (SMC)-specific markers, including calponin, alpha-smooth muscle actin (alpha-SMA) and smooth muscle-myosin heavy chain (SM-MHC). Polymerized collagen increased the expressions of p21(CIP1) and p27(KIP1); decreased cyclin A, cyclin D1, cyclin-dependent protein kinase 2 (Cdk2); and led to G(0)/G(1) arrest in PDMCs. Furthermore, PDMC-differentiated SMCs exhibited significant collagen contractility in the presence or absence of endothelin-1 (ET-1) stimulation. By using specific inhibitors and small interfering RNA (siRNA), we demonstrated that p38 MAPK pathway and serum response factor (SRF)-DNA binding activity is critical for the polymerized collagen-induced PDMC differentiation into SMCs. Thus, polymerized collagen exhibits the great potential in inducing PDMCs differentiation into SMCs, and exerts anti-proliferative effect on PDMC-differentiated SMCs.
Collapse
Affiliation(s)
- Mou-Tsy Chou
- Department of Gynecology, St. Martin De Porres Hospital, Chiayi City, Taiwan
| | | | | | | | | | | | | |
Collapse
|
17
|
Hirota JA, Nguyen TTB, Schaafsma D, Sharma P, Tran T. Airway smooth muscle in asthma: phenotype plasticity and function. Pulm Pharmacol Ther 2008; 22:370-8. [PMID: 19114115 DOI: 10.1016/j.pupt.2008.12.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 11/14/2008] [Accepted: 12/10/2008] [Indexed: 10/24/2022]
Abstract
Clinical asthma is characterized by reversible airway obstruction which is commonly due to an exaggerated airway narrowing referred to as airway hyperresponsiveness (AHR). Although debate exists on the complex etiology of AHR, it is clear that airway smooth muscle (ASM) mediated airway narrowing is a major contributor to airway dysfunction. More importantly, it is now appreciated that smooth muscle is far from being a simple cell with only contractile ability properties. Rather, it is more versatile with the capacity to exhibit numerous cellular functions as it adapts to the microenvironment to which it is exposed. The emerging ability of individual smooth muscle cells to undergo changes in their phenotype (phenotype plasticity) and function (functional plasticity) in response to physiological and pathological cues is an important and active area of research. This article provides a brief review of the current knowledge and emerging concepts in the field of ASM phenotype and function both under healthy and asthmatic conditions.
Collapse
Affiliation(s)
- Jeremy A Hirota
- Firestone Institute for Respiratory Health, McMaster University, Ontario, Canada
| | | | | | | | | |
Collapse
|
18
|
Xiao Q, Luo Z, Pepe AE, Margariti A, Zeng L, Xu Q. Embryonic stem cell differentiation into smooth muscle cells is mediated by Nox4-produced H2O2. Am J Physiol Cell Physiol 2008; 296:C711-23. [PMID: 19036941 DOI: 10.1152/ajpcell.00442.2008] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
NADPH oxidase (Nox4) produces reactive oxygen species (ROS) that are important for vascular smooth muscle cell (SMC) behavior, but the potential impact of Nox4 in stem cell differentiation is unknown. When mouse embryonic stem (ES) cells were plated on collagen IV-coated dishes/flasks, a panel of SMC-specific genes was significantly and consistently upregulated. Nox4 expression was markedly correlated with such a gene induction as confirmed by real-time PCR, immunofluorescence, and Western blot analysis. Overexpression of Nox4 specifically resulted in increased SMC marker production, whereas knockdown of Nox4 induced a decrease. Furthermore, SMC-specific transcription factors, including serum response factor (SRF) and myocardin were activated by Nox4 gene expression. Moreover, Nox4 was demonstrated to drive SMC differentiation through generation of H(2)O(2). Confocal microscopy analysis indicates that SRF was translocated into the nucleus during SMC differentiation in which SRF was phosphorylated. Additionally, autosecreted transforming growth factor (TGF)-beta(1) activated Nox4 and promoted SMC differentiation. Interestingly, cell lines generated from stem cells by Nox4 transfection and G418 selection displayed a characteristic of mature SMCs, including expression of SMC markers and cells with contractile function. Thus we demonstrate for the first time that Nox4 is crucial for SMC differentiation from ES cells, and enforced Nox4 expression can maintain differentiation status and functional features of stem cell-derived SMCs, highlighting its impact on vessel formation in vivo and vascular tissue engineering in the future.
Collapse
Affiliation(s)
- Qingzhong Xiao
- Cardiovascular Div., King's College London, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU
| | | | | | | | | | | |
Collapse
|