1
|
The Protective Effect of Simvastatin on the Systolic Function of the Heart in the Model of Acute Ischemia and Reperfusion Is Due to Inhibition of the RhoA Pathway and Independent of Reduction of MMP-2 Activity. Biomolecules 2022; 12:biom12091291. [PMID: 36139129 PMCID: PMC9496379 DOI: 10.3390/biom12091291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
The present study investigated whether Rho-associated protein kinase (RhoA/ROCK) signaling pathway inhibitor simvastatin inhibits matrix metalloproteinase 2 (MMP-2) activity in a rat ischemia-reperfusion injury (I/Ri) model by inhibiting the RhoA/ROCK pathway and reducing MMP-2 mRNA levels. Isolated rat hearts were subjected to aerobic perfusion or I/Ri control. The effect of simvastatin was assessed in hearts subjected to I/Ri. We determined cardiac mechanical function, the content of RhoA, phosphorylated myosin light chain subunit 1 (phospho-MYL9), troponin I, MMP-2, and MMP-2 mRNA in the heart homogenates, as well as MMP-2 activity in heart tissue. We showed that treatment with simvastatin caused improvement in the contractile function of the heart subjected to I/Ri which was accompanied by a decrease of MMP-2 activity in heart tissue along with inhibition of RhoA pathway, expressed in a reduction in both RhoA and its downstream product—phosphorylated myosin light chain (phospho-MYL9) in hearts treated with simvastatin. MMP-2 inactivation is not due to inhibition of MMP-2 m-RNA synthesis caused by inhibition of RhoA/ROCK pathway and is due, at least in part, to the direct drug action. The protective effect of simvastatin on systolic function in the acute ischemia-reperfusion model does not appear to be related to reduced MMP-2 activation, but other mechanisms related with the inhibition RhoA/ROCK pathway.
Collapse
|
2
|
Asare PF, Tran HB, Hurtado PR, Perkins GB, Nguyen P, Jersmann H, Roscioli E, Hodge S. Inhibition of LC3-associated phagocytosis in COPD and in response to cigarette smoke. Ther Adv Respir Dis 2021; 15:17534666211039769. [PMID: 34852704 PMCID: PMC8647217 DOI: 10.1177/17534666211039769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION/RATIONALE In chronic obstructive pulmonary disease (COPD), defective macrophage phagocytic clearance of cells undergoing apoptosis by efferocytosis may lead to secondary necrosis of the uncleared cells and contribute to airway inflammation. The precise mechanisms for this phenomenon remain unknown. LC3-associated phagocytosis (LAP) is indispensable for effective efferocytosis. We hypothesized that cigarette smoke inhibits the regulators of LAP pathway, potentially contributing to the chronic airways inflammation associated with COPD. METHODS Bronchoalveolar (BAL)-derived alveolar macrophages, lung tissue macrophages obtained from lung resection surgery, and monocyte-derived macrophages (MDM) were prepared from COPD patients and control participants. Lung/airway samples from mice chronically exposed to cigarette smoke were also investigated. Differentiated THP-1 cells were exposed to cigarette smoke extract (CSE). The LAP pathway including Rubicon, as an essential regulator of LAP, efferocytosis and inflammation was examined using western blot, ELISA, flow cytometry, and/or immunofluorescence. RESULTS Rubicon was significantly depleted in COPD alveolar macrophages compared with non-COPD control macrophages. Rubicon protein in alveolar macrophages of cigarette smoke-exposed mice and cigarette smoke-exposed MDM and THP-1 was decreased with a concomitant impairment of efferocytosis. We also noted increased expression of LC3 which is critical for LAP pathway in COPD and THP-1 macrophages. Furthermore, THP-1 macrophages exposed to cigarette smoke extract exhibited higher levels of other key components of LAP pathway including Atg5 and TIM-4. There was a strong positive correlation between Rubicon protein expression and efferocytosis. CONCLUSION LAP is a requisite for effective efferocytosis and an appropriate inflammatory response, which is impaired by Rubicon deficiency. Our findings suggest dysregulated LAP due to reduced Rubicon as a result of CSE exposure. This phenomenon could lead to a failure of macrophages to effectively process phagosomes containing apoptotic cells during efferocytosis. Restoring Rubicon protein expression has unrecognized therapeutic potential in the context of disease-related modifications caused by exposure to cigarette smoke.
Collapse
Affiliation(s)
- Patrick F Asare
- Department of Thoracic Medicine, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Hai B Tran
- Department of Thoracic Medicine, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Plinio R Hurtado
- Department of Renal Medicine, School of Medicine, The University of Adelaide, Adelaide, SA, Australia.,Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Griffith B Perkins
- Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Phan Nguyen
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Hubertus Jersmann
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Eugene Roscioli
- Department of Thoracic Medicine, School of Medicine, The University of Adelaide, Adelaide, SA, Australia.,Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Sandra Hodge
- Department of Thoracic Medicine, Faculty of Health and Medical Science, The University of Adelaide, Adelaide, SA 5005, Australia.,School of Medicine, The University of Adelaide, Adelaide, SA, Australia.,Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
3
|
Jin P, Deng S, Sherchan P, Cui Y, Huang L, Li G, Lian L, Xie S, Lenahan C, Travis ZD, Zhang JH, Gong Y, Tang J. Neurokinin Receptor 1 (NK1R) Antagonist Aprepitant Enhances Hematoma Clearance by Regulating Microglial Polarization via PKC/p38MAPK/NFκB Pathway After Experimental Intracerebral Hemorrhage in Mice. Neurotherapeutics 2021; 18:1922-1938. [PMID: 34244927 PMCID: PMC8608951 DOI: 10.1007/s13311-021-01077-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2021] [Indexed: 02/04/2023] Open
Abstract
Hematoma clearance is an important therapeutic target to improve outcome following intracerebral hemorrhage (ICH). Recent studies showed that Neurokinin receptor-1 (NK1R) inhibition exerts protective effects in various neurological disease models, but its role in ICH has not been explored. The objective of this study was to investigate the role of NK1R and its relation to hematoma clearance after ICH using an autologous blood injection mouse model. A total of 332 adult male CD1 mice were used. We found that the expression levels of NK1R and its endogenous ligand, substance P (SP), were significantly upregulated after ICH. Intraperitoneal administration of the NK1R selective antagonist, Aprepitant, significantly improved neurobehavior, reduced hematoma volume and hemoglobin levels after ICH, and promoted microglia polarization towards M2 phenotype. Aprepitant decreased phosphorylated PKC, p38MAPK, and NFκB p65, and downregulated M1 markers while upregulating M2 markers after ICH. Intracerebroventricular administration of the NK1R agonist, GR73632 or PKC agonist, phorbol 12-myristate 13-acetate (PMA) reversed the effects of Aprepitant. To demonstrate the upstream mediator of NK1R activation, we performed thrombin injection and found that it increased SP. Inhibiting thrombin suppressed SP and decreased M1 markers while increasing M2 microglia polarization. Thus, NK1R inhibition promoted hematoma clearance after ICH by increasing M2 microglial polarization via downregulating PKC/p38MAPK/NFκB signaling pathway, and thrombin may be a key upstream mediator of NK1R activation. Therapeutic interventions inhibiting NK1R signaling may be a new target for the treatment of ICH.
Collapse
Affiliation(s)
- Peng Jin
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Shuixiang Deng
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Yuhui Cui
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Gaigai Li
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Lifei Lian
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Shucai Xie
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
- Burrell College of Osteopathic Medicine, Las Cruces, NM, 88001, USA
| | - Zachary D Travis
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Ye Gong
- Department of Intensive Care Unit, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, 92350, USA.
| |
Collapse
|
4
|
Iannarelli NJ, MacNeil AJ, Dempster KS, Wade TJ, O’Leary DD. Serum MMP-3 and its association with central arterial stiffness among young adults is moderated by smoking and BMI. Physiol Rep 2021; 9:e14920. [PMID: 34110720 PMCID: PMC8191404 DOI: 10.14814/phy2.14920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 12/21/2022] Open
Abstract
Central arterial stiffness is an independent predictor of cardiovascular disease. It is characterized by a marked reduction in the elastin-collagen ratio of the arterial wall extracellular matrix (ECM), and is largely the result of degradation of various ECM components. Matrix metalloproteinase-3 (MMP-3) may contribute to central arterial stiffness via its involvement in ECM homeostasis and remodeling. This study examined the association between serum MMP-3 concentrations and central arterial stiffness and potential interactions of MMP-3 and traditional cardiovascular risk factors in a population of healthy young adults. A total of 206 participants (n = 109 females) aged 19-25 years were included in the current study. Central arterial stiffness was measured non-invasively as carotid-femoral pulse wave velocity (cfPWV) (m/s). MMP-3 concentrations (ng/ml) were measured using ELISA techniques. Regression analyses were used to examine the association between cfPWV and MMP-3, adjusting for age, sex, smoking status, body mass index (BMI), instantaneous mean arterial pressure (MAP) and heart rate, and serum C-reactive protein. Interactions between MMP-3 with smoking, BMI, sex, and MAP were analyzed in subsequent regression models. MMP-3 was an independent predictor of cfPWV (β = 0.187, p = 0.007), and significant interactions between MMP-3 and regular smoking (β = 0.291, p = 0.022), and MMP-3 and BMI (β = 0.210, p = 0.013) were observed. Higher serum MMP-3 concentrations were associated with a faster cfPWV and thus, greater central arterial stiffness. Interactions between MMP-3 and smoking, and MMP-3 and BMI may, in part, drive the association between MMP-3 and central arterial stiffness.
Collapse
Affiliation(s)
- Nathaniel J. Iannarelli
- Department of Health SciencesFaculty of Applied Health SciencesBrock UniversitySaint CatharinesONCanada
| | - Adam J. MacNeil
- Department of Health SciencesFaculty of Applied Health SciencesBrock UniversitySaint CatharinesONCanada
| | - Kylie S. Dempster
- Department of Health SciencesFaculty of Applied Health SciencesBrock UniversitySaint CatharinesONCanada
- Brock‐Niagara Centre for Health and Well‐BeingBrock UniversitySaint CatharinesONCanada
| | - Terrance J. Wade
- Department of Health SciencesFaculty of Applied Health SciencesBrock UniversitySaint CatharinesONCanada
- Brock‐Niagara Centre for Health and Well‐BeingBrock UniversitySaint CatharinesONCanada
| | - Deborah D. O’Leary
- Department of Health SciencesFaculty of Applied Health SciencesBrock UniversitySaint CatharinesONCanada
- Brock‐Niagara Centre for Health and Well‐BeingBrock UniversitySaint CatharinesONCanada
| |
Collapse
|
5
|
Mehta S, Srivastava N, Bhatia A, Dhawan V. Exposure of cigarette smoke condensate activates NLRP3 inflammasome in vitro and in vivo: A connotation of innate immunity and atherosclerosis. Int Immunopharmacol 2020; 84:106561. [PMID: 32402952 DOI: 10.1016/j.intimp.2020.106561] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/21/2020] [Accepted: 05/02/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Smoking is known to have detrimental effects on cardiovascular system. However, the potential molecular basis of smoking-induced atherosclerosis remains unclear. NLRP3 inflammasome is implicated in perpetuation of inflammatory response in atherosclerosis. Therefore, we aimed to explore the cytotoxic effects of cigarette smoke condensate (CSC) on the activation of NLRP3 inflammasome in vitro and in vivo. METHODS For in vitro study, the pro-atherogenic effects of CSC were evaluated in THP-1 monocytes with different dose concentrations (0.1, 1, 5, 10 and 20 µg/ml) for varied time periods (6, 12, 24 and 48 h). For in vivo study, 30 male C57BL/6J mice were employed. 6 mice were sacrificed for baseline investigations. 24 mice were randomly divided into four groups: Group-I:Control mice, Group-II:CSC model, Group-III:High-fat diet(HFD) model, and Group-IV:HFD + CSC model for 14 weeks (n = 6/group). The group-II and IV mice were injected with 720 µg CSC/20 g body weight intraperitoneally (6 days/week). RESULTS In vitro, higher dosage of CSC (20 µg/ml) was toxic to cells as significant decline in cell viability and proliferation was observed. Furthermore, the mRNA expression of NLRP3 inflammasome and its pro-cytokine levels were significantly augmented on CSC exposure in a dose-dependent manner but impeded in time-dependent manner. In vivo, CSC and HFD independently augmented the expression of NLRP3 inflammasome (~4-10 fold-change) along with pro-cytokine levels in Group-II and III vs Group-I mice whereas, HFD + CSC treatment demonstrated synergistic effects in Group-IV. CONCLUSION Our data suggest that CSC activates NLRP3 inflammasome in vitro and in vivo and collectively with HFD has synergistic effects in vivo that may promote atherosclerosis.
Collapse
Affiliation(s)
- Sakshi Mehta
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Niharika Srivastava
- Department of Dermatology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Veena Dhawan
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
| |
Collapse
|
6
|
Jiang W, Wang X, Wang W, Hua F, Zhang Z, Zhang Z, Xiang J, Yang X. Inhibition of NK1R attenuates LPS-induced microglial inflammation and consequent death of PC12 cells. Brain Res Bull 2020; 162:115-124. [PMID: 32540418 DOI: 10.1016/j.brainresbull.2020.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 10/24/2022]
Abstract
Microglia, the resident immune cells in the central nervous system, play a critical role under physiological conditions, but they may be activated and exaggerate the pathological development of Parkinson's disease (PD). Recent reports have suggested that neurokinin 1 receptor (NK1R) is involved in various inflammatory diseases, including PD. However, whether neurokinin 1 (NK1) is involved in the activation of microglial cells remains unclear. In the present study, we found that (1) NK1R is located in microglial cells and upregulated in lipopolysaccharide (LPS)-activated BV2 microglia. Application of CP-99994, a selective antagonist of NK1R, inhibited the production of inflammatory mediators such as tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), IL-6, inducible macrophage-type nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) in activated BV2 cells. (2) NK1R antagonist suppressed the morphological changes in LPS-stimulated BV2. (3) Microglial inactivation by NK1R antagonist resulted in decreased microglial migration. (4) NK1R antagonist reduced nuclear translocation of nuclear factor kappa-B (NF-κB) and attenuated phosphorylation of mitogen-activated protein kinases (MAPKs) in LPS-stimulated BV2. (5) The cell death of PC12 induced by microglia-mediated neuroinflammation was reversed in a Transwell co-culture system by NK1R antagonist. Collectively, these results showed that inhibition of NK1R attenuates LPS-induced microglial inflammatory response and dopaminergic neurotoxicity, which may be due to the decreased MAPK/NF-κB signal pathway. Thus, NK1R may be a therapeutic target in neuroinflammation, especially in PD.
Collapse
Affiliation(s)
- Weifeng Jiang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Xiaoying Wang
- Department of Ultrasound, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Wei Wang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zunsheng Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zuohui Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Jie Xiang
- Department of Rehabilitation, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Xinxin Yang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China; Institute of Neurological Diseases of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| |
Collapse
|
7
|
Brodskaya TA, Nevzorova VA, Vasileva MS, Lavrenyuk VV. [Endothelium-related and neuro-mediated mechanisms of emphysema development in chronic obstructive pulmonary disease]. TERAPEVT ARKH 2020; 92:116-124. [PMID: 32598803 DOI: 10.26442/00403660.2020.03.000347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Indexed: 11/22/2022]
Abstract
Emphysema is one of the main manifestations of chronic obstructive pulmonary disease (COPD), and smoking is one of the most significant risk factors. The results of studies in humans and animals show the vascular endothelium initiates and modulates the main pathological processes in COPD and smoking is an important factor initiating, developing and persisting inflammation and remodeling of blood vessels and tissues, including the destruction of small respiratory tracts with the development of lung tissue destruction and emphysema. The latest studies describe mechanisms not just associated with the endothelium, but specific neuro-mediated mechanisms. There is reason to believe that neuro-mediated and neuro-similar mechanisms associated and not related to endothelial dysfunction may play the significant role in the pathogenesis of COPD and emphysema formation. Information about components and mechanisms of neurogenic inflammation in emphysema development is fragmentary and not systematized in the literature. It is described that long-term tobacco smoking can initiate processes not only of cells and tissues damage, but also become a trigger for excessive release of neurotransmitters, which entails whole cascades of adverse reactions that have an effect on emphysema formation. With prolonged and/or intensive stimulation of sensor fibers, excessive release of neuropeptides is accompanied by a number of plastic and destructive processes due to a cascade of pathological reactions of neurogenic inflammation, the main participants of which are classical neuropeptides and their receptors. The most important consequences can be the maintenance and stagnation of chronic inflammation, activation of the mechanisms of destruction and remodeling, inadequate repair processes in response to damage, resulting in irreversible loss of lung tissue. For future research, there is interest to evaluate the possibilities of therapeutic and prophylactic effects on neuro-mediated mechanisms of endothelial dysfunction and damage emphysema in COPD and smoking development.
Collapse
|
8
|
Mehta S, Dhawan V. Exposure of cigarette smoke condensate activates NLRP3 inflammasome in THP-1 cells in a stage-specific manner: An underlying role of innate immunity in atherosclerosis. Cell Signal 2020; 72:109645. [PMID: 32305666 DOI: 10.1016/j.cellsig.2020.109645] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/24/2020] [Accepted: 04/15/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Smoking is known to affect all the phases of atherosclerosis, thus is considered as an independent and major risk factor. The underlying mechanism responsible for the atherogenic effects of smoking is still uncertain and a major concern. Recent evidence implicates NLRP3 inflammasome, an innate immunity component in the pathogenesis of atherosclerosis. Therefore, we hypothesized that NLRP3 inflammasome may be an associated pathway between smoking and atherosclerosis. METHODS AND RESULTS Differentiation in monocytes, macrophages and foam cells are the key stages in atherosclerotic plaque development, best mimicked by THP-1 cells. Therefore, to determine whether cigarette smoke condensate (CSC) could induce differentiation of THP-1 monocytes into macrophages, morphological changes and the expression levels of the inflammatory surface markers, i.e. CD11b, CD14 and CD36 were analyzed. The results showed that CD14 and CD36 levels were significantly increased in CSC-treated THP-1 monocytes. Further, we investigated the effect of CSC exposure on the status of NLRP3 inflammasome markers, i.e. NLRP3, pro-caspase-1, caspase-1, pro-IL-18, pro-IL-1β, IL-1β and IL-18 in a stage-specific manner. For this, THP-1 monocytes, PMA-differentiated macrophages and oxidized-low density lipoprotein (ox-LDL)-induced macrophage foam cells were exposed to 10 μg/ml of CSC for 6 h. CSC exposure significantly upregulated the expression of NLRP3 inflammasome in CSC-treated cells at both transcriptional and translational levels. Moreover, downstream pro-cytokines, i.e. IL-1β and IL-18 levels were also significantly increased in culture supernatants of CSC-exposed cells. CONCLUSION These observations suggest that CSC exposure may activate NLRP3 inflammasome in a stage-specific manner and may promote initiation and progression of atherosclerosis.
Collapse
Affiliation(s)
- Sakshi Mehta
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Veena Dhawan
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
| |
Collapse
|
9
|
Henkel M, Partyka J, Gregory AD, Forno E, Cho MH, Eddens T, Tout AR, Salamacha N, Horne W, Rao KS, Wu Y, Alcorn JF, Kostka D, Hirsch R, Celedón JC, Shapiro SD, Kolls JK, Campfield BT. FSTL-1 Attenuation Causes Spontaneous Smoke-Resistant Pulmonary Emphysema. Am J Respir Crit Care Med 2020; 201:934-945. [PMID: 31834999 PMCID: PMC7159415 DOI: 10.1164/rccm.201905-0973oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 12/10/2019] [Indexed: 12/19/2022] Open
Abstract
Rationale: The role of FSTL-1 (follistatin-like 1) in lung homeostasis is unknown.Objectives: We aimed to define the impact of FSTL-1 attenuation on lung structure and function and to identify FSTL-1-regulated transcriptional pathways in the lung. Further, we aimed to analyze the association of FSTL-1 SNPs with lung disease.Methods: FSTL-1 hypomorphic (FSTL-1 Hypo) mice underwent lung morphometry, pulmonary function testing, and micro-computed tomography. Fstl1 expression was determined in wild-type lung cell populations from three independent research groups. RNA sequencing of wild-type and FSTL-1 Hypo mice identified FSTL-1-regulated gene expression, followed by validation and mechanistic in vitro examination. FSTL1 SNP analysis was performed in the COPDGene (Genetic Epidemiology of Chronic Obstructive Pulmonary Disease) cohort.Measurements and Main Results: FSTL-1 Hypo mice developed spontaneous emphysema, independent of smoke exposure. Fstl1 is highly expressed in the lung by mesenchymal and endothelial cells but not immune cells. RNA sequencing of whole lung identified 33 FSTL-1-regulated genes, including Nr4a1, an orphan nuclear hormone receptor that negatively regulates NF-κB (nuclear factor-κB) signaling. In vitro, recombinant FSTL-1 treatment of macrophages attenuated NF-κB p65 phosphorylation in an Nr4a1-dependent manner. Within the COPDGene cohort, several SNPs in the FSTL1 region corresponded to chronic obstructive pulmonary disease and lung function.Conclusions: This work identifies a novel role for FSTL-1 protecting against emphysema development independent of smoke exposure. This FSTL-1-deficient emphysema implicates regulation of immune tolerance in lung macrophages through Nr4a1. Further study of the mechanisms involving FSTL-1 in lung homeostasis, immune regulation, and NF-κB signaling may provide additional insight into the pathophysiology of emphysema and inflammatory lung diseases.
Collapse
Affiliation(s)
- Matthew Henkel
- Division of Pediatric Infectious Diseases
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessica Partyka
- Division of Pediatric Infectious Diseases
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alyssa D. Gregory
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine
- Richard K. Mellon Institute for Pediatric Research, and
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael H. Cho
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts; and
| | - Taylor Eddens
- Division of Pediatric Infectious Diseases
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Nathan Salamacha
- Department of Developmental Biology
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - William Horne
- Richard K. Mellon Institute for Pediatric Research, and
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Yijen Wu
- Department of Developmental Biology
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - John F. Alcorn
- Division of Pediatric Pulmonary Medicine
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dennis Kostka
- Department of Developmental Biology
- Department of Computational and Systems Biology, and
- Center for Evolutionary Biology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Raphael Hirsch
- Stead Family Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Juan C. Celedón
- Division of Pediatric Pulmonary Medicine
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Steven D. Shapiro
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Jay K. Kolls
- Richard K. Mellon Institute for Pediatric Research, and
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - Brian T. Campfield
- Division of Pediatric Infectious Diseases
- Richard K. Mellon Institute for Pediatric Research, and
- University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Fan Y, Zhang Z, Yao C, Bai J, Yang H, Ma P, Fan Y, Li S, Yuan J, Lin M, Hou Q. Amurensin H, a Derivative From Resveratrol, Ameliorates Lipopolysaccharide/Cigarette Smoke-Induced Airway Inflammation by Blocking the Syk/NF-κB Pathway. Front Pharmacol 2019; 10:1157. [PMID: 31636566 PMCID: PMC6787933 DOI: 10.3389/fphar.2019.01157] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/09/2019] [Indexed: 12/20/2022] Open
Abstract
Amurensin H, a resveratrol dimer derived from Vitis amurensis Rupr, has several biological effects, including anti-inflammatory and antioxidant activities. Studies have found that amurensin H attenuated asthma-like allergic airway inflammation. However, its protective activity on chronic obstructive pulmonary disease (COPD) airway inflammation is not fully explored. The present study used a lipopolysaccharide (LPS)/cigarette smoke-induced mice model and an LPS-stimulated THP-1-derived macrophages model to measure the lung tissue's morphology changes. The results showed that amurensin H ameliorated the histological inflammatory alterations in the lung tissues, leading to a decrease in the expression of interleukin 6 (IL-6), IL-17A, tumor necrosis factor α (TNF-α), and interferon γ in bronchoalveolar lavage fluid. Amurensin H also significantly inhibited the release of IL-1β, IL-6, IL-8, and TNF-α in LPS-stimulated THP-1-derived macrophages. Furthermore, amurensin H markedly inhibited the expressions of p-Syk, nuclear factor κB (NF-κB), and p-NF-κB both in vivo and in vitro. Results from cotreatment with Syk inhibitor BAY61-3606 and NF-κB inhibitor BAY11-7082 in vitro revealed that amurensin H's protective effect against airway inflammation could be due partly to the inhibition of the Syk/NF-κB pathway. These findings suggest that amurensin H shows therapeutic effects on COPD airway inflammation, and inhibiting the Syk/NF-κB pathway might be part of its underlying mechanisms.
Collapse
Affiliation(s)
- Yannan Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziqian Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunsuo Yao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinye Bai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pei Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiyao Fan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyi Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiqiao Yuan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingbao Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Hou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
The effects of cigarette smoking extracts on cell cycle and tumor spread: novel evidence. Future Sci OA 2019. [DOI: 10.4155/fsoa-2019-0017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
12
|
Pezzuto A, Citarella F, Croghan I, Tonini G. The effects of cigarette smoking extracts on cell cycle and tumor spread: novel evidence. Future Sci OA 2019; 5:FSO394. [PMID: 31205749 PMCID: PMC6556819 DOI: 10.2144/fsoa-2019-0017] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cigarette smoking is a major preventable risk factor for lung cancer, contributing to lung cancer progression and metastasis. Moreover, cigarette smoking correlates with increased metastasis frequency of pancreatic, breast and bladder cancer. The aim of this review was to examine the role of cigarette smoke extract in cell cycle and cancer progression. Clinical impact and the effects of cigarette smoke extract on carcinogenesis are discussed. 98 of the over 5000 chemicals in tobacco smoke are known carcinogens that can act on cancer genes such as K-RAS and p53. Through various mechanisms these compounds can activate molecules involved in the cell cycle, such as cyclins, and molecules involved in apoptosis and autophagy, such as Beclin-1 or LC3B. A search of the literature, including in vitro and in vivo studies, was carried out and the results summarized. There is evidence of cancerogenic effects of cigarette smoke compounds. Cigarette smoke extract is a tobacco condensate obtained by filtration processes. Studies have shown that it can modify the cell cycle, inducing uncontrolled cell proliferation. This effect occurs through activation of genetic and epigenetic pathways and increasing the expression of proteins involved in inflammation. The pathways activated by cigarette smoke extract open up opportunities for researchers to develop new targeted therapies toward the specific molecules involved. Furthermore, the effects exerted by cigarette smoke extract on normal epithelial cells hold potential for use in the development of prevention medicine and early cancer diagnosis.
Collapse
Affiliation(s)
- Aldo Pezzuto
- Cardiovascular & Thoracic Department, AOU Sant'Andrea, Sapienza - Università di Roma, Roma, Italy
| | | | - Ivana Croghan
- Department of Medicine Clinical Research Office & Primary Care Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Giuseppe Tonini
- Oncology Department, Campus Bio-Medico Università di Roma, Roma, Italy
| |
Collapse
|
13
|
Yang DC, Chen CH. Cigarette Smoking-Mediated Macrophage Reprogramming: Mechanistic Insights and Therapeutic Implications. JOURNAL OF NATURE AND SCIENCE 2018; 4:e539. [PMID: 30801020 PMCID: PMC6383770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Macrophages, the mature form of the monocytes, play a significant role in tissue homeostasis and immunity. In response to environmental cues, they can undergo classical or alternative activation, polarizing into specialized functional subsets. A common hallmark of the pathologic environment is represented by cigarette smoking. Although the contribution of cigarette smoke to various cellular processes has been extensively studied, its roles in macrophage polarization have been conflicting. This review discusses the molecular and functional differences of cigarette smoke-exposed macrophages that exist between pro-inflammatory and anti-inflammatory states. We also highlight the most recent advances in therapeutic potential of targeting signaling molecules associated with smoking to modulate macrophage plasticity and polarized activation.
Collapse
Affiliation(s)
- David C Yang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine and Center for Comparative Respiratory Biology and Medicine, University of California Davis, Davis, California, USA
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, California, USA
| | - Ching-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, California, USA
- Comprehensive Cancer Center, University of California Davis, Davis, California, USA
| |
Collapse
|
14
|
Neuroepithelial control of mucosal inflammation in acute cystitis. Sci Rep 2018; 8:11015. [PMID: 30030504 PMCID: PMC6054610 DOI: 10.1038/s41598-018-28634-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/03/2018] [Indexed: 12/18/2022] Open
Abstract
The nervous system is engaged by infection, indirectly through inflammatory cascades or directly, by bacterial attack on nerve cells. Here we identify a neuro-epithelial activation loop that participates in the control of mucosal inflammation and pain in acute cystitis. We show that infection activates Neurokinin-1 receptor (NK1R) and Substance P (SP) expression in nerve cells and bladder epithelial cells in vitro and in vivo in the urinary bladder mucosa. Specific innate immune response genes regulated this mucosal response, and single gene deletions resulted either in protection (Tlr4−/− and Il1b−/− mice) or in accentuated bladder pathology (Asc−/− and Nlrp3−/− mice), compared to controls. NK1R/SP expression was lower in Tlr4−/− and Il1b−/− mice than in C56BL/6WT controls but in Asc−/− and Nlrp3−/− mice, NK1R over-activation accompanied the exaggerated disease phenotype, due, in part to transcriptional de-repression of Tacr1. Pharmacologic NK1R inhibitors attenuated acute cystitis in susceptible mice, supporting a role in disease pathogenesis. Clinical relevance was suggested by elevated urine SP levels in patients with acute cystitis, compared to patients with asymptomatic bacteriuria identifying NK1R/SP as potential therapeutic targets. We propose that NK1R and SP influence the severity of acute cystitis through a neuro-epithelial activation loop that controls pain and mucosal inflammation.
Collapse
|
15
|
Berg T, Hegelund-Myrbäck T, Öckinger J, Zhou XH, Brännström M, Hagemann-Jensen M, Werkström V, Seidegård J, Grunewald J, Nord M, Gustavsson L. Expression of MATE1, P-gp, OCTN1 and OCTN2, in epithelial and immune cells in the lung of COPD and healthy individuals. Respir Res 2018; 19:68. [PMID: 29678179 PMCID: PMC5910606 DOI: 10.1186/s12931-018-0760-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/27/2018] [Indexed: 02/03/2023] Open
Abstract
Background Several inhaled drugs are dependent on organic cation transporters to cross cell membranes. To further evaluate their potential to impact on inhaled drug disposition, the localization of MATE1, P-gp, OCTN1 and OCTN2 were investigated in human lung. Methods Transporter proteins were analysed by immunohistochemistry in lung tissue from healthy subjects and COPD patients. Transporter mRNA was analysed by qPCR in lung tissue and in bronchoalveolar lavage (BAL) cells from smokers and non-smokers. Results We demonstrate for the first time MATE1 protein expression in the lung with localization to the apical side of bronchial and bronchiolar epithelial cells. Interestingly, MATE1 was strongly expressed in alveolar macrophages as demonstrated both in lung tissue and in BAL cells, and in inflammatory cells including CD3 positive T cells. P-gp, OCTN1 and OCTN2 were also expressed in the alveolar epithelial cells and in inflammatory cells including alveolar macrophages. In BAL cells from smokers, MATE1 and P-gp mRNA expression was significantly lower compared to cells from non-smokers whereas no difference was observed between COPD patients and healthy subjects. THP-1 cells were evaluated as a model for alveolar macrophages but did not reflect the transporter expression observed in BAL cells. Conclusions We conclude that MATE1, P-gp, OCTN1 and OCTN2 are expressed in pulmonary lung epithelium, in alveolar macrophages and in other inflammatory cells. This is important to consider in the development of drugs treating pulmonary disease as the transporters may impact drug disposition in the lung and consequently affect pharmacological efficacy and toxicity. Electronic supplementary material The online version of this article (10.1186/s12931-018-0760-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tove Berg
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tove Hegelund-Myrbäck
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca R&D, Gothenburg, Sweden.
| | - Johan Öckinger
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xiao-Hong Zhou
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca R&D, Gothenburg, Sweden
| | - Marie Brännström
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca R&D, Gothenburg, Sweden
| | - Michael Hagemann-Jensen
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Viktoria Werkström
- Respiratory GMed, Global Medicines Development, AstraZeneca R&D, Gothenburg, Sweden
| | - Janeric Seidegård
- Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca R&D, Gothenburg, Sweden
| | - Johan Grunewald
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Nord
- Respiratory Medicine Unit, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Global Patient Safety, Global Medicines Development, AstraZeneca R&D, Gothenburg, Sweden
| | - Lena Gustavsson
- Department of Drug Metabolism, H. Lundbeck A/S, Ottiliavej 9, 2500, Valby, Denmark
| |
Collapse
|
16
|
Bruno A, Cipollina C, Di Vincenzo S, Siena L, Dino P, Di Gaudio F, Gjomarkaj M, Pace E. Ceftaroline modulates the innate immune and host defense responses of immunocompetent cells exposed to cigarette smoke. Toxicol Lett 2017; 279:9-15. [PMID: 28720485 DOI: 10.1016/j.toxlet.2017.07.878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/03/2017] [Accepted: 07/09/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cigarette smoke, the principal risk factor for chronic obstructive pulmonary disease (COPD), negatively influences the effectiveness of the immune system's response to a pathogen. The antibiotic ceftaroline exerts immune-modulatory effects in bronchial epithelial cells exposed to cigarette smoke. AIMS AND METHODS The present study aims to assess the effects of ceftaroline on TLR2 and TLR4 expression, LPS binding and TNF-α and human beta defensin (HBD2) release in an undifferentiated and PMA-differentiated human monocyte cell line (THP-1) exposed or not to cigarette smoke extracts (CSE). TLR2, TLR4, and LPS binding were assessed by flow cytometry, TNF-α and HBD2 release were evaluated by ELISA. RESULTS The constitutive expression of TLR2 and TLR4 and LPS binding were higher in differentiated compared to undifferentiated THP-1 cells. In undifferentiated THP-1 cells, CSE increased TLR2 and TLR4 protein levels, LPS binding and TNF-α release and reduced HBD2 release and ceftaroline counteracted all these effects. In differentiated THP-1, CSE did not significantly affect TLR2 and TLR4 expression and LPS binding but reduced HBD2 release and increased TNF-α release. Ceftaroline counteracted the effects of CSE on HBD2 release in differentiated THP-1. CONCLUSION Ceftaroline counteracts the effect of CSE in immune cells by increasing the effectiveness of the innate immune system. This effect may also assist in reducing pathogen activity and recurrent exacerbations in COPD patients.
Collapse
Affiliation(s)
- A Bruno
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - C Cipollina
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy; Fondazione Ri.MED, Palermo, Italy
| | - S Di Vincenzo
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - L Siena
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - P Dino
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - F Di Gaudio
- DiBiMeF (Biopatologia e Biotecnologie Mediche e Forensi), Università degli Studi di Palermo, Italy
| | - M Gjomarkaj
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - E Pace
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy.
| |
Collapse
|
17
|
Nennig SE, Schank JR. The Role of NFkB in Drug Addiction: Beyond Inflammation. Alcohol Alcohol 2017; 52:172-179. [PMID: 28043969 DOI: 10.1093/alcalc/agw098] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Indexed: 12/19/2022] Open
Abstract
Aims Nuclear factor kappa light chain enhancer of activated B cells (NFkB) is a ubiquitous transcription factor well known for its role in the innate immune response. As such, NFkB is a transcriptional activator of inflammatory mediators such as cytokines. It has recently been demonstrated that alcohol and other drugs of abuse can induce NFkB activity and cytokine expression in the brain. A number of reviews have been published highlighting this effect of alcohol, and have linked increased NFkB function to neuroimmune-stimulated toxicity. However, in this review we focus on the potentially non-immune functions of NFkB as possible links between NFkB and addiction. Methods An extensive review of the literature via Pubmed searches was used to assess the current state of the field. Results NFkB can induce the expression of a diverse set of gene targets besides inflammatory mediators, some of which are involved in addictive processes, such as opioid receptors and neuropeptides. NFkB mediates complex behaviors including learning and memory, stress responses, anhedonia and drug reward, processes that may lie outside the role of NFkB in the classic neuroimmune response. Conclusions Future studies should focus on these non-immune functions of NFkB signaling and their association with addiction-related processes.
Collapse
Affiliation(s)
- S E Nennig
- Department of Physiology and Pharmacology, University of Georgia, 501 D.W. Brooks Drive, Athens, GA 30602, USA
| | - J R Schank
- Department of Physiology and Pharmacology, University of Georgia, 501 D.W. Brooks Drive, Athens, GA 30602, USA
| |
Collapse
|
18
|
Muñoz M, Coveñas R. Targeting NK-1 Receptors to Prevent and Treat Pancreatic Cancer: a New Therapeutic Approach. Cancers (Basel) 2015; 7:1215-32. [PMID: 26154566 PMCID: PMC4586765 DOI: 10.3390/cancers7030832] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/27/2015] [Accepted: 06/30/2015] [Indexed: 12/28/2022] Open
Abstract
Pancreatic cancer (PC) is the fourth leading cause of cancer related-deaths in both men and women, and the 1- and 5-year relative survival rates are 25% and 6%, respectively. It is known that smoking, alcoholism and psychological stress are risk factors that can promote PC and increase PC progression. To date, the prevention of PC is crucial because there is no curative treatment. After binding to the neurokinin-1 (NK-1) receptor (a receptor coupled to the stimulatory G-protein Gαs that activates adenylate cyclase), the peptide substance P (SP)-at high concentrations-is involved in many pathophysiological functions, such as depression, smoking, alcoholism, chronic inflammation and cancer. It is known that PC cells and samples express NK-1 receptors; that the NK-1 receptor is overexpressed in PC cells in comparison with non-tumor cells, and that nanomolar concentrations of SP induce PC cell proliferation. By contrast, NK-1 receptor antagonists exert antidepressive, anxiolytic and anti-inflammatory effects and anti-alcohol addiction. These antagonists also exert An antitumor action since in vitro they inhibit PC cell proliferation (PC cells death by apoptosis), and in a xenograft PC mouse model they exert both antitumor and anti-angiogenic actions. NK-1 receptor antagonists could be used for the treatment of PC and hence the NK-1 receptor could be a new promising therapeutic target in PC.
Collapse
Affiliation(s)
- Miguel Muñoz
- Research Laboratory on Neuropeptides (IBIS), Virgen del Rocío University Hospital,41013 Sevilla, Spain.
| | - Rafael Coveñas
- Laboratory of Neuroanatomy of the Peptidergic System (Lab. 14), Institute of Neurosciences ofCastilla y León (INCYL), University of Salamanca, 37008 Salamanca, Spain.
| |
Collapse
|
19
|
Novel drug targets for asthma and COPD: lessons learned from in vitro and in vivo models. Pulm Pharmacol Ther 2014; 29:181-98. [PMID: 24929072 DOI: 10.1016/j.pupt.2014.05.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 05/20/2014] [Accepted: 05/31/2014] [Indexed: 12/28/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are highly prevalent respiratory diseases characterized by airway inflammation, airway obstruction and airway hyperresponsiveness. Whilst current therapies, such as β-agonists and glucocorticoids, may be effective at reducing symptoms, they do not reduce disease progression. Thus, there is a need to identify new therapeutic targets. In this review, we summarize the potential of novel targets or tools, including anti-inflammatories, phosphodiesterase inhibitors, kinase inhibitors, transient receptor potential channels, vitamin D and protease inhibitors, for the treatment of asthma and COPD.
Collapse
|
20
|
Giunzioni I, Bonomo A, Bishop E, Castiglioni S, Corsini A, Bellosta S. Cigarette smoke condensate affects monocyte interaction with endothelium. Atherosclerosis 2014; 234:383-90. [PMID: 24747113 DOI: 10.1016/j.atherosclerosis.2014.03.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 03/12/2014] [Accepted: 03/24/2014] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Circulating monocytes adhere to the endothelium and migrate into the intima contributing to atherosclerotic plaque growth. Cigarette smoke is a risk factor for atherosclerosis, but it is not completely known how it affects monocyte behavior in atherogenesis. METHODS We studied the effects of cigarette smoke condensate (CSC) on human monocytes (HM) chemotaxis and transmigration through an endothelial cell (EC) monolayer. RESULTS Pre-treatment with CSC caused a decrease in HM chemotaxis and transmigration (-55% and -18% vs control, p < 0.05, respectively), paralleled by a reduced expression of Rac 1 GTPase. On the contrary, direct exposure of both HM and EC to CSC increased (+23% vs control, p < 0.05) HM transmigration, paralleled by a strong stimulation of VCAM1 and ICAM1 expression by ECs, and by a slight increase in monocyte integrin expression. An enhancement of monocyte transmigration was obtained after the exposure of both HM and EC to medium conditioned by HM previously incubated with CSC (+265% vs control, p < 0.001). CSC showed a stimulatory effect on the expression by HM of TLR4, MCP1, IL8, IL1beta, and TNFalfa, which was ablated by pre treatment with PDTC. Incubation with neutralizing antibodies against both MCP1 or IL8 completely abolished the CSC-conditioned medium induced HM transmigration. CONCLUSIONS CSC induces HM to release chemotactic factor(s), which amplify the recruitment and transmigration of inflammatory cells through EC, but CSC may also reduce HM migratory capacity. Therefore, exposure to CSC affects monocyte behavior and interaction with the endothelium, thus potentially facilitating and/or further aggravating the atherogenic process.
Collapse
Affiliation(s)
- I Giunzioni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - A Bonomo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - E Bishop
- British American Tobacco Group Research & Development, Southampton, UK
| | - S Castiglioni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - A Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - S Bellosta
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy.
| |
Collapse
|
21
|
Šmerdová L, Neča J, Svobodová J, Topinka J, Schmuczerová J, Kozubík A, Machala M, Vondráček J. Inflammatory mediators accelerate metabolism of benzo[a]pyrene in rat alveolar type II cells: The role of enhanced cytochrome P450 1B1 expression. Toxicology 2013; 314:30-8. [DOI: 10.1016/j.tox.2013.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 08/29/2013] [Accepted: 09/02/2013] [Indexed: 12/15/2022]
|
22
|
Melnik BC, Plewig G. Impaired Notch-MKP-1 signalling in hidradenitis suppurativa: an approach to pathogenesis by evidence from translational biology. Exp Dermatol 2013; 22:172-7. [PMID: 23489419 DOI: 10.1111/exd.12098] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2013] [Indexed: 12/13/2022]
Abstract
Recent findings in familial hidradenitis suppurativa (HS) demonstrated loss-of-function mutations of components of the γ-secretase (GS) complex leading to decreased protease cleaving activity, which may compromise canonical Notch signalling. Appropriate Notch signalling is of pivotal importance for maintaining the inner and outer root sheath of the hair follicle and skin appendages. This viewpoint on the pathogenesis of HS is primarily supported by circumstantial evidence derived from translational biology. Impaired Notch signalling is proposed to be the major pathogenic mechanism of HS. Deficient Notch signalling switches the fate of outer root sheath cells, resulting in conversion of hair follicles to keratin-enriched epidermal cysts. Impaired Notch signalling may compromise apocrine gland homoeostasis as well. Damage-associated molecular pattern molecules released by either ruptured epidermal cysts exposing keratin fibres or altered structural components of less maintained apocrine glands may both stimulate TLR-mediated innate immunity. All aggravating factors of HS, that is, smoking, obesity, skin occlusion, androgens and progesterone, may further promote inflammation by release of proinflammatory cytokines derived from activated monocyte/macrophages. Inappropriate Notch signalling may not only initiate inflammation in HS but may lead to insufficient feedback inhibition of overstimulated innate immunity. Regular Notch signalling via induction of MAPK phosphatase-1 (MKP-1) terminates TLR-MAPK-signalling in macrophages and IL-23 secreting DCs, the key players for Th17 cell polarization. Thus, impaired Notch signalling links HS to other Th17-driven comorbidities. All major therapeutic interventions in HS appear to attenuate increased MAPK activation of innate immune cells due to impaired Notch-mediated feedback regulation of innate immunity.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Osnabrück, Germany.
| | | |
Collapse
|
23
|
Campesi I, Carru C, Zinellu A, Occhioni S, Sanna M, Palermo M, Tonolo G, Mercuro G, Franconi F. Regular cigarette smoking influences the transsulfuration pathway, endothelial function, and inflammation biomarkers in a sex-gender specific manner in healthy young humans. Am J Transl Res 2013; 5:497-509. [PMID: 23977409 PMCID: PMC3745437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/09/2013] [Indexed: 06/02/2023]
Abstract
Cigarette smoking (CS) is the primary cause of preventable morbidity and mortality. Abundant clinical evidence suggests that CS is more harmful to women; however, the mechanisms responsible for these differences are not yet known. CS alters endothelial function, the redox state, inflammation, and global DNA methylation, which is associated with one-carbon metabolism and the transsulfuration pathway. However, it is not known whether the previously identified alterations are sex-gender related. Healthy adult men and oral contraceptive-free women with regular menstrual cycles were enrolled; women were examined during the follicular phase. Men had higher plasma levels of uric acid, total bilirubin, homocysteine, glutamylcysteine, total glutathione, cysteinylglycine; had more monocytes and released more TNF-alpha from human monocytes derived macrophages (hMDMs), but they had fewer platelets and lower levels of DNA methylation, and their hMDMs released less TNF-alpha after LPS stimulation. MDA, taurine, cysteine, arginine, ADMA, and SDMA were not different. CS decreased global DNA methylation more in women and increased the platelet, monocyte, and lymphocyte counts and the homocysteine, arginine, and ADMA levels only in women, whereas increased the neutrophil and eosinophil counts only in men. Additionally, CS reduced the sex-gender differences in total bilirubin, basal and LPS-induced TNF-alpha release, total glutathione, and glutamylcysteine, leaving unchanged cysteinylglycine, taurine, SDMA, MDA, and cysteine. These data suggest that cardiovascular risk factors seem to come earlier in young healthy female smokers than in young healthy male smokers, supporting the greater alarmism regarding the effects of CS in women and providing a basis for understanding the sex-gender differences. These results also suggest that cessation programs targeting women are needed.
Collapse
Affiliation(s)
- Ilaria Campesi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and BiosystemsViale S. Antonio, Osilo, Italy
- Department of Biomedical Sciences, University of SassariVia Muroni 23, Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of SassariVia Muroni 23, Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of SassariVia Muroni 23, Sassari, Italy
| | - Stefano Occhioni
- Department of Biomedical Sciences, University of SassariVia Muroni 23, Sassari, Italy
| | - Manuela Sanna
- Department of Biomedical Sciences, University of SassariVia Muroni 23, Sassari, Italy
| | - Mario Palermo
- Servizio di Diagnosi e Cura di Endocrinologia, Azienda Ospedaliero-UniversitariaSassari, Italy
| | - Giancarlo Tonolo
- SC Diabetologia Aziendale ASL 2 Olbia, Hospital San Giovanni di DioOlbia, Italy
| | | | - Flavia Franconi
- Laboratory of Sex-Gender Medicine, National Institute of Biostructures and BiosystemsViale S. Antonio, Osilo, Italy
- Department of Biomedical Sciences, University of SassariVia Muroni 23, Sassari, Italy
| |
Collapse
|
24
|
Okwan-Duodu D, Landry J, Shen XZ, Diaz R. Angiotensin-converting enzyme and the tumor microenvironment: mechanisms beyond angiogenesis. Am J Physiol Regul Integr Comp Physiol 2013; 305:R205-15. [DOI: 10.1152/ajpregu.00544.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The renin angiotensin system (RAS) is a network of enzymes and peptides that coalesce primarily on the angiotensin II type 1 receptor (AT1R) to induce cell proliferation, angiogenesis, fibrosis, and blood pressure control. Angiotensin-converting enzyme (ACE), the key peptidase of the RAS, is promiscuous in that it cleaves other substrates such as substance P and bradykinin. Accumulating evidence implicates ACE in the pathophysiology of carcinogenesis. While the role of ACE and its peptide network in modulating angiogenesis via the AT1R is well documented, its involvement in shaping other aspects of the tumor microenvironment remains largely unknown. Here, we review the role of ACE in modulating the immune compartment of the tumor microenvironment, which encompasses the immunosuppressive, cancer-promoting myeloid-derived suppressor cells, alternatively activated tumor-associated macrophages, and T regulatory cells. We also discuss the potential roles of peptides that accumulate in the setting of chronic ACE inhibitor use, such as bradykinin, substance P, and N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP), and how they may undercut the gains of anti-angiogenesis from ACE inhibition. These emerging mechanisms may harmonize the often-conflicting results on the role of ACE inhibitors and ACE polymorphisms in various cancers and call for further investigations into the potential benefit of ACE inhibitors in some neoplasms.
Collapse
Affiliation(s)
- Derick Okwan-Duodu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia; and
| | - Jerome Landry
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia; and
| | - Xiao Z. Shen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Roberto Diaz
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia; and
| |
Collapse
|
25
|
Pierson T, Learmonth-Pierson S, Pinto D, van Hoek ML. Cigarette smoke extract induces differential expression levels of beta-defensin peptides in human alveolar epithelial cells. Tob Induc Dis 2013; 11:10. [PMID: 23627872 PMCID: PMC3648470 DOI: 10.1186/1617-9625-11-10] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/12/2013] [Indexed: 12/16/2022] Open
Abstract
Background The damaging effects of cigarette smoke on the lungs are well known in terms of cancer risks. Additional molecular changes within the lung tissue can also occur as a result of exposure to cigarette smoke. The human β-defensin (hBD) class of antimicrobial peptides is the focus of our research. In addition to antimicrobial activity, β-defensins also have immunomodulatory functions. Over 30 previously unrecognized β-defensin genes have recently been identified in the human genome, many with yet to be determined functions. We postulated that altered β-defensin production may play a role in the pathogenesis observed in the lungs of smokers. Our hypothesis is that cigarette smoke exposure will affect the expression of β-defensins in human lung alveolar epithelial cells (A549). Methods We exposed A549 cells to cigarette smoke extract (CSE) and measured the changes in mRNA levels of several antimicrobial peptides by quantitative real-time PCR, and directly observed peptide expression in cells by immunofluorescence (IF) microscopy. Results We found that hBD3, hBD5, and hBD9 gene expression was upregulated in A549 cells exposed to CSE. HBD1, hBD8, hBD18 and LL-37 gene expression did not significantly change upon exposure to CSE. Expression of hBD3 and hBD4 peptides was visualized by IF. Conclusions This differential expression suggests that hBD3, hBD5, and hBD9 may play a role in the changes to the lung tissue observed in smokers. Establishing differential β-defensin expression following CSE treatment will add to our understanding of the molecular response of the lung alveolar epithelium to cigarette smoke exposure.
Collapse
Affiliation(s)
- Tony Pierson
- School of Systems Biology, George Mason University, Manassas, VA, USA.
| | | | | | | |
Collapse
|
26
|
Van Scott MR, Chandler J, Olmstead S, Brown JM, Mannie M. Airway Anatomy, Physiology, and Inflammation. THE TOXICANT INDUCTION OF IRRITANT ASTHMA, RHINITIS, AND RELATED CONDITIONS 2013. [PMCID: PMC7122617 DOI: 10.1007/978-1-4614-9044-9_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
27
|
Arimilli S, Damratoski BE, Bombick B, Borgerding MF, Prasad G. Evaluation of cytotoxicity of different tobacco product preparations. Regul Toxicol Pharmacol 2012; 64:350-60. [DOI: 10.1016/j.yrtph.2012.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 08/20/2012] [Accepted: 09/11/2012] [Indexed: 01/01/2023]
|
28
|
Nagy A, Zane A, Cole SL, Severance M, Dutta PK, Waldman WJ. Contrast of the biological activity of negatively and positively charged microwave synthesized CdSe/ZnS quantum dots. Chem Res Toxicol 2011; 24:2176-88. [PMID: 22092015 DOI: 10.1021/tx2003195] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Quantum dots (QDs) are semiconductor nanocrystals that have found use in bioimaging, cell tracking, and drug delivery. This article compares the cytotoxicity and cellular interactions of positively and negatively charged CdSe/CdS/ZnS QDs prepared by a microwave method using a murine alveolar macrophage-like cell culture model. Keeping the core semiconductor the same, QD charge was varied by altering the surface capping molecule; negatively charged QDs were formed with mercaptopropionic acid (MPA-QDs) and positively charged QDs with thiocholine (THIO-QDs). The size and charge of these two QDs were investigated in three types of media (RPMI, RPMI + FBS, and X-VIVO serum-free media) relevant for the biological studies. MPA-QDs were found to have negative zeta potential in RPMI, RPMI + FBS, and serum-free media and had sizes ranging from 8 to 54 nm. THIO-QDs suspended in RPMI alone were <62 nm in size, while large aggregates (greater than 1000 nm) formed when these QDs were suspended in RPMI + FBS and serum-free media. THIO-QDs retained positive zeta potential in RPMI and were found to have a negative zeta potential in RPMI + FBS and nearly neutral zeta potential in serum-free media. In a cell culture model, both MPA-QDs and THIO-QDs caused comparable levels of apoptosis and necrosis. Both QDs induced significant tumor necrosis factor-alpha (TNF-α) secretion only at high concentrations (>250 nM). Both types of QDs were internalized via clathrin-dependent endocytosis. Using real-time, live cell imaging, we found that MPA-QDs interact with the cell surface within minutes and progress through the endocytic pathway to the lysosomes upon internalization. With the THIO-QDs, the internalization process was slower, but the pathways could not be mapped because of spectroscopic interference caused by QD aggregates. Finally, MPA-QDs were found to associate with cell surface scavenger receptors, while the THIO-QDs did not. This study indicates that the surface charge and aggregation characteristics of QDs change drastically in biological culture conditions and, in turn, influence nanoparticle and cellular interactions.
Collapse
Affiliation(s)
- Amber Nagy
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | | | |
Collapse
|
29
|
Haspel JA, Choi AMK. Autophagy: a core cellular process with emerging links to pulmonary disease. Am J Respir Crit Care Med 2011; 184:1237-46. [PMID: 21836133 DOI: 10.1164/rccm.201106-0966ci] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Autophagy is a highly conserved homeostatic pathway by which cells transport damaged proteins and organelles to lysosomes for degradation. Dysregulation of autophagy contributes to the pathogenesis of clinically important disorders in a variety of organ systems but, until recently, little was known about its relationship to diseases of the lung. However, there is now growing evidence at the basic research level that autophagy is linked to the pathogenesis of important pulmonary disorders such as chronic obstructive pulmonary disease, cystic fibrosis, and tuberculosis. In this review, we provide an introduction to the field of autophagy research geared to clinical and research pulmonologists. We focus on the best-studied autophagic mechanism, macroautophagy, and summarize studies that link the regulation of this pathway to pulmonary disease. Last, we offer our perspective on how a better understanding of macroautophagy might be used for designing novel therapies for pulmonary disorders.
Collapse
Affiliation(s)
- Jeffrey A Haspel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | |
Collapse
|