51
|
Yamaguchi Y, Watanabe Y, Watanabe T, Komitsu N, Aihara M. Decreased Expression of Caveolin-1 Contributes to the Pathogenesis of Psoriasiform Dermatitis in Mice. J Invest Dermatol 2015; 135:2764-2774. [PMID: 26134945 DOI: 10.1038/jid.2015.249] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 06/02/2015] [Accepted: 06/12/2015] [Indexed: 02/08/2023]
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by excessive proliferation and abnormal keratinocyte development, in which T helper type 17 cells and signal transducer and activator of transcription 3 (STAT3) activation have pivotal roles. Moreover, caveolin-1 (CAV-1) has been implicated in the regulation of signal transduction, and aberrant CAV-1 expression is involved in a variety of diseases. However, whether CAV-1 is involved in psoriasis is unknown. Here we examined CAV-1 expression in the psoriatic epidermis and investigated its role in the pathogenesis of psoriasis. CAV-1 was markedly reduced in lesional epidermis of psoriasis patients. CAV1 silencing in keratinocytes in vitro revealed significant activation of STAT3, leading to increased expression of keratin 16 and several cytokine/chemokines, such as IL-6, C-X-C chemokine ligand 8 (CXCL8), CXCL9, and C-C chemokine ligand 20. In addition, psoriasis-related cytokines, including tumor necrosis factor-α (TNF-α), decreased CAV-1 expression in keratinocytes. Finally, administration of CAV-1 scaffolding domain peptide in a murine model of psoriasis-like skin inflammation induced by imiquimod improved the skin phenotype and reduced epidermal thickness and infiltrating cell counts. Furthermore, expression of TNF-α, IL-17A, and IL-23 was significantly suppressed by this treatment. Collectively, our study indicated that CAV-1 participates in the pathogenesis of psoriasis by regulating the STAT3 pathway and cytokine networks.
Collapse
Affiliation(s)
- Yukie Yamaguchi
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Yuko Watanabe
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tomoya Watanabe
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Noriko Komitsu
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Michiko Aihara
- Department of Environmental Immuno-Dermatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
52
|
Transient Receptor Potential Channel 1 Deficiency Impairs Host Defense and Proinflammatory Responses to Bacterial Infection by Regulating Protein Kinase Cα Signaling. Mol Cell Biol 2015; 35:2729-39. [PMID: 26031335 DOI: 10.1128/mcb.00256-15] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/19/2015] [Indexed: 02/05/2023] Open
Abstract
Transient receptor potential channel 1 (TRPC1) is a nonselective cation channel that is required for Ca(2+) homeostasis necessary for cellular functions. However, whether TRPC1 is involved in infectious disease remains unknown. Here, we report a novel function for TRPC1 in host defense against Gram-negative bacteria. TRPC1(-/-) mice exhibited decreased survival, severe lung injury, and systemic bacterial dissemination upon infection. Furthermore, silencing of TRPC1 showed decreased Ca(2+) entry, reduced proinflammatory cytokines, and lowered bacterial clearance. Importantly, TRPC1 functioned as an endogenous Ca(2+) entry channel critical for proinflammatory cytokine production in both alveolar macrophages and epithelial cells. We further identified that bacterium-mediated activation of TRPC1 was dependent on Toll-like receptor 4 (TLR4), which induced endoplasmic reticulum (ER) store depletion. After activation of phospholipase Cγ (PLC-γ), TRPC1 mediated Ca(2+) entry and triggered protein kinase Cα (PKCα) activity to facilitate nuclear translocation of NF-κB/Jun N-terminal protein kinase (JNK) and augment the proinflammatory response, leading to tissue damage and eventually mortality. These findings reveal that TRPC1 is required for host defense against bacterial infections through the TLR4-TRPC1-PKCα signaling circuit.
Collapse
|
53
|
Ye Y, Tan S, Zhou X, Li X, Jundt MC, Lichter N, Hidebrand A, Dhasarathy A, Wu M. Inhibition of p-IκBα Ubiquitylation by Autophagy-Related Gene 7 to Regulate Inflammatory Responses to Bacterial Infection. J Infect Dis 2015; 212:1816-26. [PMID: 26022442 DOI: 10.1093/infdis/jiv301] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/14/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Klebsiella pneumoniae causes serious infections and healthcare burdens in humans. We have previously reported that the deficiency of autophagy-related gene (Atg) 7 in macrophages (murine alveolar macrophage cell line [MH-S]) induced irregular host immunity against K. pneumoniae and worsened pathologic effects in the lung. In the current study, we investigated the molecular mechanism by which Atg7 influenced K. pneumoniae-induced inflammatory responses. METHODS Expression levels of Atg7, ubiquitin (Ub), and tumor necrosis factor (TNF) α and phosphorylation of IκBα (p-IκBα) were determined with immunoblotting. Ubiquitylation of p-IκBα was determined with immunoprecipitation. RESULTS We noted an interaction between Atg7 and p-IκBα, which was decreased in MH-S after K. pneumoniae infection, whereas the interaction between Ub and p-IκBα was increased. Knock-down of Atg7 with small interfering RNA increased p-IκBα ubiquitylation, promoted nuclear factor κB translocation into the nucleus, and increased the production of TNF-α. Moreover, knock-down of Ub with lentivirus-short hairpin RNA Ub particles decreased binding of p-IκBα to Ub and inhibited TNF-α expression in the primary alveolar macrophages and lung tissue of atg7-knockout mice on K. pneumoniae infection. CONCLUSIONS Loss of Atg7 switched binding of p-IκBα from Atg7 to Ub, resulting in increased ubiquitylation of p-IκBα and intensified inflammatory responses against K. pneumoniae. Our findings not only reveal a regulatory role of Atg7 in ubiquitylation of p-IκBα but also indicate potential therapeutic targets for K. pneumoniae control.
Collapse
Affiliation(s)
- Yan Ye
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks
| | - Shirui Tan
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming
| | - Xikun Zhou
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Xuefeng Li
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Michael C Jundt
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks
| | - Natalie Lichter
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks
| | - Alec Hidebrand
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks
| | - Archana Dhasarathy
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks
| | - Min Wu
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks
| |
Collapse
|
54
|
Feng Q, Gao N. Keeping Wnt signalosome in check by vesicular traffic. J Cell Physiol 2015; 230:1170-80. [PMID: 25336320 DOI: 10.1002/jcp.24853] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 10/17/2014] [Indexed: 01/01/2023]
Abstract
Wg/Wnts are paracrine and autocrine ligands that activate distinct signaling pathways while being internalized through surface receptors. Converging and contrasting views are shaping our understanding of whether, where, and how endocytosis may modulate Wnt signaling. We gather considerable amount of evidences to elaborate the point that signal-receiving cells utilize distinct, flexible, and sophisticated vesicular trafficking mechanisms to keep Wnt signaling activity in check. Same molecules in a highly context-dependent fashion serve as regulatory hub for various signaling purposes: amplification, maintenance, inhibition, and termination. Updates are provided for the regulatory mechanisms related to the three critical cell surface complexes, Wnt-Fzd-LRP6, Dkk1-Kremen-LRP6, and R-spondin-LGR5-RNF43, which potently influence Wnt signaling. We pay particular attentions to how cells achieve sustained and delicate control of Wnt signaling strength by employing comprehensive aspects of vesicular trafficking.
Collapse
Affiliation(s)
- Qiang Feng
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | | |
Collapse
|
55
|
Could caveolae be acting as warnings of mitochondrial ageing? Mech Ageing Dev 2015; 146-148:81-7. [PMID: 25959712 DOI: 10.1016/j.mad.2015.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 04/09/2015] [Accepted: 04/28/2015] [Indexed: 11/20/2022]
Abstract
Ageing is a cellular process with many facets, some of which are currently undergoing a paradigm change. It is the case of "mitochondrial theory of ageing", which, interestingly, has been found lately to cross paths with another ageing dysfunctional process - intracellular signalling - in an unexpected point (or place) - caveolae. The latter represent membrane microdomains altered in senescent cells, scaffolded by proteins modified (posttranslational or as expression) with ageing. An important determinant of these alterations is oxidative stress, through increased production of reactive oxygen species that originate at mitochondrial site. Spanning from physical contact points, to shared structural proteins and similar function domains, caveolae and mitochondria might have more in common than originally thought. By reviewing recent data on oxidative stress impact on caveolae and caveolins, as well as possible interactions between caveolae and mitochondria, we propose a hypothesis for senescence-related involvement of caveolins.
Collapse
|
56
|
Yu Q, Chen X, Fang X, Chen Q, Hu C. Caveolin-1 aggravates cigarette smoke extract-induced MUC5AC secretion in human airway epithelial cells. Int J Mol Med 2015; 35:1435-42. [PMID: 25776934 DOI: 10.3892/ijmm.2015.2133] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/04/2015] [Indexed: 11/06/2022] Open
Abstract
Airway mucus hypersecretion is a major pathological characteristic of chronic obstructive pulmonary disease (COPD), and cigarette smoke is highly implicated in mucus secretion and the development of COPD. Cigarette smoke reportedly induces mucin overproduction through the epidermal growth factor receptor (EGFR) in the airway epithelium; however, the underlying mechanisms responsible for the activation of EGFR remain unknown. Caveolin-1, a component protein in the cytomembrane, reportedly regulates airway inflammation and lung injury. In this study, we aimed to determine whether caveolin-1 modulates mucin hyperproduction induced by cigarette smoke. Our results revealed that cigarette smoke extract (CSE) significantly increased MUC5AC production, as well as the levels of phosphorylated EGFR (p-EGFR) and phosphorylated Akt (p-Akt) in human bronchial epithelial cells (16HBE cells), as shown by ELISA, RT-PCR and western blot analysis. These effects were prevented by treatment with EGFR inhibitor (AG1478) and phosphatidylinostol-3-kinase (PI3K) inhibitor (LY294002). We also found that the overexpression of caveolin-1 enhanced the expression of MUC5AC, p-EGFR and p-Akt induced by CSE. Conversely, the downregulation of caveolin-1 by siRNA against caveolin-1 inhibited the expression of MUC5AC, p-EGFR and p-Akt. Taken together, our data suggest that caveolin-1 enhances CSE-induced MUC5AC hypersecretion through the EGFR/PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Qiao Yu
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xi Chen
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xia Fang
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qiong Chen
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Chengping Hu
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
57
|
Zhang PX, Cheng J, Zou S, D’Souza AD, Koff JL, Lu J, Lee PJ, Krause DS, Egan ME, Bruscia EM. Pharmacological modulation of the AKT/microRNA-199a-5p/CAV1 pathway ameliorates cystic fibrosis lung hyper-inflammation. Nat Commun 2015; 6:6221. [PMID: 25665524 PMCID: PMC4324503 DOI: 10.1038/ncomms7221] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 01/07/2015] [Indexed: 12/12/2022] Open
Abstract
In cystic fibrosis (CF) patients, hyper-inflammation is a key factor in lung destruction and disease morbidity. We have previously demonstrated that macrophages drive the lung hyper-inflammatory response to LPS in CF mice, because of reduced levels of the scaffold protein CAV1 with subsequent uncontrolled TLR4 signalling. Here we show that reduced CAV1 and, consequently, increased TLR4 signalling, in human and murine CF macrophages and murine CF lungs, is caused by high microRNA-199a-5p levels, which are PI3K/AKT-dependent. Downregulation of microRNA-199a-5p or increased AKT signalling restores CAV1 expression and reduces hyper-inflammation in CF macrophages. Importantly, the FDA-approved drug celecoxib re-establishes the AKT/miR-199a-5p/CAV1 axis in CF macrophages, and ameliorates lung hyper-inflammation in Cftr-deficient mice. Thus, we identify the AKT/miR-199a-5p/CAV1 pathway as a regulator of innate immunity, which is dysfunctional in CF macrophages contributing to lung hyper-inflammation. In addition, we found that this pathway can be targeted by celecoxib.
Collapse
Affiliation(s)
- Ping-xia Zhang
- Department of Pediatrics, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
- Department of Laboratory Medicine, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| | - Jijun Cheng
- Department of Genetics, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
- Yale Stem Cell Center, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
- Yale Cancer Center, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| | - Siying Zou
- Department of Cell Biology, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| | - Anthony D. D’Souza
- Department of Laboratory Medicine, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| | - Jonathan L. Koff
- Department of Pulmonary, Critical Care and Sleep Medicine, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| | - Jun Lu
- Department of Genetics, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
- Yale Stem Cell Center, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
- Yale Cancer Center, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| | - Patty J. Lee
- Department of Pulmonary, Critical Care and Sleep Medicine, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| | - Diane S. Krause
- Department of Laboratory Medicine, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
- Yale Stem Cell Center, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
- Yale Cancer Center, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
- Department of Cell Biology, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| | - Marie E. Egan
- Department of Pediatrics, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
- Department of Cellular and Molecular Physiology, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| | - Emanuela M. Bruscia
- Department of Pediatrics, 333 Cedar Street, Yale University School of Medicine, New Haven Connecticut, USA
| |
Collapse
|
58
|
Tan S, Gan C, Li R, Ye Y, Zhang S, Wu X, Yang YY, Fan W, Wu M. A novel chemosynthetic peptide with β-sheet motif efficiently kills Klebsiella pneumoniae in a mouse model. Int J Nanomedicine 2015; 10:1045-59. [PMID: 25709431 PMCID: PMC4330034 DOI: 10.2147/ijn.s73303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Klebsiella pneumoniae (Kp) is one of the most common pathogens in nosocomial infections and is increasingly becoming multiple drug resistant. However, the molecular pathogenesis of Kp in causing tissue injury and dysregulated host defense remains elusive, further dampening the development of novel therapeutic measures. We have previously screened a series of synthetic antimicrobial beta-sheet forming peptides and identified a peptide (IRIKIRIK; ie, IK8L) with a broad range of bactericidal activity and low cytotoxicity in vitro. Here, employing an animal model, we investigated the antibacterial effects of IK8L in acute infection and demonstrated that peritoneal injection of IK8L to mice down-regulated inflammatory cytokines, alleviated lung injury, and importantly, decreased mortality compared to sham-injected controls. In addition, a math model was used to evaluate in vivo imaging data and predict infection progression in infected live animals. Mechanistically, IK8L can kill Kp by inhibiting biofilm formation and modulating production of inflammatory cytokines through the STAT3/JAK signaling both in vitro and in vivo. Collectively, these findings reveal that IK8L may have potential for preventing or treating Kp infection.
Collapse
Affiliation(s)
- Shirui Tan
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA ; Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
| | - Changpei Gan
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA ; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Rongpeng Li
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA
| | - Yan Ye
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA
| | - Shuang Zhang
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA ; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xu Wu
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, The Nanos, Singapore
| | - Weimin Fan
- Program of Innovative Cancer Therapeutics, First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, People's Republic of China
| | - Min Wu
- Department of Basic Sciences, School of Medicine and Health Sciences University of North Dakota, Grand Forks, ND, USA
| |
Collapse
|
59
|
Li X, Ye Y, Zhou X, Huang C, Wu M. Atg7 enhances host defense against infection via downregulation of superoxide but upregulation of nitric oxide. THE JOURNAL OF IMMUNOLOGY 2014; 194:1112-21. [PMID: 25535282 DOI: 10.4049/jimmunol.1401958] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic bacterium that can cause serious infection in immunocompromised individuals. Although autophagy may augment immune responses against P. aeruginosa infection in macrophages, the critical components and their role of autophagy in host defense are largely unknown. In this study, we show that P. aeruginosa infection-induced autophagy activates JAK2/STAT1α and increases NO production. Knocking down Atg7 resulted in increased IFN-γ release, excessive reactive oxygen species, and increased Src homology-2 domain-containing phosphatase 2 activity, which led to lowered phosphorylation of JAK2/STAT1α and subdued expression of NO synthase 2 (NOS2). In addition, we demonstrated the physiological relevance of dysregulated NO under Atg7 deficiency as atg7(-/-) mice were more susceptible to P. aeruginosa infection with increased mortality and severe lung injury than wild-type mice. Furthermore, P. aeruginosa-infected atg7(-/-) mice exhibited increased oxidation but decreased bacterial clearance in the lung and other organs compared with wild-type mice. Mechanistically, atg7 deficiency suppressed NOS2 activity by downmodulating JAK2/STAT1α, leading to decreased NO both in vitro and in vivo. Taken together, these findings revealed that the JAK2/STAT1α/NOS2 dysfunction leads to dysregulated immune responses and worsened disease phenotypes.
Collapse
Affiliation(s)
- Xuefeng Li
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203; and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yan Ye
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203; and
| | - Xikun Zhou
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203; and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Canhua Huang
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Min Wu
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203; and
| |
Collapse
|
60
|
Cao Q, Wang Y, Chen F, Xia Y, Lou J, Zhang X, Yang N, Sun X, Zhang Q, Zhuo C, Huang X, Deng X, Yang CG, Ye Y, Zhao J, Wu M, Lan L. A novel signal transduction pathway that modulates rhl quorum sensing and bacterial virulence in Pseudomonas aeruginosa. PLoS Pathog 2014; 10:e1004340. [PMID: 25166864 PMCID: PMC4148453 DOI: 10.1371/journal.ppat.1004340] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/15/2014] [Indexed: 01/10/2023] Open
Abstract
The rhl quorum-sensing (QS) system plays critical roles in the pathogenesis of P. aeruginosa. However, the regulatory effects that occur directly upstream of the rhl QS system are poorly understood. Here, we show that deletion of gene encoding for the two-component sensor BfmS leads to the activation of its cognate response regulator BfmR, which in turn directly binds to the promoter and decreases the expression of the rhlR gene that encodes the QS regulator RhlR, causing the inhibition of the rhl QS system. In the absence of bfmS, the Acka-Pta pathway can modulate the regulatory activity of BfmR. In addition, BfmS tunes the expression of 202 genes that comprise 3.6% of the P. aeruginosa genome. We further demonstrate that deletion of bfmS causes substantially reduced virulence in lettuce leaf, reduced cytotoxicity, enhanced invasion, and reduced bacterial survival during acute mouse lung infection. Intriguingly, specific missense mutations, which occur naturally in the bfmS gene in P. aeruginosa cystic fibrosis (CF) isolates such as DK2 strains and RP73 strain, can produce BfmS variants (BfmSL181P, BfmSL181P/E376Q, and BfmSR393H) that no longer repress, but instead activate BfmR. As a result, BfmS variants, but not the wild-type BfmS, inhibit the rhl QS system. This study thus uncovers a previously unexplored signal transduction pathway, BfmS/BfmR/RhlR, for the regulation of rhl QS in P. aeruginosa. We propose that BfmRS TCS may have an important role in the regulation and evolution of P. aeruginosa virulence during chronic infection in CF lungs. The rhl quorum-sensing (QS) system allows P. aeruginosa to regulate diverse metabolic adaptations and virulence. However, how rhl QS system is regulated remains largely unknown. Here, we report that two-component sensor BfmS controls rhl QS system by repressing its cognate response regulator BfmR, which directly suppresses the expression of rhl QS regulator RhlR gene and reduces the production of QS signal molecule N-butanoyl-L-homoserine lactone (C4-HSL). We find that BfmS is critical to the ability of P. aeruginosa to modulate the expression of virulence-associated traits and adapt to the host. Intriguingly, although wild-type BfmS is a repressor of BfmR, naturally occurring missense mutation (L181P, L181P/E376Q, or R393H) can convert its function from a repressor to an activator of BfmR, leading to BfmR activation, which in turn reduces the level of rhl QS signal C4-HSL. These results, therefore, provide important and novel insight into the regulation and evolution of P. aeruginosa virulence.
Collapse
Affiliation(s)
- Qiao Cao
- Hainan University, Haikou, Hainan, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yue Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Institute of Chemistry and BioMedical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Feifei Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yongjie Xia
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jingyu Lou
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xue Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Nana Yang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoxu Sun
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Qin Zhang
- State Key Laboratory of Respiratory Diseases and the First Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | - Chao Zhuo
- State Key Laboratory of Respiratory Diseases and the First Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | - Xi Huang
- Hainan University, Haikou, Hainan, China
| | - Xin Deng
- Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois, United States of America
| | - Cai-Guang Yang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yan Ye
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| | - Jing Zhao
- Institute of Chemistry and BioMedical Sciences, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- * E-mail: (JZ); (MW); (LL)
| | - Min Wu
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
- * E-mail: (JZ); (MW); (LL)
| | - Lefu Lan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- * E-mail: (JZ); (MW); (LL)
| |
Collapse
|
61
|
Vandermeulen E, Ruttens D, Verleden SE, Vos R, Van Raemdonck DE, Kastelijn EA, Wauters E, Lambrechts D, Nawrot TS, Cox B, Verleden GM, Vanaudenaerde BM. Genetic Variation in Caveolin-1 Affects Survival After Lung Transplantation. Transplantation 2014; 98:354-9. [DOI: 10.1097/tp.0000000000000058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
62
|
Ma JQ, Ding J, Xiao ZH, Liu CM. Ursolic acid ameliorates carbon tetrachloride-induced oxidative DNA damage and inflammation in mouse kidney by inhibiting the STAT3 and NF-κB activities. Int Immunopharmacol 2014; 21:389-95. [PMID: 24880019 DOI: 10.1016/j.intimp.2014.05.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 05/02/2014] [Accepted: 05/02/2014] [Indexed: 01/01/2023]
Abstract
Ursolic acid (UA), a common pentacyclic triterpenoid compound, has been reported to have many benefits and medicinal properties. However, its protective effects against carbon tetrachloride (CCl4) induced injury in kidneys are not yet clear. In the current report, we investigated whether UA inhibited the oxidative stress and inflammation in the kidneys of CCl4 treated mice. Male ICR mice were injected with CCl4 with or without UA co-administration (25 and 50mg/kg intragastrically once daily) for six weeks. Our data showed that UA significantly prevented CCl4-induced nephrotoxicity in a dose-dependent manner, indicated by both diagnostic indicators of kidney damage and histopathological analysis. Moreover, CCl4-induced profound elevation of ROS and oxidative stress, as evidenced by the increase of lipid peroxidation level and the depletion of the total antioxidant capacity (TAC) level in the kidney, was suppressed by treatment with UA. UA also decreased 8-hydroxy-2-deoxyguanosine (one product of oxidative DNA damage) levels. Furthermore, protein expression by Western blot analysis showed that UA significantly decreased production of pro-inflammatory markers including tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), interleukin-17 (IL-17) and cyclooxygenase-2 (COX-2) in CCl4-treated mouse kidney. In exploring the underlying mechanisms of UA action, we found that UA increased the phosphorylation of transcription 3 (STAT3), which in turn activated the nuclear factor kappa B (NF-kappaB) and the inflammatory cytokines in the kidneys. In conclusion, these results suggested that the inhibition of CCl4-induced inflammation by UA is due at least in part to its anti-oxidant activity and its ability to modulate the STAT3 and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Jie-Qiong Ma
- School of Chemistry and Pharmaceutical Engineering, Sichuan University of Science and Engineering, 643000 Zigong City, Sichuan Province, PR China.
| | - Jie Ding
- School of Chemistry and Pharmaceutical Engineering, Sichuan University of Science and Engineering, 643000 Zigong City, Sichuan Province, PR China
| | - Zheng-Hua Xiao
- School of Chemistry and Pharmaceutical Engineering, Sichuan University of Science and Engineering, 643000 Zigong City, Sichuan Province, PR China
| | - Chan-Min Liu
- School of Life Science, Jiangsu Normal University, No. 101, Shanghai Road, Tangshan New Area, 221116 Xuzhou City, Jiangsu Province, PR China
| |
Collapse
|
63
|
Chen F, Barman S, Yu Y, Haigh S, Wang Y, Black SM, Rafikov R, Dou H, Bagi Z, Han W, Su Y, Fulton DJR. Caveolin-1 is a negative regulator of NADPH oxidase-derived reactive oxygen species. Free Radic Biol Med 2014; 73:201-13. [PMID: 24835767 PMCID: PMC4228786 DOI: 10.1016/j.freeradbiomed.2014.04.029] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 04/25/2014] [Accepted: 04/27/2014] [Indexed: 01/14/2023]
Abstract
Changes in the expression and function of caveolin-1 (Cav-1) have been proposed as a pathogenic mechanism underlying many cardiovascular diseases. Cav-1 binds to and regulates the activity of numerous signaling proteins via interactions with its scaffolding domain. In endothelial cells, Cav-1 has been shown to reduce reactive oxygen species (ROS) production, but whether Cav-1 regulates the activity of NADPH oxidases (Noxes), a major source of cellular ROS, has not yet been shown. Herein, we show that Cav-1 is primarily expressed in the endothelium and adventitia of pulmonary arteries (PAs) and that Cav-1 expression is reduced in isolated PAs from multiple models of pulmonary artery hypertension (PH). Reduced Cav-1 expression correlates with increased ROS production in the adventitia of hypertensive PA. In vitro experiments revealed a significant ability of Cav-1 and its scaffolding domain to inhibit Nox1-5 activity and it was also found that Cav-1 binds to Nox5 and Nox2 but not Nox4. In addition to posttranslational actions, in primary cells, Cav-1 represses the mRNA and protein expression of Nox2 and Nox4 through inhibition of the NF-κB pathway. Last, in a mouse hypoxia model, the genetic ablation of Cav-1 increased the expression of Nox2 and Nox4 and exacerbated PH. Together, these results suggest that Cav-1 is a negative regulator of Nox function via two distinct mechanisms, acutely through direct binding and chronically through alteration of expression levels. Accordingly, the loss of Cav-1 expression in cardiovascular diseases such as PH may account for the increased Nox activity and greater production of ROS.
Collapse
Affiliation(s)
- Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA.
| | - Scott Barman
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Yanfang Yu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu 210029, China; Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA
| | - Steven Haigh
- Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA
| | - Yusi Wang
- Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA
| | | | | | | | - Zsolt Bagi
- Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA
| | - Weihong Han
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - David J R Fulton
- Vascular Biology Center and Georgia Regents University, Augusta, GA 30912, USA; Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
64
|
Liang H, Deng X, Li X, Ye Y, Wu M. Molecular mechanisms of master regulator VqsM mediating quorum-sensing and antibiotic resistance in Pseudomonas aeruginosa. Nucleic Acids Res 2014; 42:10307-20. [PMID: 25034696 PMCID: PMC4176358 DOI: 10.1093/nar/gku586] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The Pseudomonas aeruginosa quorum-sensing (QS) systems contribute to bacterial homeostasis and pathogenicity. Although the AraC-family transcription factor VqsM has been characterized to control the production of virulence factors and QS signaling molecules, its detailed regulatory mechanisms still remain elusive. Here, we report that VqsM directly binds to the lasI promoter region, and thus regulates its expression. To identify additional targets of VqsM in P. aeruginosa PAO1, we performed chromatin immunoprecipitation (ChIP) followed by high-throughput DNA sequencing (ChIP-seq) and detected 48 enriched loci harboring VqsM-binding peaks in the P. aeruginosa genome. The direct regulation of these genes by VqsM has been confirmed by electrophoretic mobility shift assays and quantitative real-time polymerase chain reactions. A VqsM-binding motif was identified by using the MEME suite and verified by footprint assays in vitro. In addition, VqsM directly bound to the promoter regions of the antibiotic resistance regulator NfxB and the master type III secretion system (T3SS) regulator ExsA. Notably, the vqsM mutant displayed more resistance to two types of antibiotics and promoted bacterial survival in a mouse model, compared to wild-type PAO1. Collectively, this work provides new cues to better understand the detailed regulatory networks of QS systems, T3SS, and antibiotic resistance.
Collapse
Affiliation(s)
- Haihua Liang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, ShaanXi 710069, China Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Xin Deng
- Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Xuefeng Li
- Department of Basic Science, School of Medicine and Health Science, University of North Dakota, 501 North Columbia Rd, EJRF Building, Room 2726, ND 58203, USA
| | - Yan Ye
- Department of Basic Science, School of Medicine and Health Science, University of North Dakota, 501 North Columbia Rd, EJRF Building, Room 2726, ND 58203, USA
| | - Min Wu
- Department of Basic Science, School of Medicine and Health Science, University of North Dakota, 501 North Columbia Rd, EJRF Building, Room 2726, ND 58203, USA
| |
Collapse
|
65
|
Zhao K, Li Y, Yue B, Wu M. Genes as early responders regulate quorum-sensing and control bacterial cooperation in Pseudomonas aeruginosa. PLoS One 2014; 9:e101887. [PMID: 25006971 PMCID: PMC4090235 DOI: 10.1371/journal.pone.0101887] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 06/12/2014] [Indexed: 11/19/2022] Open
Abstract
Quorum-sensing (QS) allows bacterial communication to coordinate the production of extracellular products essential for population fitness at higher cell densities. It has been generally accepted that a significant time duration is required to reach appropriate cell density to activate the relevant quiescent genes encoding these costly but beneficial public goods. Which regulatory genes are involved and how these genes control bacterial communication at the early phases are largely un-explored. By determining time-dependent expression of QS-related genes of the opportunistic pathogen Pseudomonas aerugionsa, we show that the induction of social cooperation could be critically influenced by environmental factors to optimize the density of population. In particular, small regulatory RNAs (RsmY and RsmZ) serving as early responders, can promote the expression of dependent genes (e.g. lasR) to boost the synthesis of intracellular enzymes and coordinate instant cooperative behavior in bacterial cells. These early responders, acting as a rheostat to finely modulate bacterial cooperation, which may be quickly activated under environment threats, but peter off when critical QS dependent genes are fully functional for cooperation. Our findings suggest that RsmY and RsmZ critically control the timing and levels of public goods production, which may have implications in sociomicrobiology and infection control.
Collapse
Affiliation(s)
- Kelei Zhao
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Yi Li
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Bisong Yue
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Min Wu
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| |
Collapse
|
66
|
Ye Y, Li X, Wang W, Ouedraogo KC, Li Y, Gan C, Tan S, Zhou X, Wu M. Atg7 deficiency impairs host defense against Klebsiella pneumoniae by impacting bacterial clearance, survival and inflammatory responses in mice. Am J Physiol Lung Cell Mol Physiol 2014; 307:L355-63. [PMID: 24993132 DOI: 10.1152/ajplung.00046.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Klebsiella pneumoniae (Kp) is a Gram-negative bacterium that can cause serious infections in humans. Autophagy-related gene 7 (Atg7) has been implicated in certain bacterial infections; however, the role of Atg7 in macrophage-mediated immunity against Kp infection has not been elucidated. Here we showed that Atg7 expression was significantly increased in murine alveolar macrophages (MH-S) upon Kp infection, indicating that Atg7 participated in host defense. Knocking down Atg7 with small-interfering RNA increased bacterial burdens in MH-S cells. Using cell biology assays and whole animal imaging analysis, we found that compared with wild-type mice atg7 knockout (KO) mice exhibited increased susceptibility to Kp infection, with decreased survival rates, decreased bacterial clearance, and intensified lung injury. Moreover, Kp infection induced excessive proinflammatory cytokines and superoxide in the lung of atg7 KO mice. Similarly, silencing Atg7 in MH-S cells markedly increased expression levels of proinflammatory cytokines. Collectively, these findings reveal that Atg7 offers critical resistance to Kp infection by modulating both systemic and local production of proinflammatory cytokines.
Collapse
Affiliation(s)
- Yan Ye
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Xuefeng Li
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Wenxue Wang
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Kiswendsida Claude Ouedraogo
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Yi Li
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Changpei Gan
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Shirui Tan
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Xikun Zhou
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Min Wu
- Department of Basic Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota
| |
Collapse
|
67
|
Chen J, Shetty S, Zhang P, Gao R, Hu Y, Wang S, Li Z, Fu J. Aspirin-triggered resolvin D1 down-regulates inflammatory responses and protects against endotoxin-induced acute kidney injury. Toxicol Appl Pharmacol 2014; 277:118-23. [DOI: 10.1016/j.taap.2014.03.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 02/19/2014] [Accepted: 03/25/2014] [Indexed: 12/22/2022]
|
68
|
Li Y, Gan CP, Zhang S, Zhou XK, Li XF, Wei YQ, Yang JL, Wu M. FIP200 is involved in murine pseudomonas infection by regulating HMGB1 intracellular translocation. Cell Physiol Biochem 2014; 33:1733-44. [PMID: 24923305 PMCID: PMC4393741 DOI: 10.1159/000362954] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2014] [Indexed: 02/05/2023] Open
Abstract
Background FIP200, a critical autophagy initiating protein, can participate in numerous cellular functions including cancer development; however, its functional role in P. aeruginosa infection of alveolar macrophages is unknown. Methods To investigate the role of FIP200 in host defense, we transfected murine alveolar macrophage MH-S cells with FIP200 siRNA. Having confirmed that FIP200 knockdown inhibited PAO1-induced autophagosme formation, we sought to characterize the underlying signaling pathways by immunoblotting. Further, we used fip200 KO mice to study the effects of fip200 deficiency on HMGB1 translocation. Results We showed that Pseudomonas PAO1 strain infection facilitated autophagosome formation, whereas knockdown of FIP200 inhibited autophagosome formation and HMGB1 expression in MH-S cells. Silencing FIP200 impaired the translocation of HMGB1 to cytosol of MH-S cells and almost abolished acetylation of HMGB1 during PAO1 infection. In contrast, FIP200 overexpression facilitated the cytosol translocation of HMGB1 from nuclei and increased acetylation of HMGB1 in PAO1-infected MH-S cells. Importantly, expression and acetylation of HMGB1 were also significantly down-regulated in fip200 KO mice following PAO1 infection. Conclusions Collectively, these findings elucidate that FIP200 may regulate expression and translocation of HMGB1 during PAO1 infection, which may indicate novel therapeutic targets to control pulmonary infection.
Collapse
Affiliation(s)
- Yi Li
- State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Zhang H, Lei Y, Yuan P, Li L, Luo C, Gao R, Tian J, Feng Z, Nice EC, Sun J. ROS-mediated autophagy induced by dysregulation of lipid metabolism plays a protective role in colorectal cancer cells treated with gambogic acid. PLoS One 2014; 9:e96418. [PMID: 24810758 PMCID: PMC4014500 DOI: 10.1371/journal.pone.0096418] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Accepted: 04/07/2014] [Indexed: 12/26/2022] Open
Abstract
Gambogic acid (GA), the main active component of gamboge resin, has potent antitumor activity both in vivo and in vitro. However, the underlying molecular mechanisms remain unclear. In this study, we found that GA could initiate autophagy in colorectal cancer cells, and inhibition of the autophagy process accelerated the effect of proliferative inhibition and apoptotic cell death induced by GA, implying a protective role of autophagy. Two-dimensional electrophoresis-based proteomics showed that GA treatment altered the expression of multiple proteins involved in redox signaling and lipid metabolism. Functional studies revealed that GA-induced dysregulation of lipid metabolism could activate 5-lipoxygenase (5-LOX), resulting in intracellular ROS accumulation, followed by inhibition of Akt-mTOR signaling and autophagy initiation. Finally, results using a xenograft model suggested ROS-induced autophagy protect against the antitumor effect of GA. Taken together, these data showed new biological activities of GA against colorectal cancer underlying the protective role of ROS-induced autophagy. This study will provide valuable insights for future studies regarding the anticancer mechanisms of GA.
Collapse
Affiliation(s)
- Haiyuan Zhang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of science and Technology, Wuhan, People's Republic of China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ping Yuan
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of science and Technology, Wuhan, People's Republic of China
| | - Lingjun Li
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of science and Technology, Wuhan, People's Republic of China
| | - Chao Luo
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of science and Technology, Wuhan, People's Republic of China
| | - Rui Gao
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of science and Technology, Wuhan, People's Republic of China
| | - Jun Tian
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of science and Technology, Wuhan, People's Republic of China
| | - Zuohua Feng
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of science and Technology, Wuhan, People's Republic of China
| | - Edouard C Nice
- Monash University, Department of Biochemistry and Molecular Biology, Clayton, Victoria, Australia
| | - Jun Sun
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
70
|
Zhou X, Li X, Ye Y, Zhao K, Zhuang Y, Li Y, Wei Y, Wu M. MicroRNA-302b augments host defense to bacteria by regulating inflammatory responses via feedback to TLR/IRAK4 circuits. Nat Commun 2014; 5:3619. [PMID: 24717937 PMCID: PMC4011559 DOI: 10.1038/ncomms4619] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 03/11/2014] [Indexed: 02/05/2023] Open
Abstract
MicroRNAs (miRNAs) have been implicated in a spectrum of physiological and pathological conditions, including immune responses. miR-302b has been implicated in stem cell differentiation but its role in immunity remains unknown. Here we show that miR-302b is induced by TLR2 and TLR4 through ERK-p38-NF-κB signaling upon Gram-negative bacterium Pseudomonas aeruginosa infection. Suppression of inflammatory responses to bacterial infection is mediated by targeting IRAK4, a protein required for the activation and nuclear translocation of NF-κB. Through negative feedback, enforced expression of miR-302b or IRAK4 siRNA silencing inhibits downstream NF-κB signaling and airway leukocyte infiltration, thereby alleviating lung injury and increasing survival in P. aeruginosa-infected mice. In contrast, miR-302b inhibitors exacerbate inflammatory responses and decrease survival in P. aeruginosa-infected mice and lung cells. These findings reveal that miR-302b is a novel inflammatory regulator of NF-κB activation in respiratory bacterial infections by providing negative feedback to TLRs-mediated immunity.
Collapse
Affiliation(s)
- Xikun Zhou
- 1] Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA [2] State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China [3]
| | - Xuefeng Li
- 1] Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA [2] State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China [3]
| | - Yan Ye
- Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| | - Kelei Zhao
- Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| | - Yan Zhuang
- Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| | - Yi Li
- 1] Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA [2] State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Wu
- Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| |
Collapse
|
71
|
Li X, Zhou X, Li Y, Li J, Privratsky B, Ye Y, Wu E, Gao H, Huang C, Wu M. Lyn regulates inflammatory responses in Klebsiella pneumoniae infection via the p38/NF-κB pathway. Eur J Immunol 2014; 44:763-773. [PMID: 24338528 PMCID: PMC4103995 DOI: 10.1002/eji.201343972] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 10/07/2013] [Accepted: 11/11/2013] [Indexed: 02/05/2023]
Abstract
Klebsiella pneumoniae (Kp) is one of the most common pathogens in nosocomial infections and is becoming increasingly multidrug resistant. However, the underlying molecular pathogenesis of this bacterium remains elusive, limiting the therapeutic options. Understanding the mechanism of its pathogenesis may facilitate the development of anti-bacterial therapeutics. Here, we show that Lyn, a pleiotropic Src tyrosine kinase, is involved in host defense against Kp by regulating phagocytosis process and simultaneously downregulating inflammatory responses. Using acute infection mouse models, we observed that lyn(-/-) mice were more susceptible to Kp with increased mortality and severe lung injury compared with WT mice. Kp infected-lyn(-/-) mice exhibited elevated inflammatory cytokines (IL-6 and TNF-α), and increased superoxide in the lung and other organs. In addition, the phosphorylation of p38 and NF-κB p65 subunit increased markedly in response to Kp infection in lyn(-/-) mice. We also demonstrated that the translocation of p65 from cytoplasm to nuclei increased in cultured murine lung epithelial cells by Lyn siRNA knockdown. Furthermore, lipid rafts clustered with activated Lyn and accumulated in the site of Kp invasion. Taken together, these findings revealed that Lyn may participate in host defense against Kp infection through the negative modulation of inflammatory cytokines.
Collapse
Affiliation(s)
- Xuefeng Li
- The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| | - Xikun Zhou
- The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| | - Yi Li
- The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| | - Jiaxin Li
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| | - Breanna Privratsky
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| | - Yan Ye
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| | - Erxi Wu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Hongwei Gao
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative & Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Canhua Huang
- The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Min Wu
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota 58203-9037, USA
| |
Collapse
|
72
|
Zhao Y, Ye Y, Zhou X, Chen J, Jin Y, Hanson A, Zhao JX, Wu M. Photosensitive fluorescent dye contributes to phototoxicity and inflammatory responses of dye-doped silica NPs in cells and mice. Am J Cancer Res 2014; 4:445-59. [PMID: 24578727 PMCID: PMC3936296 DOI: 10.7150/thno.7653] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 01/27/2014] [Indexed: 11/18/2022] Open
Abstract
Dye-doped fluorescent silica nanoparticles provide highly intense and photostable fluorescence signals. However, some dopant dye molecules are photosensitive. A widely-used photosensitive fluorescent dopant, RuBpy, was chosen to systematically investigate the phototoxicity of the dye-doped silica nanoparticles (NPs). We investigated cell viability, DNA damage, and Reactive Oxygen Species (ROS) levels in alveolar macrophages using the dye-doped NPs with or without irradiation. Our results showed that the RuBpy-doped silica NPs could induce significant amount of ROS, DNA damage, apoptosis and impaired proliferation in MH-S cells. In vivo studies in mice showed that RuBpy-doped silica NPs induced significant inflammatory cytokine production and lowered expression in signaling proteins such as ERK1/2 and NF-κB as well as increased lung injury determined by myeloperoxidase and lipid peroxidation. Strikingly, we also found that both RuBpy alone and NPs induced systemic signaling activation in the kidney compared to the liver and lung where showed highly selective signaling patterns, which is more pronounced than RuBpy-doped silica NPs. Moreover, we discovered a critical biomarker (e.g., HMGB1) for silica NPs-induced stress and toxicity and demonstrated differentially-regulated response patterns in various organs. Our results indicate for the first time that the RuBpy-doped silica NPs may impose less inflammatory responses but stronger thermotherapeutic effects on target cells in animals than naked NPs in a time- and dose-dependent manner.
Collapse
|
73
|
Carnesecchi S, Dunand-Sauthier I, Zanetti F, Singovski G, Deffert C, Donati Y, Cagarelli T, Pache JC, Krause KH, Reith W, Barazzone-Argiroffo C. NOX1 is responsible for cell death through STAT3 activation in hyperoxia and is associated with the pathogenesis of acute respiratory distress syndrome. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:537-551. [PMID: 24551274 PMCID: PMC3925898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 12/21/2013] [Indexed: 06/03/2023]
Abstract
Reactive oxygen species (ROS) contribute to alveolar cell death in acute respiratory distress syndrome (ARDS) and we previously demonstrated that NOX1-derived ROS contributed to hyperoxia-induced alveolar cell death in mice. The study investigates whether NOX1 expression is modulated in epithelial cells concomitantly to cell death and associated to STAT3 signaling in the exudative phase of ARDS. In addition, the role of STAT3 activation in NOX1-dependent epithelial cell death was confirmed by using a lung epithelial cell line and in mice exposed to hyperoxia. NOX1 expression, cell death and STAT3 staining were evaluated in the lungs of control and ARDS patients by immunohistochemistry. In parallel, a stable NOX1-silenced murine epithelial cell line (MLE12) and NOX1-deficient mice were used to characterize signalling pathways. In the present study, we show that NOX1 is detected in alveolar epithelial cells of ARDS patients in the exudative stage. In addition, increased alveolar epithelial cell death and phosphorylated STAT3 are observed in ARDS patients and associated with NOX1 expression. Phosphorylated STAT3 is also correlated with TUNEL staining. We also confirmed that NOX1-dependent STAT3 activation participates to alveolar epithelial cell death. Silencing and acute inhibition of NOX1 in MLE12 led to decreased cell death and cleaved-caspase 3 induced by hyperoxia. Additionally, hyperoxia-induced STAT3 phosphorylation is dependent on NOX1 expression and associated with cell death in MLE12 and mice. This study demonstrates that NOX1 is involved in human ARDS pathophysiology and is responsible for the damage occurring in alveolar epithelial cells at least in part via STAT3 signalling pathways.
Collapse
Affiliation(s)
- Stephanie Carnesecchi
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | | | - Filippo Zanetti
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Grigory Singovski
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Christine Deffert
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Yves Donati
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Thomas Cagarelli
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Jean-Claude Pache
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Walter Reith
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| | - Constance Barazzone-Argiroffo
- Department of PediatricsGeneva, Switzerland
- Department of Pathology and Immunology, Medical School, University of GenevaSwitzerland
| |
Collapse
|
74
|
Huang H, Weaver A, Wu E, Li Y, Gao H, Fan W, Wu M. Lipid-based signaling modulates DNA repair response and survival against Klebsiella pneumoniae infection in host cells and in mice. Am J Respir Cell Mol Biol 2013; 49:798-807. [PMID: 23742126 DOI: 10.1165/rcmb.2013-0069oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Klebsiella pneumoniae causes serious infections in the urinary tract, respiratory tract, and blood. Lipid rafts, also known as membrane microdomains, have been linked to the pathogenesis of bacterial infection. However, whether lipid rafts affect K. pneumoniae internalization into host cells remains unknown. Here, we show for the first time that K. pneumoniae was internalized into lung cells by activating lipid rafts. Disrupting lipid rafts by methyl-β-cyclodextrin inhibited pathogen internalization, impairing host defense. A deficient mutant of capsule polysaccharide (CPS) showed a higher internalization rate than a wild-type strain, indicating that CPS may inhibit bacterial entry to host cells. Furthermore, lipid rafts may affect the function of extracellular regulated kinase (ERK)-1/2, and knocking down ERK1/2 via short, interfering RNA increased apoptosis in both alveolar macrophages and epithelial cells after infection. To gain insights into bacterial pathogenesis, we evaluated the impact of lipid rafts on DNA integrity, and showed that raft aggregates also affect DNA damage and DNA repair responses (i.e., 8-oxoguanine DNA glycosylase [Ogg1]) through the regulation of reactive oxygen species. Importantly, cells overexpressing Ogg1 demonstrated reduced cytotoxicity during bacterial infection. Taken together, these results suggest that lipid rafts may modulate bacterial internalization, thereby affecting DNA damage and repair, which is critical to host defense against K. pneumoniae.
Collapse
Affiliation(s)
- Huang Huang
- 1 Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota
| | | | | | | | | | | | | |
Collapse
|
75
|
Pseudomonas aeruginosa outer membrane vesicles modulate host immune responses by targeting the Toll-like receptor 4 signaling pathway. Infect Immun 2013; 81:4509-18. [PMID: 24082079 DOI: 10.1128/iai.01008-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Bacteria can naturally secrete outer membrane vesicles (OMVs) as pathogenic factors, while these vesicles may also serve as immunologic regulators if appropriately prepared. However, it is largely unknown whether Pseudomonas aeruginosa OMVs can activate inflammatory responses and whether immunization with OMVs can provide immune protection against subsequent infection. We purified and identified OMVs, which were then used to infect lung epithelial cells in vitro as well as C57BL/6J mice to investigate the immune response and the underlying signaling pathway. The results showed that OMVs generated from P. aeruginosa wild-type strain PAO1 were more cytotoxic to alveolar epithelial cells than those from quorum-sensing (QS)-deficient strain PAO1-ΔlasR. The levels of Toll-like receptor 4 (TLR4) and proinflammatory cytokines, including interleukin-1β (IL-1β) and IL-6, increased following OMV infection. Compared with lipopolysaccharide (LPS), lysed OMVs in which the membrane structures were broken induced a weak immune response. Furthermore, expression levels of TLR4-mediated responders (i.e., cytokines) were markedly downregulated by the TLR4 inhibitor E5564. Active immunization with OMVs or passive transfer of sera with a high cytokine quantity acquired from OMV-immunized mice could protect healthy mice against subsequent lethal PAO1 challenges (1.5 × 10(11) CFU). Collectively, these findings indicate that naturally secreted P. aeruginosa OMVs may trigger significant inflammatory responses via the TLR4 signaling pathway and protect mice against pseudomonal lung infection.
Collapse
|
76
|
Feng H, Guo W, Han J, Li XA. Role of caveolin-1 and caveolae signaling in endotoxemia and sepsis. Life Sci 2013; 93:1-6. [PMID: 23727353 DOI: 10.1016/j.lfs.2013.05.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 05/09/2013] [Accepted: 05/14/2013] [Indexed: 12/28/2022]
Abstract
Caveolae, plasma membrane invaginations of 60-80nm in diameter, are a subset of lipid rafts enriched in cholesterol and sphingolipids. Caveolae are expressed in various tissues and cell types, such as endothelial cells, macrophages, neutrophils and adipocytes. The functions of caveolae are diverse and include endocytosis, transcytosis, potocytosis, calcium signaling, and regulation of various signaling events. Although growing evidence has increased our understanding of caveolae function, the role of caveolae in sepsis is still a controversial issue. In this review, we present a number of studies addressing caveolae and sepsis and describe the signaling pathways involved, including the LPS-eNOS-TLR4-NFκB, MKK3/p38 MAPK, cPLA2/p38 MAPK, STAT3/NFκB and IL-1β-IL-1R1 pathways. Different studies using endotoxemia and bacteremia animal models have provided distinct conclusions about the function of caveolae, and we discuss these inconsistencies. Taken together, the current data suggest that the function of caveolae in sepsis, which involves a number of signaling pathways, is complex and warrants further studies.
Collapse
Affiliation(s)
- Hong Feng
- Department of Tumor Research and Therapy Center, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Wen Guo
- Taian Central Hospital, Taian, Shandong 271000, China
| | - Junqing Han
- Department of Tumor Research and Therapy Center, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Xiang-An Li
- Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States.
| |
Collapse
|
77
|
Deng L, Lei Y, Liu R, Li J, Yuan K, Li Y, Chen Y, Liu Y, Lu Y, Edwards CK, Huang C, Wei Y. Pyrvinium targets autophagy addiction to promote cancer cell death. Cell Death Dis 2013; 4:e614. [PMID: 23640456 PMCID: PMC3674351 DOI: 10.1038/cddis.2013.142] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Autophagy is a cellular catabolic process by which long-lived proteins and damaged organelles are degradated by lysosomes. Activation of autophagy is an important survival mechanism that protects cancer cells from various stresses, including anticancer agents. Recent studies indicate that pyrvinium pamoate, an FDA-approved antihelminthic drug, exhibits wide-ranging anticancer activity. Here we demonstrate that pyrvinium inhibits autophagy both in vitro and in vivo. We further demonstrate that the inhibition of autophagy is mammalian target of rapamycin independent but depends on the transcriptional inhibition of autophagy genes. Moreover, the combination of pyrvinium with autophagy stimuli improves its toxicity against cancer cells, and pretreatment of cells with 3-MA or siBeclin1 partially protects cells from pyrvinium-induced cell death under glucose starvation, suggesting that targeted autophagy addiction is involved in pyrvinium-mediated cytotoxicity. Finally, in vivo studies show that the combination therapy of pyrvinium with the anticancer and autophagy stimulus agent, 2-deoxy-𝒟-glucose (2-DG), is significantly more effective in inhibiting tumor growth than pyrvinium or 2-DG alone. This study supports a novel cancer therapeutic strategy based on targeting autophagy addiction and implicates using pyrvinium as an autophagy inhibitor in combination with chemotherapeutic agents to improve their therapeutic efficacy.
Collapse
Affiliation(s)
- Longfei Deng
- The State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Zhang PX, Murray TS, Villella VR, Ferrari E, Esposito S, D'Souza A, Raia V, Maiuri L, Krause DS, Egan ME, Bruscia EM. Reduced caveolin-1 promotes hyperinflammation due to abnormal heme oxygenase-1 localization in lipopolysaccharide-challenged macrophages with dysfunctional cystic fibrosis transmembrane conductance regulator. THE JOURNAL OF IMMUNOLOGY 2013; 190:5196-206. [PMID: 23606537 DOI: 10.4049/jimmunol.1201607] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We have previously reported that TLR4 signaling is increased in LPS-stimulated cystic fibrosis (CF) macrophages (MΦs), contributing to the robust production of proinflammatory cytokines. The heme oxygenase-1 (HO-1)/CO pathway modulates cellular redox status, inflammatory responses, and cell survival. The HO-1 enzyme, together with the scaffold protein caveolin 1 (CAV-1), also acts as a negative regulator of TLR4 signaling in MΦs. In this study, we demonstrate that in LPS-challenged CF MΦs, HO-1 does not compartmentalize normally to the cell surface and instead accumulates intracellularly. The abnormal HO-1 localization in CF MΦs in response to LPS is due to decreased CAV-1 expression, which is controlled by the cellular oxidative state, and is required for HO-1 delivery to the cell surface. Overexpression of HO-1 or stimulating the pathway with CO-releasing molecules enhances CAV-1 expression in CF MΦs, suggesting a positive-feed forward loop between HO-1/CO induction and CAV-1 expression. These manipulations re-established HO-1 and CAV-1 cell surface localization in CF MΦs. Consistent with restoration of HO-1/CAV-1-negative regulation of TLR4 signaling, genetic or pharmacological (CO-releasing molecule 2) induced enhancement of this pathway decreased the inflammatory response of CF MΦs and CF mice treated with LPS. In conclusion, our results demonstrate that the counterregulatory HO-1/CO pathway, which is critical in balancing and limiting the inflammatory response, is defective in CF MΦs through a CAV-1-dependent mechanism, exacerbating the CF MΦ response to LPS. This pathway could be a potential target for therapeutic intervention for CF lung disease.
Collapse
Affiliation(s)
- Ping-Xia Zhang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06509, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Hitkova I, Yuan G, Anderl F, Gerhard M, Kirchner T, Reu S, Röcken C, Schäfer C, Schmid RM, Vogelmann R, Ebert MPA, Burgermeister E. Caveolin-1 protects B6129 mice against Helicobacter pylori gastritis. PLoS Pathog 2013; 9:e1003251. [PMID: 23592983 PMCID: PMC3623771 DOI: 10.1371/journal.ppat.1003251] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 02/04/2013] [Indexed: 02/07/2023] Open
Abstract
Caveolin-1 (Cav1) is a scaffold protein and pathogen receptor in the mucosa of the gastrointestinal tract. Chronic infection of gastric epithelial cells by Helicobacter pylori (H. pylori) is a major risk factor for human gastric cancer (GC) where Cav1 is frequently down-regulated. However, the function of Cav1 in H. pylori infection and pathogenesis of GC remained unknown. We show here that Cav1-deficient mice, infected for 11 months with the CagA-delivery deficient H. pylori strain SS1, developed more severe gastritis and tissue damage, including loss of parietal cells and foveolar hyperplasia, and displayed lower colonisation of the gastric mucosa than wild-type B6129 littermates. Cav1-null mice showed enhanced infiltration of macrophages and B-cells and secretion of chemokines (RANTES) but had reduced levels of CD25+ regulatory T-cells. Cav1-deficient human GC cells (AGS), infected with the CagA-delivery proficient H. pylori strain G27, were more sensitive to CagA-related cytoskeletal stress morphologies (“humming bird”) compared to AGS cells stably transfected with Cav1 (AGS/Cav1). Infection of AGS/Cav1 cells triggered the recruitment of p120 RhoGTPase-activating protein/deleted in liver cancer-1 (p120RhoGAP/DLC1) to Cav1 and counteracted CagA-induced cytoskeletal rearrangements. In human GC cell lines (MKN45, N87) and mouse stomach tissue, H. pylori down-regulated endogenous expression of Cav1 independently of CagA. Mechanistically, H. pylori activated sterol-responsive element-binding protein-1 (SREBP1) to repress transcription of the human Cav1 gene from sterol-responsive elements (SREs) in the proximal Cav1 promoter. These data suggested a protective role of Cav1 against H. pylori-induced inflammation and tissue damage. We propose that H. pylori exploits down-regulation of Cav1 to subvert the host's immune response and to promote signalling of its virulence factors in host cells. Infection with the bacterium Helicobacter pylori (H. pylori) mainly affects children in the developing countries who are at risk to progress to gastric cancer (GC) as adults after many years of persistent infection, especially with strains which are positive for the oncogenic virulence factor CagA. Eradication of H. pylori by antibiotics is a treatment of choice but may also alter the susceptibility to allergies and other tumor types. Thus, novel diagnostic or prognostic markers are needed which detect early molecular changes in the stomach mucosa during the transition of chronic inflammation to cancer. In our study, we found that the tumor suppressor caveolin-1 (Cav1) is reduced upon infection with H. pylori, and CagA was sufficient but not necessary for this down-regulation. Loss of Cav1 was caused by H. pylori-dependent activation of sterol-responsive element-binding protein-1 (SREBP1), and this event abolished the interaction of Cav1 with p120 RhoGTPase-activating protein/deleted in liver cancer-1 (p120RhoGAP/DLC1), a second bona fide tumor suppressor in gastric tissue. Conclusively, Cav1 and DLC1 may constitute novel molecular markers in the H. pylori-infected gastric mucosa before neoplastic transformation of the epithelium.
Collapse
Affiliation(s)
- Ivana Hitkova
- Department of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gang Yuan
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Florian Anderl
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, München, Germany
| | - Markus Gerhard
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, München, Germany
- German Centre for Infection Research (DZIF), München, Germany
| | - Thomas Kirchner
- Institute of Pathology, Klinikum der Universität München, München, Germany
| | - Simone Reu
- Institute of Pathology, Klinikum der Universität München, München, Germany
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts Universität, Kiel, Germany
| | - Claus Schäfer
- Department of Medicine II, Klinikum der Universität München, München, Germany
| | - Roland M. Schmid
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Roger Vogelmann
- Department of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P. A. Ebert
- Department of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Elke Burgermeister
- Department of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- * E-mail:
| |
Collapse
|
80
|
Phenotypic and Molecular Characterization of MCF10DCIS and SUM Breast Cancer Cell Lines. Int J Breast Cancer 2013; 2013:872743. [PMID: 23401782 PMCID: PMC3562669 DOI: 10.1155/2013/872743] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/31/2012] [Accepted: 11/08/2012] [Indexed: 12/20/2022] Open
Abstract
We reviewed the phenotypic and molecular characteristics of MCF10DCIS.com and the SUM cell lines based on numerous studies performed over the years. The major signaling pathways that give rise to the phenotype of these cells may serve as a good resource of information when researchers in drug discovery and development use these cells to identify novel targets and biomarkers. Major signaling pathways and mutations affecting the coding sequence are also described providing important information when using these cells as a model in a variety of studies.
Collapse
|
81
|
Guo Q, Shen N, Yuan K, Li J, Wu H, Zeng Y, Fox J, Bansal AK, Singh BB, Gao H, Wu M. Caveolin-1 plays a critical role in host immunity against Klebsiella pneumoniae by regulating STAT5 and Akt activity. Eur J Immunol 2012; 42:1500-1511. [PMID: 22678904 PMCID: PMC3644991 DOI: 10.1002/eji.201142051] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Caveolin-1 (Cav1) is a structural protein of caveolae. Although Cav1 is associated with certain bacterial infections, it is unknown whether Cav1 is involved in host immunity against Klebsiella pneumoniae, the third most commonly isolated microorganism from bacterial sepsis patients. Here, we showed that cav1 knockout mice succumbed to K. pneumoniae infection with markedly decreased survival rates, increased bacterial burdens, intensified tissue injury, hyperactive proinflammatory cytokines, and systemic bacterial dissemination as compared with WT mice. Knocking down Cav1 by a dominant negative approach in lung epithelial MLE-12 cells resulted in similar outcomes (decreased bacterial clearance and increased proinflammatory cytokine production). Furthermore, we revealed that STAT5 influences the GSK3β-β-catenin-Akt pathway, which contributes to the intensive inflammatory response and rapid infection dissemination seen in Cav1 deficiency. Collectively, our findings indicate that Cav1 may offer resistance to K. pneumoniae infection, by affecting both systemic and local production of proinflammatory cytokines via the actions of STAT5 and the GSK3β-β-catenin-Akt pathway.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota, USA
- Department of Rheumatology Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Nan Shen
- Department of Rheumatology Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Kefei Yuan
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota, USA
| | - Jiaxin Li
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota, USA
- Department of Hepatobiliary and Pancreatic Surgery West China Hospital, Sichuan University, Chengdu, China
| | - Hong Wu
- Department of Hepatobiliary and Pancreatic Surgery West China Hospital, Sichuan University, Chengdu, China
| | - Yong Zeng
- Department of Hepatobiliary and Pancreatic Surgery West China Hospital, Sichuan University, Chengdu, China
| | - John Fox
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota, USA
| | - Arvind K. Bansal
- Pulmonary, Critical Care and Sleep Medicine, the Altru Hospital, Grand Forks, North Dakota, USA
| | - Brij B. Singh
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota, USA
| | - Hongwei Gao
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Min Wu
- Department of Biochemistry and Molecular Biology, University of North Dakota, Grand Forks, North Dakota, USA
| |
Collapse
|
82
|
Abstract
Despite the progress in medical treatment sepsis remains one of the major causes of death in pediatric and elderly patients. Understanding signaling pathways associated with sepsis may be of key significance for designing more efficient therapeutic approaches which could alleviate sepsis outcome. Earlier studies suggested that cholesteroland sphingolipid-rich lipid rafts and their morphologically distinct subset, caveolaecan be utilized by certain bacterial pathogens to enter and invade host cells. Moreover, there is also evidence that the expression levels of the major caveolar coat proteincaveolin-1 can be regulated by the major component of the outer membrane of Gram-negative bacteria,lipopolysaccharide (LPS) in various cell types involved in sepsis. In particular recent studies using caveolin-1 knockout mice and cells have revealed that caveolin-1 is directly involved in regulating numerous signalingpathways and functions in various cell types of the immune system and other cell types involved in sepsis. Moreover, the most recent report implies that in addition to extensively studied caveolin-1, caveolin-2 is also important in regulating LPS-induced sepsis and might possibly play an opposite role to caveolin-1 in regulating certain pro-inflammatory signaling pathways. The purpose of this review is to discuss these new exciting discoveries relatedto the specific role of caveolin-1 and the less studiedcaveolin-2in regulating signaling and outcome associated with sepsis induced by LPS and pathogenic bacteria at molecular, cellular and systemic levels.
Collapse
Affiliation(s)
- Grzegorz Sowa
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia,USA
| |
Collapse
|