151
|
Li Q, Fang Y, Zhu P, Ren CY, Chen H, Gu J, Jia YP, Wang K, Tong WD, Zhang WJ, Pan J, Lu DS, Tang B, Mao XH. Burkholderia pseudomallei survival in lung epithelial cells benefits from miRNA-mediated suppression of ATG10. Autophagy 2016; 11:1293-307. [PMID: 26151773 DOI: 10.1080/15548627.2015.1058474] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Burkholderia pseudomallei is the causative agent of melioidosis, a disease with high mortality, which is prevalent in tropical regions of the world. A recent study shows that B. pseudomallei can survive inside mammalian cells because of its ability to actively evade cell autophagy. However, the underlying mechanisms remain unclear. In the present study, based on microarray screening, we found that ATG10 was downregulated following B. pseudomallei infection in A549 human lung epithelial cells. Forced expression of ATG10 accelerated the elimination of intracellular B. pseudomallei by enhancing the process of autophagy. Moreover, MIR4458, MIR4667-5p, and MIR4668-5p were found, by microarray screening, to be upregulated in response to B. pseudomallei infection. These 3 novel miRNAs, MIR4458, MIR4667-5p, and MIR4668-5p, targeted to the 3'-untranslated region of ATG10 in different time-course and spatial manners. Upregulation of these miRNAs reduced the level of ATG10 and inhibited autophagy, leading to increasing survival rate of intracellular B. pseudomallei. Furthermore, the increase of these miRNAs was correlated with the reduced promoter methylation status in A549 cells in response to B. pseudomallei infection. Our results reveal that 3 novel miRNAs regulate autophagy-mediated elimination of B. pseudomallei by targeting ATG10, and provide potential targets for clinical treatment.
Collapse
Affiliation(s)
- Qian Li
- a Department of Clinical Microbiology and Immunology ; Southwest Hospital & College of Medical Laboratory Science; Third Military Medical University ; Chongqing , China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Wu Y, Li D, Wang Y, Chen K, Yang K, Huang X, Zhang Y, Wu M. Pseudomonas aeruginosa promotes autophagy to suppress macrophage-mediated bacterial eradication. Int Immunopharmacol 2016; 38:214-22. [PMID: 27295610 DOI: 10.1016/j.intimp.2016.04.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/12/2016] [Accepted: 04/29/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To explore the role of autophagy on macrophage-mediated phagocytosis and intracellular killing of Pseudomonas aeruginosa (PA), a common extracellular bacterium which often causes various opportunistic infections. METHODS Macrophages were infected with PA or stimulated with zymosan bioparticles. Autophagy was tested by fluorescent microscopy and Western blot for LC3. Phagocytosis and killing efficiency were assessed by plate count assay, flow cytometry or immunofluorescent staining. Phagocytic receptor expression, ROS generation and NO production were examined by PCR, flow cytometry and Griess reaction, respectively. RESULTS PA infection induced autophagy activation in both mouse and human macrophages. Induction of autophagy by rapamycin or starvation significantly inhibited PA internalization by downregulating phagocytosis receptor expression, and suppressed intracellular killing of PA via reducing ROS and NO production in macrophages. While knockdown of autophagy molecules ATG7 or Beclin1 enhanced macrophage-mediated phagocytosis and intracellular killing of PA. Additionally, confocal microscopy data showed that induction of autophagy reduced the number of phagosomes and phagolysosomes in macrophages after stimulation with zymosan bioparticles. CONCLUSIONS Our study suggested that PA promotes autophagy to suppress macrophage-mediated bacterial phagocytosis and intracellular killing. These insights demonstrated a novel immune evasion mechanism employed by PA, which may provide potential therapeutic strategies of PA infectious diseases.
Collapse
Affiliation(s)
- Yongjian Wu
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Dandan Li
- Department of Clinical Laboratory, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 243000, China
| | - Yi Wang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Kang Chen
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Division of Clinical Laboratory, Zhongshan Hospital of Sun Yat-sen University, Zhongshan 528403, China
| | - Kun Yang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Xi Huang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Minhao Wu
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China.
| |
Collapse
|
153
|
Yang K, Wu Y, Xie H, Li M, Ming S, Li L, Li M, Wu M, Gong S, Huang X. Macrophage-mediated inflammatory response decreases mycobacterial survival in mouse MSCs by augmenting NO production. Sci Rep 2016; 6:27326. [PMID: 27251437 PMCID: PMC4890015 DOI: 10.1038/srep27326] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 05/12/2016] [Indexed: 12/28/2022] Open
Abstract
Mycobacterium tuberculosis (MTB) is a hard-to-eradicate intracellular microbe, which escapes host immune attack during latent infection. Recent studies reveal that mesenchymal stem cells (MSCs) provide a protective niche for MTB to maintain latency. However, the regulation of mycobacterial residency in MSCs in the infectious microenvironment remains largely unknown. Here, we found that macrophage-mediated inflammatory response during MTB infection facilitated the clearance of bacilli residing in mouse MSCs. Higher inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production were observed in mouse MSCs under macrophage-mediated inflammatory circumstance. Blocking NO production in MSCs increased the survival of intracellular mycobacteria, indicating NO-mediated antimycobacterial activity. Moreover, both nuclear factor κB (NF-κB) and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathways were involved in iNOS expression and NO production in inflammatory microenvironment. Furthermore, pro-inflammatory cytokine interleukin-1β could trigger NO production in MSCs and exert anti-mycobacterial activity via NF-κB signaling pathway. Neutralization of interleukin-1β in macrophage-mediated inflammatory microenvironment dampened the ability of mouse MSCs to produce NO. Together, our findings demonstrated that macrophage-mediated inflammatory response during mycobacterial infection promotes the clearance of bacilli in mouse MSCs by increasing NO production, which may provide a better understanding of latent MTB infection.
Collapse
Affiliation(s)
- Kun Yang
- Program of Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Institute of Tuberculosis Control, Key laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Yongjian Wu
- Program of Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Institute of Tuberculosis Control, Key laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Heping Xie
- Department of Traditional Chinese Medicine, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Miao Li
- Program of Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Institute of Tuberculosis Control, Key laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Siqi Ming
- Program of Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Institute of Tuberculosis Control, Key laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Liyan Li
- Program of Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Institute of Tuberculosis Control, Key laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Meiyu Li
- Program of Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Institute of Tuberculosis Control, Key laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Minhao Wu
- Program of Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Institute of Tuberculosis Control, Key laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Sitang Gong
- Program of Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xi Huang
- Program of Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Institute of Tuberculosis Control, Key laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| |
Collapse
|
154
|
Bah A, Lacarrière C, Vergne I. Autophagy-Related Proteins Target Ubiquitin-Free Mycobacterial Compartment to Promote Killing in Macrophages. Front Cell Infect Microbiol 2016; 6:53. [PMID: 27242971 PMCID: PMC4863073 DOI: 10.3389/fcimb.2016.00053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/26/2016] [Indexed: 01/02/2023] Open
Abstract
Autophagy is a lysosomal degradative process that plays essential functions in innate immunity, particularly, in the clearance of intracellular bacteria such as Mycobacterium tuberculosis. The molecular mechanisms involved in autophagy activation and targeting of mycobacteria, in innate immune responses of macrophages, are only partially characterized. Autophagy targets pathogenic M. tuberculosis via a cytosolic DNA recognition- and an ubiquitin-dependent pathway. In this report, we show that non-pathogenic M. smegmatis induces a robust autophagic response in THP-1 macrophages with an up regulation of several autophagy-related genes. Autophagy activation relies in part on recognition of mycobacteria by Toll-like receptor 2 (TLR2). Notably, LC3 targeting of M. smegmatis does not rely on membrane damage, ubiquitination, or autophagy receptor recruitment. Lastly, M. smegmatis promotes recruitment of several autophagy proteins, which are required for mycobacterial killing. In conclusion, our study uncovered an alternative autophagic pathway triggered by mycobacteria which involves cell surface recognition but not bacterial ubiquitination.
Collapse
Affiliation(s)
- Aïcha Bah
- Tuberculosis and Infection Biology, Institut de Pharmacologie et de Biologie Structurale, UMR 5089 Centre National de la Recherche Scientifique - Université de Toulouse Toulouse, France
| | - Camille Lacarrière
- Tuberculosis and Infection Biology, Institut de Pharmacologie et de Biologie Structurale, UMR 5089 Centre National de la Recherche Scientifique - Université de Toulouse Toulouse, France
| | - Isabelle Vergne
- Tuberculosis and Infection Biology, Institut de Pharmacologie et de Biologie Structurale, UMR 5089 Centre National de la Recherche Scientifique - Université de Toulouse Toulouse, France
| |
Collapse
|
155
|
Guo W, Han H, Wang Y, Zhang X, Liu S, Zhang G, Hu S. miR-200a regulates Rheb-mediated amelioration of insulin resistance after duodenal-jejunal bypass. Int J Obes (Lond) 2016; 40:1222-32. [PMID: 27121251 PMCID: PMC4973218 DOI: 10.1038/ijo.2016.60] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 02/06/2016] [Accepted: 03/16/2016] [Indexed: 12/20/2022]
Abstract
Objectives: Duodenal–jejunal bypass (DJB) surgery can induce the rapid and durable remission of diabetes. Recent studies indicate that ameliorated hepatic insulin resistance and improved insulin signaling might contribute to the diabetic control observed after DJB. Ras homolog enriched in brain (Rheb) is reported to have an important role in insulin pathway, and some microRNAs (miRNAs) have been found to regulate Rheb. This study was conducted to investigate the effects of DJB on hepatic insulin resistance and the effects of miRNA-200a, a Rheb-targeting miRNA, on the development of DJB-induced amelioration in hepatic insulin resistance. Subjects: We investigated hepatic insulin signaling change and mapped the hepatic miRNAome involved in a rat model of DJB. We studied the effects of miR-200a on Rheb signaling pathway in buffalo rat liver cell lines. Liver tissues were studied and glucose tolerance tests were conducted in DJB rats injected with lentivirus encoding miR-200a inhibitor and diabetic rats injected with miR-200a mimic. Results: Rheb is a potential target of miR-200a. Transfection with an miR-200a inhibitor increased Rheb protein levels and enhanced the feedback action on insulin receptor substrate-dependent insulin signaling, whereas transfection with an miR-200a mimic produced the opposite effects. A luciferase assay confirmed that miR-200a bind to the 3′UTR (untranslated regions) of Rheb. Global downregulation of miR-200a in DJB rats showed impaired insulin sensitivity whereas upregulation of miR-200a in diabetic rats showed amelioration of diabetes. Conclusions: A novel mechanism was identified, in which miR-200a regulates the Rheb-mediated amelioration of insulin resistance in DJB. The findings suggest miR-200a should be further explored as a potential target for the treatment of diabetes.
Collapse
Affiliation(s)
- W Guo
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - H Han
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - Y Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - X Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - S Liu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - G Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| | - S Hu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People's Republic of China
| |
Collapse
|
156
|
Inhibition of IFN-γ-Induced Nitric Oxide Dependent Antimycobacterial Activity by miR-155 and C/EBPβ. Int J Mol Sci 2016; 17:535. [PMID: 27070591 PMCID: PMC4848991 DOI: 10.3390/ijms17040535] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 03/25/2016] [Accepted: 04/01/2016] [Indexed: 12/16/2022] Open
Abstract
miR-155 (microRNA-155) is an important non-coding RNA in regulating host crucial biological regulators. However, its regulatory function in mycobacterium infection remains unclear. Our study demonstrates that miR-155 expression is significantly increased in macrophages after Mycobacterium marinum (M.m) infection. Transfection with anti-miR-155 enhances nitric oxide (NO) synthesis and decreases the mycobacterium burden, and vice versa, in interferon γ (IFN-γ) activated macrophages. More importantly, miR-155 can directly bind to the 3'UTR of CCAAT/enhancer binding protein β (C/EBPβ), a positive transcriptional regulator of nitric oxide synthase (NOS2), and regulate C/EBPβ expression negatively. Knockdown of C/EBPβ inhibit the production of nitric oxide synthase and promoted mycobacterium survival. Collectively, these data suggest that M.m-induced upregulation of miR-155 downregulated the expression of C/EBPβ, thus decreasing the production of NO and promoting mycobacterium survival, which may provide an insight into the function of miRNA in subverting the host innate immune response by using mycobacterium for its own profit. Understanding how miRNAs partly regulate microbicidal mechanisms may represent an attractive way to control tuberculosis infectious.
Collapse
|
157
|
microRNA-146a promotes mycobacterial survival in macrophages through suppressing nitric oxide production. Sci Rep 2016; 6:23351. [PMID: 27025258 PMCID: PMC4812255 DOI: 10.1038/srep23351] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/02/2016] [Indexed: 12/17/2022] Open
Abstract
Macrophages play a crucial role in host innate anti-mycobacterial defense, which is tightly regulated by multiple factors, including microRNAs. Our previous study showed that a panel of microRNAs was markedly up-regulated in macrophages upon mycobacterial infection. Here, we investigated the biological function of miR-146a during mycobacterial infection. miR-146a expression was induced both in vitro and in vivo after Mycobacterium bovis BCG infection. The inducible miR-146a could suppress the inducible nitric oxide (NO) synthase (iNOS) expression and NO generation, thus promoting mycobacterial survival in macrophages. Inhibition of endogenous miR-146a increased NO production and mycobacterial clearance. Moreover, miR-146a attenuated the activation of nuclear factor κB and mitogen-activated protein kinases signaling pathways during BCG infection, which in turn repressed iNOS expression. Mechanistically, miR-146a directly targeted tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) at post-transcriptional level. Silencing TRAF6 decreased iNOS expression and NO production in BCG-infected macrophages, while overexpression of TRAF6 reversed miR-146a-mediated inhibition of NO production and clearance of mycobacteria. Therefore, we demonstrated a novel role of miR-146a in the modulation of host defense against mycobacterial infection by repressing NO production via targeting TRAF6, which may provide a promising therapeutic target for tuberculosis.
Collapse
|
158
|
Pawar K, Sharbati J, Einspanier R, Sharbati S. Mycobacterium bovis BCG Interferes with miR-3619-5p Control of Cathepsin S in the Process of Autophagy. Front Cell Infect Microbiol 2016; 6:27. [PMID: 27014637 PMCID: PMC4783571 DOI: 10.3389/fcimb.2016.00027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/22/2016] [Indexed: 12/14/2022] Open
Abstract
Main survival mechanism of pathogenic mycobacteria is to escape inimical phagolysosomal environment inside the macrophages. Many efforts have been made to unravel the molecular mechanisms behind this process. However, little is known about the involvement of microRNAs (miRNAs) in the regulation of phagolysosomal biosynthesis and maturation. Based on a bottom up approach, we searched for miRNAs that were involved in phagolysosomal processing events in the course of mycobacterial infection of macrophages. After infecting THP-1 derived macrophages with viable and heat killed Mycobacterium bovis BCG (BCG), early time points were identified after co-localization studies of the phagosomal marker protein LAMP1 and BCG. Differences in LAMP1 localization on the phagosomes of both groups were observed at 30 min and 4 h. After in silico based pre-selection of miRNAs, expression analysis at the identified time points revealed down-regulation of three miRNAs: miR-3619-5p, miR-637, and miR-324-3p. Consequently, most likely targets were predicted that were supposed to be mutually regulated by these three studied miRNAs. The lysosomal cysteine protease Cathepsin S (CTSS) and Rab11 family-interacting protein 4 (RAB11FIP4) were up-regulated and were considered to be connected to lysosomal trafficking and autophagy. Interaction studies verified the regulation of CTSS by miR-3619-5p. Down-regulation of CTSS by ectopic miR-3619-5p as well as its specific knockdown by siRNA affected the process of autophagy in THP-1 derived macrophages.
Collapse
Affiliation(s)
- Kamlesh Pawar
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| | - Jutta Sharbati
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| | - Ralf Einspanier
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| | - Soroush Sharbati
- Department of Veterinary Medicine, Institute of Veterinary Biochemistry, Freie Universität Berlin Berlin, Germany
| |
Collapse
|
159
|
The Transcriptional Foundations of Sp110-mediated Macrophage (RAW264.7) Resistance to Mycobacterium tuberculosis H37Ra. Sci Rep 2016; 6:22041. [PMID: 26912204 PMCID: PMC4766572 DOI: 10.1038/srep22041] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 02/04/2016] [Indexed: 12/24/2022] Open
Abstract
Human tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a leading global health problem, causing 1.3 million deaths each year. The nuclear body protein, Sp110, has been linked to TB resistance and previous work showed that it enhances macrophage apoptosis upon Mtb infection. Here, we report on the role of Sp110 in transcriptional regulation of macrophage responses to Mtb through integrated transcriptome and mechanistic studies. Transcriptome analysis revealed that Sp110 regulates genes involved in immune responses, apoptosis, defence responses, and inflammatory responses. Detailed investigation revealed that, in addition to apoptosis-related genes, Sp110 regulates cytokines, chemokines and genes that regulate intracellular survival of Mtb. Moreover, Sp110 regulates miRNA expression in macrophages, with immune and apoptosis-related miRNAs such as miR-125a, miR-146a, miR-155, miR-21a and miR-99b under Sp110 regulation. Additionally, our results showed that Sp110 upregulates BCL2 modifying factor (Bmf) by inhibiting miR-125a, and forced expression of Bmf induces macrophage apoptosis. These findings not only reveal the transcriptional basis of Sp110-mediated macrophage resistance to Mtb, but also suggest potential regulatory roles for Sp110 related to inflammatory responses, miRNA profiles, and the intracellular growth of Mtb.
Collapse
|
160
|
Su Z, Yang Z, Xu Y, Chen Y, Yu Q. MicroRNAs in apoptosis, autophagy and necroptosis. Oncotarget 2016; 6:8474-90. [PMID: 25893379 PMCID: PMC4496162 DOI: 10.18632/oncotarget.3523] [Citation(s) in RCA: 278] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/10/2015] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are endogenous 22 nt non-coding RNAs that target mRNAs for cleavage or translational repression. Numerous miRNAs regulate programmed cell death including apoptosis, autophagy and necroptosis. We summarize how miRNAs regulate apoptotic, autophagic and necroptotic pathways and cancer progression. We also discuss how miRNAs link different types of cell death.
Collapse
Affiliation(s)
- Zhenyi Su
- Department of Biochemistry and Molecular Biology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China.,Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Zuozhang Yang
- Bone and Soft Tissue Tumors Research Center of Yunnan Province, Department of Orthopaedics, the Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, China.,Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan 650118, China
| | - Yongqing Xu
- Department of Orthopaedics, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan 650118, China
| | - Yongbin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Qiang Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
161
|
Deng Q, Wang Y, Zhang Y, Li M, Li D, Huang X, Wu Y, Pu J, Wu M. Pseudomonas aeruginosa Triggers Macrophage Autophagy To Escape Intracellular Killing by Activation of the NLRP3 Inflammasome. Infect Immun 2016; 84:56-66. [PMID: 26467446 PMCID: PMC4694000 DOI: 10.1128/iai.00945-15] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/12/2015] [Indexed: 02/07/2023] Open
Abstract
Assembly of the inflammasome has recently been identified to be a critical event in the initiation of inflammation. However, its role in bacterial killing remains unclear. Our study demonstrates that Pseudomonas aeruginosa infection induces the assembly of the NLRP3 inflammasome and the sequential secretion of caspase1 and interleukin-1β (IL-1β) in human macrophages. More importantly, activation of the NLRP3 inflammasome reduces the killing of P. aeruginosa in human macrophages, without affecting the generation of antimicrobial peptides, reactive oxygen species, and nitric oxide. In addition, our results demonstrate that P. aeruginosa infection increases the amount of the LC3-II protein and triggers the formation of autophagosomes in human macrophages. The P. aeruginosa-induced autophagy was enhanced by overexpression of NLRP3, ASC, or caspase1 but was reduced by knockdown of these core molecules of the NLRP3 inflammasome. Treatment with IL-1β enhanced autophagy in human macrophages. More importantly, IL-1β decreased the macrophage-mediated killing of P. aeruginosa, whereas knockdown of ATG7 or Beclin1 restored the IL-1β-mediated suppression of bacterial killing. Collectively, our study explores a novel mechanism employed by P. aeruginosa to escape from phagocyte killing and may provide a better understanding of the interaction between P. aeruginosa and host immune cells, including macrophages.
Collapse
Affiliation(s)
- Qiuchan Deng
- Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology, Institute of Tuberculosis Control, Sun Yat-sen University, Guangzhou, China Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Yi Wang
- Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology, Institute of Tuberculosis Control, Sun Yat-sen University, Guangzhou, China Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Meiyu Li
- Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology, Institute of Tuberculosis Control, Sun Yat-sen University, Guangzhou, China Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Dandan Li
- Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology, Institute of Tuberculosis Control, Sun Yat-sen University, Guangzhou, China Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Xi Huang
- Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology, Institute of Tuberculosis Control, Sun Yat-sen University, Guangzhou, China Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Yongjian Wu
- Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology, Institute of Tuberculosis Control, Sun Yat-sen University, Guangzhou, China Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Jieying Pu
- Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology, Institute of Tuberculosis Control, Sun Yat-sen University, Guangzhou, China Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| | - Minhao Wu
- Department of Immunology, Zhongshan School of Medicine, Institute of Human Virology, Institute of Tuberculosis Control, Sun Yat-sen University, Guangzhou, China Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China
| |
Collapse
|
162
|
Abstract
A multifunctional microRNA, miR-155, has been recently recognized as an important modulator of numerous biological processes. In our previous in vitro studies, miR-155 was identified as a potential regulator of the endothelial morphogenesis. The present study demonstrates that in vivo inhibition of miR-155 supports cerebral vasculature after experimental stroke. Intravenous injections of a specific miR-155 inhibitor were initiated at 48 h after mouse distal middle cerebral artery occlusion (dMCAO). Microvasculature in peri-infarct area, infarct size, and animal functional recovery were assessed at 1, 2, and 3 weeks after dMCAO. Using in vivo two-photon microscopy, we detected improved blood flow and microvascular integrity in the peri-infarct area of miR-155 inhibitor-injected mice. Electron microscopy revealed that, in contrast to the control group, these animals demonstrated well preserved capillary tight junctions (TJs). Western blot analysis data indicate that improved TJ integrity in the inhibitor-injected animals could be associated with stabilization of the TJ protein ZO-1 and mediated by the miR-155 target protein Rheb. MRI analysis showed significant (34%) reduction of infarct size in miR-155 inhibitor-injected animals at 21 d after dMCAO. Reduced brain injury was confirmed by electron microscopy demonstrating decreased neuronal damage in the peri-infarct area of stroke. Preservation of brain tissue was reflected in efficient functional recovery of inhibitor-injected animals. Based on our findings, we propose that in vivo miR-155 inhibition after ischemia supports brain microvasculature, reduces brain tissue damage, and improves the animal functional recovery. Significance statement: In the present study, we investigated an effect of the in vivo inhibition of a microRNA, miR-155, on brain recovery after experimental cerebral ischemia. To our knowledge, this is the first report describing the efficiency of intravenous anti-miRNA injections in a mouse model of ischemic stroke. The role of miRNAs in poststroke revascularization has been unexplored and in vivo regulation of miRNAs during the subacute phase of stroke has not yet been proposed. Our investigation introduces a new and unexplored approach to cerebral regeneration: regulation of poststroke angiogenesis and recovery through direct modulation of specific miRNA activity. We expect that our findings will lead to the development of novel strategies for regulating neurorestorative processes in the postischemic brain.
Collapse
|
163
|
Kimmey JM, Huynh JP, Weiss LA, Park S, Kambal A, Debnath J, Virgin HW, Stallings CL. Unique role for ATG5 in neutrophil-mediated immunopathology during M. tuberculosis infection. Nature 2015; 528:565-9. [PMID: 26649827 PMCID: PMC4842313 DOI: 10.1038/nature16451] [Citation(s) in RCA: 292] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 11/16/2015] [Indexed: 02/08/2023]
Abstract
Mycobacterium tuberculosis (Mtb), a major global health threat, replicates in macrophages (MΦ) in part by inhibiting phagosome-lysosome fusion, until IFN-γ activates the MΦ to traffic Mtb to the lysosome. How IFN-γ elicits this effect is unknown, but many studies suggest a role for macroautophagy (autophagy herein), a cellular process by which cytoplasmic contents are sequestered into an autophagosome and targeted for lysosomal degradation1. The involvement of autophagy has been defined based on studies in cultured MΦ or dendritic cells (DC) where Mtb colocalizes with autophagy (ATG) factors ATG5, ATG12, ATG16L1, p62, NDP52, Beclin1 and LC32–6, stimulation of autophagy increases bacterial killing6–8, and inhibition of autophagy allows for increased bacterial survival1,2,4,6,7. Notably, these studies reveal modest (e.g. 1.5- to 3-fold change) effects on Mtb replication. In contrast, Atg5fl/fl-LysM-Cre mice lacking ATG5 in monocyte-derived cells and neutrophils (polymorphic mononuclear cells, PMN) succumb to Mtb within 30 days4,9, an extremely severe phenotype similar to mice lacking IFN-γ signaling10,11. Importantly, ATG5 is the only ATG factor that has been studied during Mtb infection in vivo and autophagy-independent functions of ATG5 have been described12–18. For this reason, we used a genetic approach to elucidate the role for multiple ATG genes and the requirement for autophagy in resistance to Mtb infection in vivo. We have discovered that, contrary to expectation, autophagic capacity does not correlate with the outcome of Mtb infection. Instead, ATG5 plays a unique role in protection against Mtb by preventing PMN-mediated immunopathology. Furthermore, while ATG5 is dispensable in alveolar MΦ during Mtb infection, loss of Atg5 in PMN can sensitize mice to Mtb. These findings shift our understanding of the role of ATG5 during Mtb infection, reveal a new outcome of ATG5 activity, and shed light on early events in innate immunity that are required to regulate tuberculosis disease pathology and Mtb replication.
Collapse
Affiliation(s)
- Jacqueline M Kimmey
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Jeremy P Huynh
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Leslie A Weiss
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Sunmin Park
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Amal Kambal
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Jayanta Debnath
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94143, USA
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | - Christina L Stallings
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| |
Collapse
|
164
|
Kumar R, Sahu SK, Kumar M, Jana K, Gupta P, Gupta UD, Kundu M, Basu J. MicroRNA 17-5p regulates autophagy in Mycobacterium tuberculosis-infected macrophages by targeting Mcl-1 and STAT3. Cell Microbiol 2015; 18:679-91. [PMID: 26513648 DOI: 10.1111/cmi.12540] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/23/2015] [Accepted: 10/23/2015] [Indexed: 12/30/2022]
Abstract
Autophagy plays a crucial role in the control of bacterial burden during Mycobacterium tuberculosis infection. MicroRNAs (miRNAs) are small non-coding RNAs that regulate immune signalling and inflammation in response to challenge by pathogens. Appreciating the potential of host-directed therapies designed to control autophagy during mycobacterial infection, we focused on the role of miRNAs in regulating M. tuberculosis-induced autophagy in macrophages. Here, we demonstrate that M. tuberculosis infection leads to downregulation of miR-17 and concomitant upregulation of its targets Mcl-1 and STAT3, a transcriptional activator of Mcl-1. Forced expression of miR-17 reduces expression of Mcl-1 and STAT3 and also the interaction between Mcl-1 and Beclin-1. This is directly linked to enhanced autophagy, because Mcl-1 overexpression attenuates the effects of miR-17. At the same time, transfection with a kinase-inactive mutant of protein kinase C δ (PKCδ) (an activator of STAT3) augments M. tuberculosis-induced autophagy, and miR-17 overexpression diminishes phosphorylation of PKCδ, suggesting that an miR-17/PKC δ/STAT3 axis regulates autophagy during M. tuberculosis infection.
Collapse
Affiliation(s)
- Ranjeet Kumar
- Department of Chemistry, Bose Institute, Kolkata, 700009, India
| | | | - Manish Kumar
- Department of Chemistry, Bose Institute, Kolkata, 700009, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, 700054, India
| | - Pushpa Gupta
- National Jalma Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282006, India
| | - Umesh D Gupta
- National Jalma Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, 282006, India
| | | | - Joyoti Basu
- Department of Chemistry, Bose Institute, Kolkata, 700009, India
| |
Collapse
|
165
|
Holla S, Balaji KN. Epigenetics and miRNA during bacteria-induced host immune responses. Epigenomics 2015; 7:1197-212. [PMID: 26585338 DOI: 10.2217/epi.15.75] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Various cellular processes including the pathogen-specific immune responses, host-pathogen interactions and the related evasion mechanisms rely on the ability of the immune cells to be reprogrammed accurately and in many cases instantaneously. In this context, the exact functions of epigenetic and miRNA-mediated regulation of genes, coupled with recent advent in techniques that aid such studies, make it an attractive field for research. Here, we review examples that involve the epigenetic and miRNA control of the host immune system during infection with bacteria. Interestingly, many pathogens utilize the epigenetic and miRNA machinery to modify and evade the host immune responses. Thus, we believe that global epigenetic and miRNA mapping of such host-pathogen interactions would provide key insights into their cellular functions and help to identify various determinants for therapeutic value.
Collapse
Affiliation(s)
- Sahana Holla
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | | |
Collapse
|
166
|
Bhaduri A, Misra R, Dhamija N. The Tussle Between Mycobacteria and Host: To Eat or Not To Eat. Indian J Microbiol 2015; 55:456-9. [PMID: 26543272 DOI: 10.1007/s12088-015-0541-9] [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: 05/26/2015] [Accepted: 07/04/2015] [Indexed: 11/24/2022] Open
Abstract
Autophagy is a catabolic process of cellular homeostasis evolutionarily conserved in eukaryotes. To block infection of intracellular bacterial pathogens, metazoans deploy autophagy for pathogen clearance through phago-lysosome formation and specific bactericidal peptides. Although an array of research have publicized the host regulatory factors, the function of bacterial effectors are yet to be understood in detail. In this article, we focus on the autophagic response to one of the most successful intracellular bacteria Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Asani Bhaduri
- Cluster Innovation Centre, University of Delhi, Delhi, 110007 India
| | - Richa Misra
- Miranda House, University of Delhi, Delhi, 110007 India
| | - Neeru Dhamija
- Daulat Ram College, University of Delhi, Delhi, 110007 India
| |
Collapse
|
167
|
Abstract
The innate immune system is central for the maintenance of tissue homeostasis and quickly responds to local or systemic perturbations by pathogenic or sterile insults. This rapid response must be metabolically supported to allow cell migration and proliferation and to enable efficient production of cytokines and lipid mediators. This Review focuses on the role of mammalian target of rapamycin (mTOR) in controlling and shaping the effector responses of innate immune cells. mTOR reconfigures cellular metabolism and regulates translation, cytokine responses, antigen presentation, macrophage polarization and cell migration. The mTOR network emerges as an integrative rheostat that couples cellular activation to the environmental and intracellular nutritional status to dictate and optimize the inflammatory response. A detailed understanding of how mTOR metabolically coordinates effector responses by myeloid cells will provide important insights into immunity in health and disease.
Collapse
Affiliation(s)
- Thomas Weichhart
- Medical University of Vienna, Institute of Medical Genetics, Währingerstrasse 10, 1090 Vienna, Austria
| | - Markus Hengstschläger
- Medical University of Vienna, Institute of Medical Genetics, Währingerstrasse 10, 1090 Vienna, Austria
| | - Monika Linke
- Medical University of Vienna, Institute of Medical Genetics, Währingerstrasse 10, 1090 Vienna, Austria
| |
Collapse
|
168
|
Liu Y, Wang R, Jiang J, Yang B, Cao Z, Cheng X. miR-223 is upregulated in monocytes from patients with tuberculosis and regulates function of monocyte-derived macrophages. Mol Immunol 2015; 67:475-481. [PMID: 26296289 DOI: 10.1016/j.molimm.2015.08.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 08/04/2015] [Accepted: 08/06/2015] [Indexed: 12/19/2022]
Abstract
Tuberculosis (TB) is a serious infectious disease that most commonly affects the lungs. Macrophages are among the first line defenders against establishment of Mycobacterium tuberculosis infection in the lungs. In this study, we found that activation and cytokine production in monocyte-derived macrophages (MDM) from patients with active TB was impaired. miR-223 expression was significantly elevated in monocytes and MDM from patients with TB compared with healthy controls. To determine the functional role of miR-223 in macrophages, stable miR-223-expressing and miR-223 antisense-expressing U937 cells were established. Compared with empty vector controls, expression of IL-1β, IL-6, TNF-α and IL-12p40 genes was significantly higher in miR-223 antisense-expressing U937 cells, but lower in miR-223-expressing U937 cells. miR-223 can negatively regulate activation of NF-κB by inhibition of p65 phosphorylation and nuclear translocation. It is concluded that miR-223 can regulate macrophage function by inhibition of cytokine production and NF-κB activation.
Collapse
Affiliation(s)
- Yanhua Liu
- Key Laboratory of Tuberculosis Prevention and Treatment, Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Ruo Wang
- Key Laboratory of Tuberculosis Prevention and Treatment, Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Jing Jiang
- Key Laboratory of Tuberculosis Prevention and Treatment, Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Bingfen Yang
- Key Laboratory of Tuberculosis Prevention and Treatment, Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Zhihong Cao
- Key Laboratory of Tuberculosis Prevention and Treatment, Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China
| | - Xiaoxing Cheng
- Key Laboratory of Tuberculosis Prevention and Treatment, Beijing Key Laboratory of New Techniques for Tuberculosis Diagnosis and Treatment, Institute of Tuberculosis, 309th Hospital, Beijing, China.
| |
Collapse
|
169
|
Frank B, Marcu A, de Oliveira Almeida Petersen AL, Weber H, Stigloher C, Mottram JC, Scholz CJ, Schurigt U. Autophagic digestion of Leishmania major by host macrophages is associated with differential expression of BNIP3, CTSE, and the miRNAs miR-101c, miR-129, and miR-210. Parasit Vectors 2015; 8:404. [PMID: 26226952 PMCID: PMC4521392 DOI: 10.1186/s13071-015-0974-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 06/30/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Autophagy participates in innate immunity by eliminating intracellular pathogens. Consequently, numerous microorganisms have developed strategies to impair the autophagic machinery in phagocytes. In the current study, interactions between Leishmania major (L. m.) and the autophagic machinery of bone marrow-derived macrophages (BMDM) were analyzed. METHODS BMDM were generated from BALB/c mice, and the cells were infected with L. m. promastigotes. Transmission electron microscopy (TEM) and electron tomography were used to investigate the ultrastructure of BMDM and the intracellular parasites. Affymetrix chip analyses were conducted to identify autophagy-related messenger RNAs (mRNAs) and microRNAs (miRNAs). The protein expression levels of autophagy related 5 (ATG5), BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), cathepsin E (CTSE), mechanistic target of rapamycin (MTOR), microtubule-associated proteins 1A/1B light chain 3B (LC3B), and ubiquitin (UB) were investigated through western blot analyses. BMDM were transfected with specific small interfering RNAs (siRNAs) against autophagy-related genes and with mimics or inhibitors of autophagy-associated miRNAs. The infection rates of BMDM were determined by light microscopy after a parasite-specific staining. RESULTS The experiments demonstrated autophagy induction in BMDM after in vitro infection with L. m.. The results suggested a putative MTOR phosphorylation-dependent counteracting mechanism in the early infection phase and indicated that intracellular amastigotes were cleared by autophagy in BMDM in the late infection phase. Transcriptomic analyses and specific downregulation of protein expression with siRNAs suggested there is an association between the infection-specific over expression of BNIP3, as well as CTSE, and the autophagic activity of BMDM. Transfection with mimics of mmu-miR-101c and mmu-miR-129-5p, as well as with an inhibitor of mmu-miR-210-5p, demonstrated direct effects of the respective miRNAs on parasite clearance in L. m.-infected BMDM. Furthermore, Affymetrix chip analyses revealed a complex autophagy-related RNA network consisting of differentially expressed mRNAs and miRNAs in BMDM, which indicates high glycolytic and inflammatory activity in the host macrophages. CONCLUSIONS Autophagy in L. m.-infected host macrophages is a highly regulated cellular process at both the RNA level and the protein level. Autophagy has the potential to clear parasites from the host. The results obtained from experiments with murine host macrophages could be translated in the future to develop innovative and therapeutic antileishmanial strategies for human patients.
Collapse
Affiliation(s)
- Benjamin Frank
- Institute for Molecular Infection Biology, University of Wuerzburg, Josef-Schneider-Str. 2/D15, 97080, Wuerzburg, Germany.
| | - Ana Marcu
- Institute for Molecular Infection Biology, University of Wuerzburg, Josef-Schneider-Str. 2/D15, 97080, Wuerzburg, Germany.
| | - Antonio Luis de Oliveira Almeida Petersen
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK.
- Laboratório de Patologia e Biointervenção, Fundação Oswaldo Cruz-BA, Salvador, Bahia, Brazil.
| | - Heike Weber
- Interdisciplinary Center for Clinical Research (IZKF), University of Wuerzburg, Wuerzburg, Germany.
| | - Christian Stigloher
- Division of Electron Microscopy, Biocenter of the University of Wuerzburg, Wuerzburg, Germany.
| | - Jeremy C Mottram
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, G12 8TA, UK.
| | - Claus Juergen Scholz
- Interdisciplinary Center for Clinical Research (IZKF), University of Wuerzburg, Wuerzburg, Germany.
| | - Uta Schurigt
- Institute for Molecular Infection Biology, University of Wuerzburg, Josef-Schneider-Str. 2/D15, 97080, Wuerzburg, Germany.
| |
Collapse
|
170
|
Zeng FR, Tang LJ, He Y, Garcia RC. An update on the role of miRNA-155 in pathogenic microbial infections. Microbes Infect 2015; 17:613-21. [PMID: 26072128 DOI: 10.1016/j.micinf.2015.05.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 04/28/2015] [Accepted: 05/25/2015] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are evolutionarily conserved and naturally abundant molecules of single-stranded, non-coding RNA from ∼17 to 25 nucleotides long. MiRNAs act at post-transcriptional level either to suppress gene translation or to induce mRNA degradation, according to the degree of complementarity with their target sequences. MiR-155 is a typical representative of the miRNA family that plays a crucial role in cell differentiation and organism development. A number of studies have shown that miR-155 can not only regulate cell proliferation, apoptosis and lymphoma progression, but also plays an important part in various other physiological and pathological processes. For instance, it is involved in hematopoietic cell differentiation, cardiovascular disease, inflammation and immune responses. In recent years, the role of miR-155 in infectious diseases has attracted considerable attention. This review will highlight the participation of miR-155 in the responses to infections caused by different pathogens.
Collapse
Affiliation(s)
- Fu-Rong Zeng
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha 410078, Hunan, China
| | - Li-Jun Tang
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha 410078, Hunan, China.
| | - Ye He
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha 410078, Hunan, China
| | - R C Garcia
- International Centre for Genetic Engineering and Biotechnology, Area Science Park, Padriciano 99, 34012 Trieste, Italy
| |
Collapse
|
171
|
Espert L, Beaumelle B, Vergne I. Autophagy in Mycobacterium tuberculosis and HIV infections. Front Cell Infect Microbiol 2015; 5:49. [PMID: 26082897 PMCID: PMC4451423 DOI: 10.3389/fcimb.2015.00049] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/18/2015] [Indexed: 12/31/2022] Open
Abstract
Human Immunodeficiency Virus (HIV) and Mycobacterium tuberculosis (M.tb) are among the most lethal human pathogens worldwide, each being responsible for around 1.5 million deaths annually. Moreover, synergy between acquired immune deficiency syndrome (AIDS) and tuberculosis (TB) has turned HIV/M.tb co-infection into a major public health threat in developing countries. In the past decade, autophagy, a lysosomal catabolic process, has emerged as a major host immune defense mechanism against infectious agents like M.tb and HIV. Nevertheless, in some instances, autophagy machinery appears to be instrumental for HIV infection. Finally, there is mounting evidence that both pathogens deploy various countermeasures to thwart autophagy. This mini-review proposes an overview of the roles and regulations of autophagy in HIV and M.tb infections with an emphasis on microbial factors. We also discuss the role of autophagy manipulation in the context of HIV/M.tb co-infection. In future, a comprehensive understanding of autophagy interaction with these pathogens will be critical for development of autophagy-based prophylactic and therapeutic interventions for AIDS and TB.
Collapse
Affiliation(s)
- Lucile Espert
- CPBS FRE 3689 Centre National de la Recherche Scientifique, UM Montpellier, France
| | - Bruno Beaumelle
- CPBS FRE 3689 Centre National de la Recherche Scientifique, UM Montpellier, France
| | - Isabelle Vergne
- Institut de Pharmacologie et de Biologie Structurale, UMR 5089 Centre National de la Recherche Scientifique - Université de Toulouse Toulouse, France
| |
Collapse
|
172
|
Huang N, Wu J, Qiu W, Lyu Q, He J, Xie W, Xu N, Zhang Y. MiR-15a and miR-16 induce autophagy and enhance chemosensitivity of Camptothecin. Cancer Biol Ther 2015; 16:941-8. [PMID: 25945419 PMCID: PMC4622988 DOI: 10.1080/15384047.2015.1040963] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
It has been reported that persistent or excessive autophagy promotes cancer cell death during chemotherapy, either by enhancing the induction of apoptosis or mediating autophagic cell death. Here, we show that miR-15a and miR-16 are potent inducers of autophagy. Rictor, a component of mTORC2 complex, is directly targeted by miR-15a/16. Overexpression of miR-15a/16 or depletion of endogenous Rictor attenuates the phosphorylation of mTORC1 and p70S6K, inhibits cell proliferation and G1/S cell cycle transition in human cervical carcinoma HeLa cells. Moreover, miR-15a/16 dramatically enhances anticancer drug camptothecin (CPT)-induced autophagy and apoptotic cell death in HeLa cells. Collectively, these data demonstrate that miR-15a/16 induced autophagy contribute partly to their inhibition of cell proliferation and enhanced chemotherapeutic efficacy of CPT.
Collapse
Affiliation(s)
- Nunu Huang
- a School of Life Sciences; Tsinghua University ; Beijing , China
| | | | | | | | | | | | | | | |
Collapse
|
173
|
Kim JK, Yuk JM, Kim SY, Kim TS, Jin HS, Yang CS, Jo EK. MicroRNA-125a Inhibits Autophagy Activation and Antimicrobial Responses during Mycobacterial Infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:5355-65. [PMID: 25917095 DOI: 10.4049/jimmunol.1402557] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 03/21/2015] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding nucleotides that play critical roles in the regulation of diverse biological functions, including the response of host immune cells. Autophagy plays a key role in activating the antimicrobial host defense against Mycobacterium tuberculosis. Although the pathways associated with autophagy must be tightly regulated at a posttranscriptional level, the contribution of miRNAs and whether they specifically influence the activation of macrophage autophagy during M. tuberculosis infection are largely unknown. In this study, we demonstrate that M. tuberculosis infection of macrophages leads to increased expression of miRNA-125a-3p (miR-125a), which targets UV radiation resistance-associated gene (UVRAG), to inhibit autophagy activation and antimicrobial responses to M. tuberculosis. Forced expression of miR-125a significantly blocked M. tuberculosis-induced activation of autophagy and phagosomal maturation in macrophages, and inhibitors of miR-125a counteracted these effects. Both TLR2 and MyD88 were required for biogenesis of miR-125a during M. tuberculosis infection. Notably, activation of the AMP-activated protein kinase significantly inhibited the expression of miR-125a in M. tuberculosis-infected macrophages. Moreover, either overexpression of miR-125a or silencing of UVRAG significantly attenuated the antimicrobial effects of macrophages against M. tuberculosis. Taken together, these data indicate that miR-125a regulates the innate host defense by inhibiting the activation of autophagy and antimicrobial effects against M. tuberculosis through targeting UVRAG.
Collapse
Affiliation(s)
- Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea; Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Jae-Min Yuk
- Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea; Department of Infection Biology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Soo Yeon Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea; Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Tae Sung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea; Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Hyo Sun Jin
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea; Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Chul-Su Yang
- Department of Molecular and Life Science, College of Science and Technology, Hanyang University, Ansan 426-791, South Korea; and
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea; Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea; Research Institute for Medical Sciences, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| |
Collapse
|
174
|
Tay HL, Kaiko GE, Plank M, Li J, Maltby S, Essilfie AT, Jarnicki A, Yang M, Mattes J, Hansbro PM, Foster PS. Antagonism of miR-328 increases the antimicrobial function of macrophages and neutrophils and rapid clearance of non-typeable Haemophilus influenzae (NTHi) from infected lung. PLoS Pathog 2015; 11:e1004549. [PMID: 25894560 PMCID: PMC4404141 DOI: 10.1371/journal.ppat.1004549] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 11/01/2014] [Indexed: 11/23/2022] Open
Abstract
Pathogenic bacterial infections of the lung are life threatening and underpin chronic lung diseases. Current treatments are often ineffective potentially due to increasing antibiotic resistance and impairment of innate immunity by disease processes and steroid therapy. Manipulation miRNA directly regulating anti-microbial machinery of the innate immune system may boost host defence responses. Here we demonstrate that miR-328 is a key element of the host response to pulmonary infection with non-typeable haemophilus influenzae and pharmacological inhibition in mouse and human macrophages augments phagocytosis, the production of reactive oxygen species, and microbicidal activity. Moreover, inhibition of miR-328 in respiratory models of infection, steroid-induced immunosuppression, and smoke-induced emphysema enhances bacterial clearance. Thus, miRNA pathways can be targeted in the lung to enhance host defence against a clinically relevant microbial infection and offer a potential new anti-microbial approach for the treatment of respiratory diseases. MicroRNAs regulate pathogen recognition pathways by modulating translation. In the immune system, miRNAs have been identified as important regulators of gene expression programs, which regulate differentiation, growth and function of innate and adaptive immune cells. Using miRNA microarray, we demonstrated that lung miRNAs were differentially expressed following non-typeable Haemophilus Influenzae (NTHi) infection in mice. To study the role of a specific miRNA in macrophages, we used antagomir (chemically modified single-stranded RNA analogues, complementary to the target miRNA) to block miRNA function. Interestingly, inhibition of microRNA-328 in mouse and human macrophages increases microbicidal activity by amplifying phagocytosis and production of reactive oxygen species. Inhibition of mR-328 in the lung enhanced bacterial clearance in mouse models of immunosuppression and emphysema. Our study provides proof of principle that miRNA pathways can be targeted in the lung and offer a potential new anti-microbial approach for the treatment of respiratory infection.
Collapse
Affiliation(s)
- Hock L. Tay
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Gerard E. Kaiko
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Maximilian Plank
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - JingJing Li
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Steven Maltby
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Ama-Tawiah Essilfie
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Andrew Jarnicki
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | | | - Joerg Mattes
- Priority Research Centre for Asthma and Respiratory Disease, Discipline of Paediatrics and Child Health, School of Medicine and Public Health, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Philip M. Hansbro
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Paul S. Foster
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
- * E-mail:
| |
Collapse
|
175
|
Han W, Fu X, Xie J, Meng Z, Gu Y, Wang X, Li L, Pan H, Huang W. MiR-26a enhances autophagy to protect against ethanol-induced acute liver injury. J Mol Med (Berl) 2015; 93:1045-55. [PMID: 25877859 PMCID: PMC4577542 DOI: 10.1007/s00109-015-1282-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 03/25/2015] [Accepted: 03/31/2015] [Indexed: 02/05/2023]
Abstract
Abstract Autophagy is a process for the turnover of intracellular organelles and molecules during stress responses. microRNAs (miRNAs) are small, non-coding endogenous RNAs that may regulate almost every cellular process. However, the roles of miRNAs in autophagy are still poorly understood. In this study, we show that miR-26a enhances autophagy in both culture cells and the mouse liver. Hepatic overexpression of miR-26a in mice alleviated ethanol-induced hepatic steatosis and liver injury. Overexpression of miR-26a increased the expression of the autophagy mediator Beclin-1, which is regulated by mitogen-activated protein kinases (MAPKs). We identified DUSP4 and DUSP5, two MAPKs inhibitors, as direct targets of miR-26a. We further demonstrated that miR-26a targeted the 3′-UTRs of several other negative regulators of autophagy. Our results thus identify a novel miRNA-mediated mechanism that enhances cytoprotective autophagy in the liver. Key messages • miR-26a enhances autophagy in liver cells. • Hepatic overexpression of miR-26a in mice alleviates ethanol-induced liver injury. • Overexpression of miR-26a increases the expression of autophagy mediator Beclin-1. • DUSP4 and DUSP5, two MAPKs inhibitors, were identified as direct targets of miR-26a. Electronic supplementary material The online version of this article (doi:10.1007/s00109-015-1282-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, China.,Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Xianghui Fu
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA. .,Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China. .,Collaborative Innovation Center of Biotherapy, Chengdu, 610041, Sichuan, China.
| | - Jiansheng Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, China
| | - Zhipeng Meng
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Ying Gu
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Xichun Wang
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Ling Li
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, Zhejiang, 310016, China.
| | - Wendong Huang
- Division of Molecular Diabetes Research, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
176
|
Zhang GY, Wang J, Jia YJ, Han R, Li P, Zhu DN. MicroRNA-9 promotes the neuronal differentiation of rat bone marrow mesenchymal stem cells by activating autophagy. Neural Regen Res 2015; 10:314-20. [PMID: 25883633 PMCID: PMC4392682 DOI: 10.4103/1673-5374.143439] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2014] [Indexed: 12/31/2022] Open
Abstract
MicroRNA-9 (miR-9) has been shown to promote the differentiation of bone marrow mesenchymal stem cells into neuronal cells, but the precise mechanism is unclear. Our previous study confirmed that increased autophagic activity improved the efficiency of neuronal differentiation in bone marrow mesenchymal stem cells. Accumulating evidence reveals that miRNAs adjust the autophagic pathways. This study used miR-9-1 lentiviral vector and miR-9-1 inhibitor to modulate the expression level of miR-9. Autophagic activity and neuronal differentiation were measured by the number of light chain-3 (LC3)-positive dots, the ratio of LC3-II/LC3, and the expression levels of the neuronal markers enolase and microtubule-associated protein 2. Results showed that LC3-positive dots, the ratio of LC3-II/LC3, and expression of neuron specific enolase and microtubule-associated protein 2 increased in the miR-9+ group. The above results suggest that autophagic activity increased and bone marrow mesenchymal stem cells were prone to differentiate into neuronal cells when miR-9 was overexpressed, demonstrating that miR-9 can promote neuronal differentiation by increasing autophagic activity.
Collapse
Affiliation(s)
- Guang-Yu Zhang
- Rehabilitation and Treatment Center for Children with Cerebral Palsy of Henan Province, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jun Wang
- Rehabilitation and Treatment Center for Children with Cerebral Palsy of Henan Province, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yan-Jie Jia
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Rui Han
- Department of Pediatrics, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ping Li
- Rehabilitation and Treatment Center for Children with Cerebral Palsy of Henan Province, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Deng-Na Zhu
- Rehabilitation and Treatment Center for Children with Cerebral Palsy of Henan Province, the Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
177
|
APF lncRNA regulates autophagy and myocardial infarction by targeting miR-188-3p. Nat Commun 2015; 6:6779. [DOI: 10.1038/ncomms7779] [Citation(s) in RCA: 370] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 02/26/2015] [Indexed: 01/01/2023] Open
|
178
|
Chen Z, Wang T, Liu Z, Zhang G, Wang J, Feng S, Liang J. Inhibition of Autophagy by MiR-30A Induced by Mycobacteria tuberculosis as a Possible Mechanism of Immune Escape in Human Macrophages. Jpn J Infect Dis 2015; 68:420-4. [PMID: 25866116 DOI: 10.7883/yoken.jjid.2014.466] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The regulatory mechanism of miRNA induction in response to Mycobacterium tuberculosis (MTB) infection has not been clearly established. Autophagy has recently been identified as an effective way to control intracellular survival of MTB. In the present study, we demonstrate a novel role of miR-30A in the negative regulation of the autophagy-mediated anti-MTB response. We found that overexpression of miR-30A suppresses the elimination of intracellular MTB through the inhibition of autophagy. Furthermore, there was a negative correlation between concentrations of miR-30A and beclin-1 in MTB positive patients and miR-30A expression decreased after anti-TB treatment. Our results indicate that miR-30A plays a key role in immune response against MTB and, therefore, may serve as a potential target for future treatments of tuberculosis infection.
Collapse
Affiliation(s)
- Zhi Chen
- Institute of Tuberculosis Research, the 309th Hospital of PLA
| | | | | | | | | | | | | |
Collapse
|
179
|
Iwai H, Funatogawa K, Matsumura K, Kato-Miyazawa M, Kirikae F, Kiga K, Sasakawa C, Miyoshi-Akiyama T, Kirikae T. MicroRNA-155 knockout mice are susceptible to Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 2015; 95:246-50. [PMID: 25846955 DOI: 10.1016/j.tube.2015.03.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/15/2015] [Indexed: 12/23/2022]
Abstract
MicroRNAs (miRNAs) are short, conserved, non-coding RNA molecules that repress translation, followed by the decay of miRNA-targeted mRNAs that encode molecules involved in cell differentiation, development, immunity and apoptosis. At least six miRNAs, including microRNA-155 (miR-155), were up-regulated when born marrow-derived macrophages from C57BL/6 mice were infected with Mycobacterium tuberculosis Erdman. C57BL/6 mice intravenously infected with Erdman showed up-regulation of miR-155 in livers and lungs. Following infection, miR-155-deficient C57BL/6 mice died significantly earlier and had significantly higher numbers of CFU in lungs than wild-type mice. Moreover, fewer CD4(+) T cells, but higher numbers of monocytes and neutrophils, were present in the lungs of Erdman-infected miR-155 knockout (miR-155(-/-)) than of wild-type mice. These findings indicated that miR-155 plays a critical role in immune responses to M. tuberculosis.
Collapse
Affiliation(s)
- Hiroki Iwai
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Toyama, Shinjuku, Tokyo 162-8655, Japan
| | - Keiji Funatogawa
- Tochigi Prefectural Institute of Public Health, Tochigi 329-1196, Japan
| | - Kazunori Matsumura
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Toyama, Shinjuku, Tokyo 162-8655, Japan
| | - Masako Kato-Miyazawa
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Toyama, Shinjuku, Tokyo 162-8655, Japan
| | - Fumiko Kirikae
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Toyama, Shinjuku, Tokyo 162-8655, Japan
| | - Kotaro Kiga
- Division of Bacterial Infection, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Chihiro Sasakawa
- Nippon Institute for Biological Science, Tokyo 198-0024, Japan; Division of Bacterial Infection, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan
| | - Tohru Miyoshi-Akiyama
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Toyama, Shinjuku, Tokyo 162-8655, Japan
| | - Teruo Kirikae
- Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, Toyama, Shinjuku, Tokyo 162-8655, Japan.
| |
Collapse
|
180
|
Abstract
Macrophages and neutrophils play a decisive role in host responses to intracellular bacteria including the agent of tuberculosis (TB), Mycobacterium tuberculosis as they represent the forefront of innate immune defense against bacterial invaders. At the same time, these phagocytes are also primary targets of intracellular bacteria to be abused as host cells. Their efficacy to contain and eliminate intracellular M. tuberculosis decides whether a patient initially becomes infected or not. However, when the infection becomes chronic or even latent (as in the case of TB) despite development of specific immune activation, phagocytes have also important effector functions. Macrophages have evolved a myriad of defense strategies to combat infection with intracellular bacteria such as M. tuberculosis. These include induction of toxic anti-microbial effectors such as nitric oxide and reactive oxygen intermediates, the stimulation of microbe intoxication mechanisms via acidification or metal accumulation in the phagolysosome, the restriction of the microbe's access to essential nutrients such as iron, fatty acids, or amino acids, the production of anti-microbial peptides and cytokines, along with induction of autophagy and efferocytosis to eliminate the pathogen. On the other hand, M. tuberculosis, as a prime example of a well-adapted facultative intracellular bacterium, has learned during evolution to counter-balance the host's immune defense strategies to secure survival or multiplication within this otherwise hostile environment. This review provides an overview of innate immune defense of macrophages directed against intracellular bacteria with a focus on M. tuberculosis. Gaining more insights and knowledge into this complex network of host-pathogen interaction will identify novel target sites of intervention to successfully clear infection at a time of rapidly emerging multi-resistance of M. tuberculosis against conventional antibiotics.
Collapse
Affiliation(s)
- Günter Weiss
- Department of Internal Medicine VI, Infectious Disease, Immunology, Rheumatology, Pneumology, Medical University of InnsbruckInnsbruck, Austria
| | - Ulrich E Schaible
- Cellular Microbiology, Priority Area Infections, Research Center BorstelBorstel, Germany
- Department of Immunology, London School of Hygiene and Tropical MedicineLondon, UK
- German Centre of Infection Research, TTU-TBBorstel, Germany
| |
Collapse
|
181
|
Kumar M, Sahu SK, Kumar R, Subuddhi A, Maji RK, Jana K, Gupta P, Raffetseder J, Lerm M, Ghosh Z, van Loo G, Beyaert R, Gupta UD, Kundu M, Basu J. MicroRNA let-7 modulates the immune response to Mycobacterium tuberculosis infection via control of A20, an inhibitor of the NF-κB pathway. Cell Host Microbe 2015; 17:345-356. [PMID: 25683052 DOI: 10.1016/j.chom.2015.01.007] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 11/20/2014] [Accepted: 01/08/2015] [Indexed: 12/31/2022]
Abstract
The outcome of the interaction between Mycobacterium tuberculosis (Mtb) and a macrophage depends on the interplay between host defense and bacterial immune subversion mechanisms. MicroRNAs critically regulate several host defense mechanisms, but their role in the Mtb-macrophage interplay remains unclear. MicroRNA profiling of Mtb-infected macrophages revealed the downregulation of miR-let-7f in a manner dependent on the Mtb secreted effector ESAT-6. We establish that let-7f targets A20, a feedback inhibitor of the NF-κB pathway. Expression of let-7f decreases and A20 increases with progression of Mtb infection in mice. Mtb survival is attenuated in A20-deficient macrophages, and the production of TNF, IL-1β, and nitrite, which are mediators of immunity to Mtb, is correspondingly increased. Further, let-7f overexpression diminishes Mtb survival and augments the production of cytokines including TNF and IL-1β. These results uncover a role for let-7f and its target A20 in regulating immune responses to Mtb and controlling bacterial burden.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Chemistry, Bose Institute, Kolkata 700009, India
| | | | - Ranjeet Kumar
- Department of Chemistry, Bose Institute, Kolkata 700009, India
| | | | | | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | - Pushpa Gupta
- National Jalma Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra 282006, India
| | - Johanna Raffetseder
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, SE-581 85 Linköping, Sweden
| | - Maria Lerm
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, SE-581 85 Linköping, Sweden
| | - Zhumur Ghosh
- Bioinformatics Centre, Bose Institute, Kolkata 700054, India
| | - Geert van Loo
- VIB Inflammation Research Center, Ghent University, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Rudi Beyaert
- VIB Inflammation Research Center, Ghent University, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Umesh D Gupta
- National Jalma Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra 282006, India
| | | | - Joyoti Basu
- Department of Chemistry, Bose Institute, Kolkata 700009, India.
| |
Collapse
|
182
|
Chen Y, Fu LL, Wen X, Liu B, Huang J, Wang JH, Wei YQ. Oncogenic and tumor suppressive roles of microRNAs in apoptosis and autophagy. Apoptosis 2015; 19:1177-89. [PMID: 24850099 DOI: 10.1007/s10495-014-0999-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs), small and non-coding endogenous RNAs ∼22 nucleotides (nt) in length, have been known to regulate approximately 30 % of human gene expression at the post-transcriptional and translational levels. Accumulating data have demonstrated that certain miRNAs could exert an oncogenic and/or tumor suppressive function and might play essential roles in the regulation of apoptosis and autophagy in cancer. In this review, we summarize that certain oncogenic and tumor suppressive miRNAs could modulate apoptotic pathways in different types of cancer. Subsequently, we demonstrate that other miRNAs might play regulatory roles in the autophagic pathways of cancer. A limited number of oncogenic/tumor suppressive miRNAs could regulate apoptosis and autophagy, respectively, and cooperatively. Taken together, these findings would provide a new clue to elucidate more apoptotic and/or autophagic mechanisms of miRNAs for designing potential novel therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Y Chen
- State Key Laboratory of Biotherapy, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | | | | | | | | | | | | |
Collapse
|
183
|
Mesenchymal stem cells detect and defend against gammaherpesvirus infection via the cGAS-STING pathway. Sci Rep 2015; 5:7820. [PMID: 25592282 PMCID: PMC4296288 DOI: 10.1038/srep07820] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 11/27/2014] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are widely used in clinical settings to treat tissue injuries and autoimmune disorders due to their multipotentiality and immunomodulation. Long-term observations reveal several complications after MSCs infusion, especially herpesviral infection. However, the mechanism of host defense against herpesviruses in MSCs remains largely unknown. Here we showed that murine gammaherpesvirus-68 (MHV-68), which is genetically and biologically related to human gammaherpesviruses, efficiently infected MSCs both in vitro and in vivo. Cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) was identified as the sensor of MHV-68 in MSCs for the first time. Moreover, the cytosolic DNA sensing pathway mediated a potent anti-herpesviral effect through the adaptor STING and downstream kinase TBK1. Furthermore, blockade of IFN signaling suggested that cytosolic DNA sensing triggered both IFN-dependent and -independent anti-herpesviral responses. Our findings demonstrate that cGAS-STING mediates innate immunity to gammaherpesvirus infection in MSCs, which may provide a clue to develop therapeutic strategy.
Collapse
|
184
|
Fu YR, Gao KS, Ji R, Yi ZJ. Differential transcriptional response in macrophages infected with cell wall deficient versus normal Mycobacterium Tuberculosis. Int J Biol Sci 2015; 11:22-30. [PMID: 25552926 PMCID: PMC4278251 DOI: 10.7150/ijbs.10217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/13/2014] [Indexed: 02/06/2023] Open
Abstract
Host-pathogen interactions determine the outcome following infection by mycobacterium tuberculosis (Mtb). Under adverse circumstances, normal Mtb can form cell-wall deficient (CWD) variants within macrophages, which have been considered an adaptive strategy for facilitating bacterial survival inside macrophages. However, the molecular mechanism by which infection of macrophages with different phenotypic Mtb elicits distinct responses of macrophages is not fully understood. To explore the molecular events triggered upon Mtb infection of macrophages, differential transcriptional responses of RAW264.7 cells infected with two forms of Mtb, CWD-Mtb and normal Mtb, were studied by microarray analysis. Some of the differentially regulated genes were confirmed by RT-qPCR in both RAW264.7 cells and primary macrophages. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway was used to analyze functions of differentially expressed genes. Distinct gene expression patterns were observed between CWD-Mtb and normal Mtb group. Mapt was up-regulated, while NOS2 and IL-11 were down-regulated in CWD-Mtb infected RAW264.7 cells and primary macrophages compared with normal Mtb infected ones. Many deregulated genes were found to be related to macrophages activation, immune response, phagosome maturation, autophagy and lipid metabolism. KEGG analysis showed that the differentially expressed genes were mainly involved in MAPK signaling pathway, nitrogen metabolism, cytokine-cytokine receptor interaction and focal adhesion. Taken together, the present study showed that differential macrophage responses were induced by intracellular CWD-Mtb an normal Mtb infection, which suggested that interactions between macrophages and different phenotypic Mtb are very complex. The results provide evidence for further understanding of pathogenesis of CWD-Mtb and may help in improving strategies to eliminate intracellular CWD-Mtb.
Collapse
Affiliation(s)
- Yu-Rong Fu
- 1. Department of Laboratory Medicine of Affiliated Hospital of Weifang Medical University, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong and Medical Priority Speciality of Clinical Laboratory in Shandong Province, Weifang Medical University, Weifang 261031, China; ; 2. Department of Medical Microbiology, Weifang Medical University, Weifang 261053, China
| | - Kun-Shan Gao
- 1. Department of Laboratory Medicine of Affiliated Hospital of Weifang Medical University, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong and Medical Priority Speciality of Clinical Laboratory in Shandong Province, Weifang Medical University, Weifang 261031, China
| | - Rui Ji
- 2. Department of Medical Microbiology, Weifang Medical University, Weifang 261053, China
| | - Zheng-Jun Yi
- 1. Department of Laboratory Medicine of Affiliated Hospital of Weifang Medical University, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong and Medical Priority Speciality of Clinical Laboratory in Shandong Province, Weifang Medical University, Weifang 261031, China
| |
Collapse
|
185
|
MicroRNA-423-5p Promotes Autophagy in Cancer Cells and Is Increased in Serum From Hepatocarcinoma Patients Treated With Sorafenib. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e233. [DOI: 10.1038/mtna.2015.8] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/11/2015] [Indexed: 02/06/2023]
|
186
|
Yu C, Xue J, Zhu W, Jiao Y, Zhang S, Cao J. Warburg meets non-coding RNAs: the emerging role of ncRNA in regulating the glucose metabolism of cancer cells. Tumour Biol 2014; 36:81-94. [DOI: 10.1007/s13277-014-2875-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 11/18/2014] [Indexed: 12/26/2022] Open
|
187
|
Ni B, Rajaram MVS, Lafuse WP, Landes MB, Schlesinger LS. Mycobacterium tuberculosis Decreases Human Macrophage IFN-γ Responsiveness through miR-132 and miR-26a. THE JOURNAL OF IMMUNOLOGY 2014. [DOI: 10.4049/jimmunol.1400124 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
IFN-γ–activated macrophages play an essential role in controlling intracellular pathogens; however, macrophages also serve as the cellular home for the intracellular pathogen Mycobacterium tuberculosis. Based on previous evidence that M. tuberculosis can modulate host microRNA (miRNA) expression, we examined the miRNA expression profile of M. tuberculosis–infected primary human macrophages. We identified 31 differentially expressed miRNAs in primary human macrophages during M. tuberculosis infection by NanoString and confirmed our findings by quantitative real-time RT-PCR. In addition, we determined a role for two miRNAs upregulated upon M. tuberculosis infection, miR-132 and miR-26a, as negative regulators of transcriptional coactivator p300, a component of the IFN-γ signaling cascade. Knockdown expression of miR-132 and miR-26a increased p300 protein levels and improved transcriptional, translational, and functional responses to IFN-γ in human macrophages. Collectively, these data validate p300 as a target of miR-132 and miR-26a, and demonstrate a mechanism by which M. tuberculosis can limit macrophage responses to IFN-γ by altering host miRNA expression.
Collapse
Affiliation(s)
- Bin Ni
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Murugesan V. S. Rajaram
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - William P. Lafuse
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Michelle B. Landes
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Larry S. Schlesinger
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
188
|
Ni B, Rajaram MVS, Lafuse WP, Landes MB, Schlesinger LS. Mycobacterium tuberculosis Decreases Human Macrophage IFN-γ Responsiveness through miR-132 and miR-26a. THE JOURNAL OF IMMUNOLOGY 2014. [DOI: 10.4049/jimmunol.1400124 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
IFN-γ–activated macrophages play an essential role in controlling intracellular pathogens; however, macrophages also serve as the cellular home for the intracellular pathogen Mycobacterium tuberculosis. Based on previous evidence that M. tuberculosis can modulate host microRNA (miRNA) expression, we examined the miRNA expression profile of M. tuberculosis–infected primary human macrophages. We identified 31 differentially expressed miRNAs in primary human macrophages during M. tuberculosis infection by NanoString and confirmed our findings by quantitative real-time RT-PCR. In addition, we determined a role for two miRNAs upregulated upon M. tuberculosis infection, miR-132 and miR-26a, as negative regulators of transcriptional coactivator p300, a component of the IFN-γ signaling cascade. Knockdown expression of miR-132 and miR-26a increased p300 protein levels and improved transcriptional, translational, and functional responses to IFN-γ in human macrophages. Collectively, these data validate p300 as a target of miR-132 and miR-26a, and demonstrate a mechanism by which M. tuberculosis can limit macrophage responses to IFN-γ by altering host miRNA expression.
Collapse
Affiliation(s)
- Bin Ni
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Murugesan V. S. Rajaram
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - William P. Lafuse
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Michelle B. Landes
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Larry S. Schlesinger
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
189
|
Ni B, Rajaram MVS, Lafuse WP, Landes MB, Schlesinger LS. Mycobacterium tuberculosis Decreases Human Macrophage IFN-γ Responsiveness through miR-132 and miR-26a. THE JOURNAL OF IMMUNOLOGY 2014. [DOI: 10.4049/jimmunol.1400124 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
IFN-γ–activated macrophages play an essential role in controlling intracellular pathogens; however, macrophages also serve as the cellular home for the intracellular pathogen Mycobacterium tuberculosis. Based on previous evidence that M. tuberculosis can modulate host microRNA (miRNA) expression, we examined the miRNA expression profile of M. tuberculosis–infected primary human macrophages. We identified 31 differentially expressed miRNAs in primary human macrophages during M. tuberculosis infection by NanoString and confirmed our findings by quantitative real-time RT-PCR. In addition, we determined a role for two miRNAs upregulated upon M. tuberculosis infection, miR-132 and miR-26a, as negative regulators of transcriptional coactivator p300, a component of the IFN-γ signaling cascade. Knockdown expression of miR-132 and miR-26a increased p300 protein levels and improved transcriptional, translational, and functional responses to IFN-γ in human macrophages. Collectively, these data validate p300 as a target of miR-132 and miR-26a, and demonstrate a mechanism by which M. tuberculosis can limit macrophage responses to IFN-γ by altering host miRNA expression.
Collapse
Affiliation(s)
- Bin Ni
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Murugesan V. S. Rajaram
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - William P. Lafuse
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Michelle B. Landes
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Larry S. Schlesinger
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
190
|
Ni B, Rajaram MVS, Lafuse WP, Landes MB, Schlesinger LS. Mycobacterium tuberculosis Decreases Human Macrophage IFN-γ Responsiveness through miR-132 and miR-26a. THE JOURNAL OF IMMUNOLOGY 2014. [DOI: 10.4049/jimmunol.1400124 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
IFN-γ–activated macrophages play an essential role in controlling intracellular pathogens; however, macrophages also serve as the cellular home for the intracellular pathogen Mycobacterium tuberculosis. Based on previous evidence that M. tuberculosis can modulate host microRNA (miRNA) expression, we examined the miRNA expression profile of M. tuberculosis–infected primary human macrophages. We identified 31 differentially expressed miRNAs in primary human macrophages during M. tuberculosis infection by NanoString and confirmed our findings by quantitative real-time RT-PCR. In addition, we determined a role for two miRNAs upregulated upon M. tuberculosis infection, miR-132 and miR-26a, as negative regulators of transcriptional coactivator p300, a component of the IFN-γ signaling cascade. Knockdown expression of miR-132 and miR-26a increased p300 protein levels and improved transcriptional, translational, and functional responses to IFN-γ in human macrophages. Collectively, these data validate p300 as a target of miR-132 and miR-26a, and demonstrate a mechanism by which M. tuberculosis can limit macrophage responses to IFN-γ by altering host miRNA expression.
Collapse
Affiliation(s)
- Bin Ni
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Murugesan V. S. Rajaram
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - William P. Lafuse
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Michelle B. Landes
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Larry S. Schlesinger
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
191
|
Ni B, Rajaram MVS, Lafuse WP, Landes MB, Schlesinger LS. Mycobacterium tuberculosis Decreases Human Macrophage IFN-γ Responsiveness through miR-132 and miR-26a. THE JOURNAL OF IMMUNOLOGY 2014. [DOI: 10.4049/jimmunol.1400124 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
IFN-γ–activated macrophages play an essential role in controlling intracellular pathogens; however, macrophages also serve as the cellular home for the intracellular pathogen Mycobacterium tuberculosis. Based on previous evidence that M. tuberculosis can modulate host microRNA (miRNA) expression, we examined the miRNA expression profile of M. tuberculosis–infected primary human macrophages. We identified 31 differentially expressed miRNAs in primary human macrophages during M. tuberculosis infection by NanoString and confirmed our findings by quantitative real-time RT-PCR. In addition, we determined a role for two miRNAs upregulated upon M. tuberculosis infection, miR-132 and miR-26a, as negative regulators of transcriptional coactivator p300, a component of the IFN-γ signaling cascade. Knockdown expression of miR-132 and miR-26a increased p300 protein levels and improved transcriptional, translational, and functional responses to IFN-γ in human macrophages. Collectively, these data validate p300 as a target of miR-132 and miR-26a, and demonstrate a mechanism by which M. tuberculosis can limit macrophage responses to IFN-γ by altering host miRNA expression.
Collapse
Affiliation(s)
- Bin Ni
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Murugesan V. S. Rajaram
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - William P. Lafuse
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Michelle B. Landes
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Larry S. Schlesinger
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
192
|
MicroRNA-155 induction by Mycobacterium bovis BCG enhances ROS production through targeting SHIP1. Mol Immunol 2014; 62:29-36. [DOI: 10.1016/j.molimm.2014.05.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/20/2014] [Accepted: 05/27/2014] [Indexed: 01/12/2023]
|
193
|
Ni B, Rajaram MVS, Lafuse WP, Landes MB, Schlesinger LS. Mycobacterium tuberculosis Decreases Human Macrophage IFN-γ Responsiveness through miR-132 and miR-26a. THE JOURNAL OF IMMUNOLOGY 2014. [DOI: 10.4049/jimmunol.1400124 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
IFN-γ–activated macrophages play an essential role in controlling intracellular pathogens; however, macrophages also serve as the cellular home for the intracellular pathogen Mycobacterium tuberculosis. Based on previous evidence that M. tuberculosis can modulate host microRNA (miRNA) expression, we examined the miRNA expression profile of M. tuberculosis–infected primary human macrophages. We identified 31 differentially expressed miRNAs in primary human macrophages during M. tuberculosis infection by NanoString and confirmed our findings by quantitative real-time RT-PCR. In addition, we determined a role for two miRNAs upregulated upon M. tuberculosis infection, miR-132 and miR-26a, as negative regulators of transcriptional coactivator p300, a component of the IFN-γ signaling cascade. Knockdown expression of miR-132 and miR-26a increased p300 protein levels and improved transcriptional, translational, and functional responses to IFN-γ in human macrophages. Collectively, these data validate p300 as a target of miR-132 and miR-26a, and demonstrate a mechanism by which M. tuberculosis can limit macrophage responses to IFN-γ by altering host miRNA expression.
Collapse
Affiliation(s)
- Bin Ni
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Murugesan V. S. Rajaram
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - William P. Lafuse
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Michelle B. Landes
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Larry S. Schlesinger
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
194
|
Ni B, Rajaram MVS, Lafuse WP, Landes MB, Schlesinger LS. Mycobacterium tuberculosis Decreases Human Macrophage IFN-γ Responsiveness through miR-132 and miR-26a. THE JOURNAL OF IMMUNOLOGY 2014. [DOI: 10.4049/jimmunol.1400124 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Abstract
IFN-γ–activated macrophages play an essential role in controlling intracellular pathogens; however, macrophages also serve as the cellular home for the intracellular pathogen Mycobacterium tuberculosis. Based on previous evidence that M. tuberculosis can modulate host microRNA (miRNA) expression, we examined the miRNA expression profile of M. tuberculosis–infected primary human macrophages. We identified 31 differentially expressed miRNAs in primary human macrophages during M. tuberculosis infection by NanoString and confirmed our findings by quantitative real-time RT-PCR. In addition, we determined a role for two miRNAs upregulated upon M. tuberculosis infection, miR-132 and miR-26a, as negative regulators of transcriptional coactivator p300, a component of the IFN-γ signaling cascade. Knockdown expression of miR-132 and miR-26a increased p300 protein levels and improved transcriptional, translational, and functional responses to IFN-γ in human macrophages. Collectively, these data validate p300 as a target of miR-132 and miR-26a, and demonstrate a mechanism by which M. tuberculosis can limit macrophage responses to IFN-γ by altering host miRNA expression.
Collapse
Affiliation(s)
- Bin Ni
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Murugesan V. S. Rajaram
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - William P. Lafuse
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Michelle B. Landes
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| | - Larry S. Schlesinger
- *Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
- †Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210; and
- ‡Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
195
|
Rajaram MVS, Ni B, Dodd CE, Schlesinger LS. Macrophage immunoregulatory pathways in tuberculosis. Semin Immunol 2014; 26:471-85. [PMID: 25453226 DOI: 10.1016/j.smim.2014.09.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/17/2022]
Abstract
Macrophages, the major host cells harboring Mycobacterium tuberculosis (M.tb), are a heterogeneous cell type depending on their tissue of origin and host they are derived from. Significant discord in macrophage responses to M.tb exists due to differences in M.tb strains and the various types of macrophages used to study tuberculosis (TB). This review will summarize current concepts regarding macrophage responses to M.tb infection, while pointing out relevant differences in experimental outcomes due to the use of divergent model systems. A brief description of the lung environment is included since there is increasing evidence that the alveolar macrophage (AM) has immunoregulatory properties that can delay optimal protective host immune responses. In this context, this review focuses on selected macrophage immunoregulatory pattern recognition receptors (PRRs), cytokines, negative regulators of inflammation, lipid mediators and microRNAs (miRNAs).
Collapse
Affiliation(s)
- Murugesan V S Rajaram
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Bin Ni
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Claire E Dodd
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
196
|
Moraco AH, Kornfeld H. Cell death and autophagy in tuberculosis. Semin Immunol 2014; 26:497-511. [PMID: 25453227 DOI: 10.1016/j.smim.2014.10.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 09/30/2014] [Accepted: 10/01/2014] [Indexed: 12/13/2022]
Abstract
Mycobacterium tuberculosis has succeeded in infecting one-third of the human race though inhibition or evasion of innate and adaptive immunity. The pathogen is a facultative intracellular parasite that uses the niche provided by mononuclear phagocytes for its advantage. Complex interactions determine whether the bacillus will or will not be delivered to acidified lysosomes, whether the host phagocyte will survive infection or die, and whether the timing and mode of cell death works to the advantage of the host or the pathogen. Here we discuss cell death and autophagy in TB. These fundamental processes of cell biology feature in all aspects of TB pathogenesis and may be exploited to the treatment or prevention of TB disease.
Collapse
Affiliation(s)
- Andrew H Moraco
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Hardy Kornfeld
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
197
|
Wang J, Huang C, Wu M, Zhong Q, Yang K, Li M, Zhan X, Wen J, Zhou L, Huang X. MRP8/14 induces autophagy to eliminate intracellular Mycobacterium bovis BCG. J Infect 2014; 70:415-26. [PMID: 25312864 DOI: 10.1016/j.jinf.2014.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 08/30/2014] [Accepted: 09/15/2014] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To explore the role of myeloid-related protein 8/14 in mycobacterial infection. METHODS The mRNA and protein expression levels of MRP8 or MRP14 were measured by real-time PCR and flow cytometry, respectively. Role of MRP8/14 was tested by overexpression or RNA interference assays. Flow cytometry and colony forming unit were used to test the phagocytosis and the survival of intracellular Mycobacterium bovis BCG (BCG), respectively. Autophagy mediated by MRP8/14 was detected by Western blot and immunofluorescence. The colocalization of BCG phagosomes with autophagosomes or lysosomes was by detected by confocal microscopy. ROS production was detected by flow cytometry. RESULTS MRP8/14 expressions were up-regulated in human monocytic THP1 cells and primary macrophages after mycobacterial challenge. Silencing of MRP8/14 suppressed bacterial killing, but had no influence on the phagocytosis of BCG. Importantly, silencing MRP8/14 decreased autophagy and BCG phagosome maturation in THP1-derived macrophages, thereby increasing the BCG survival. Additionally, we demonstrated that MRP8/14 promoted autophagy in a ROS-dependent manner. CONCLUSIONS The present study revealed a novel role of MRP8/14 in the autophagy-mediated elimination of intracellular BCG by promoting ROS generation, which may provide a promising therapeutic target for tuberculosis and other intracellular bacterial infectious diseases.
Collapse
Affiliation(s)
- Jinli Wang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Department of Laboratory Medicine, Guangzhou First Municipal People's Hospital, Affiliated Hospital of Guangzhou Medical University, Guangzhou 510500, China
| | - Chunyu Huang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, China
| | - Minhao Wu
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Qiu Zhong
- Center for Tuberculosis Control of Guangdong Province, Guangzhou 510630, China
| | - Kun Yang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Miao Li
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Xiaoxia Zhan
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Jinsheng Wen
- Department of Microbiology and Immunology, Wenzhou Medical University, Wenzhou 325035, China
| | - Lin Zhou
- Center for Tuberculosis Control of Guangdong Province, Guangzhou 510630, China.
| | - Xi Huang
- Department of Immunology, Institute of Tuberculosis Control, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Key Laboratory of Tropical Diseases Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Department of Microbiology and Immunology, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
198
|
Martin EC, Rhodes LV, Elliott S, Krebs AE, Nephew KP, Flemington EK, Collins-Burow BM, Burow ME. microRNA regulation of mammalian target of rapamycin expression and activity controls estrogen receptor function and RAD001 sensitivity. Mol Cancer 2014; 13:229. [PMID: 25283550 PMCID: PMC4203920 DOI: 10.1186/1476-4598-13-229] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 09/24/2014] [Indexed: 01/28/2023] Open
Abstract
Background The AKT/mammalian target of rapamycin (mTOR) signaling pathway is regulated by 17α-estradiol (E2) signaling and mediates E2-induced proliferation and progesterone receptor (PgR) expression in breast cancer. Methods and results Here we use deep sequencing analysis of previously published data from The Cancer Genome Atlas to demonstrate that expression of a key component of mTOR signaling, rapamycin-insensitive companion of mTOR (Rictor), positively correlated with an estrogen receptor-α positive (ERα+) breast tumor signature. Through increased microRNA-155 (miR-155) expression in the ERα+ breast cancer cells we demonstrate repression of Rictor enhanced activation of mTOR complex 1 (mTORC1) signaling with both qPCR and western blot. miR-155-mediated mTOR signaling resulted in deregulated ERα signaling both in cultured cells in vitro and in xenografts in vivo in addition to repressed PgR expression and activity. Furthermore we observed that miR-155 enhanced mTORC1 signaling (observed through western blot for increased phosphorylation on mTOR S2448) and induced inhibition of mTORC2 signaling (evident through repressed Rictor and tuberous sclerosis 1 (TSC1) gene expression). mTORC1 induced deregulation of E2 signaling was confirmed using qPCR and the mTORC1-specific inhibitor RAD001. Co-treatment of MCF7 breast cancer cells stably overexpressing miR-155 with RAD001 and E2 restored E2-induced PgR gene expression. RAD001 treatment of SCID/CB17 mice inhibited E2-induced tumorigenesis of the MCF7 miR-155 overexpressing cell line. Finally we demonstrated a strong positive correlation between Rictor and PgR expression and a negative correlation with Raptor expression in Luminal B breast cancer samples, a breast cancer histological subtype known for having an altered ERα-signaling pathway. Conclusions miRNA mediated alterations in mTOR and ERα signaling establishes a new mechanism for altered estrogen responses independent of growth factor stimulation. Electronic supplementary material The online version of this article (doi:10.1186/1476-4598-13-229) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Matthew E Burow
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
199
|
Ueberberg B, Kohns M, Mayatepek E, Jacobsen M. Are microRNAs suitable biomarkers of immunity to tuberculosis? Mol Cell Pediatr 2014; 1:8. [PMID: 26567102 PMCID: PMC4530568 DOI: 10.1186/s40348-014-0008-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 09/09/2014] [Indexed: 11/23/2022] Open
Abstract
Background MicroRNAs (miRNAs) are crucial regulators of human immunity e.g. against Mycobacterium tuberculosis. Against the background of still alarming high mortality of tuberculosis effective biomarkers to improve diagnosis of M. tuberculosis infection and successful treatment are of major importance. Conclusions This review summarizes recent surrogate tissue studies for identification of miRNA biomarker candidates in human tuberculosis with a special focus on reproducibility and conformance. In addition we provide assistance for the design of biomarker studies to circumvent major pitfalls.
Collapse
Affiliation(s)
- Bianca Ueberberg
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany.
| | - Malte Kohns
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany.
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany.
| | - Marc Jacobsen
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany.
| |
Collapse
|
200
|
MicroRNA expression profiling of PPD-B stimulated PBMC from M. bovis-challenged unvaccinated and BCG vaccinated cattle. Vaccine 2014; 32:5839-44. [DOI: 10.1016/j.vaccine.2014.07.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/09/2014] [Accepted: 07/09/2014] [Indexed: 12/13/2022]
|