1
|
Xiong D, Gong M, Hou Y, Chen H, Gao T, He L. Euphorbia helioscopia L. extract suppresses hepatitis B virus-related hepatocellular carcinoma via alpha serine/threonine-protein kinase and Caspase-3. J Cancer Res Clin Oncol 2024; 150:442. [PMID: 39356361 PMCID: PMC11446964 DOI: 10.1007/s00432-024-05972-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND Hepatitis B virus (HBV)-related hepatocellular carcinoma (HBV-HCC) has poor prognosis and high mortality rate. Euphorbia helioscopia L. (EHL) is a classic Chinese medicinal herb. AIM This study aimed to evaluate in vitro anti-HBV-HCC properties of EHL, and explore it targets in HBV-HCC based on molecular docking. METHODS The anti-tumor effect of EHL on HBV-HCC was evaluated using the cell viability, migration, invasion, and apoptosis of Hep 3B2.1-7 and HepG2.2.15 cells. Next, network pharmacological analysis was performed to predicted the key targets of EHL against HBV-HCC. Then the prognostic targets, including RAC-alpha serine/threonine-protein kinase (AKT1) and Caspase-3 (CASP3), were verified using molecular docking and rescue experiments. RESULTS EHL exhibited inhibitory effects on cell proliferation/migration/invasion and induced cell apoptosis. Network pharmacological analysis proposed 12 active compounds in EHL, which targeted 22 HBV-HCC-related genes. AKT1 and CASP3 were identified to be key targets for EHL against HBV-HCC. AKT1 and CASP3 had prognostic significance in liver cancer. Overexpression of AKT1 and caspase-3 inhibitor can counteract the EHL effect. CONCLUSION EHL can exert anticancer effects on HBV-HCC by inhibiting migration/invasion, and inducing apoptosis, which may be achieved through AKT1 and CASP3.
Collapse
Affiliation(s)
- Dan Xiong
- Department of Psychiatry 2, The Fifth People's Hospital of Jiujiang, Jiujiang, 332000, China
| | - Minyong Gong
- Department of Oncology, Hospital of Traditional Chinese Medicine of Jiujiang, No. 555, Dehua Road, Wuli Street, Lianxi District, Jiujiang City, 332000, Jiangxi Province, China
| | - Yanjun Hou
- Department of Oncology, Hospital of Traditional Chinese Medicine of Jiujiang, No. 555, Dehua Road, Wuli Street, Lianxi District, Jiujiang City, 332000, Jiangxi Province, China
| | - Haibing Chen
- Department of Psychiatry 2, The Fifth People's Hospital of Jiujiang, Jiujiang, 332000, China
| | - Tiexin Gao
- Department of Oncology, Hospital of Traditional Chinese Medicine of Jiujiang, No. 555, Dehua Road, Wuli Street, Lianxi District, Jiujiang City, 332000, Jiangxi Province, China
| | - Liuxin He
- Department of Oncology, Hospital of Traditional Chinese Medicine of Jiujiang, No. 555, Dehua Road, Wuli Street, Lianxi District, Jiujiang City, 332000, Jiangxi Province, China.
| |
Collapse
|
2
|
Zhang XY, Li YY, Huang HX, Zhao CC, Lei XX, Zhao BP, Lu JY, Lan T, Sun WC. Seneca Valley virus 3C pro antagonizes host innate immune responses and programmed cell death. Front Microbiol 2023; 14:1235620. [PMID: 37869659 PMCID: PMC10588452 DOI: 10.3389/fmicb.2023.1235620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
Seneca Valley virus (SVV), a member of the Picornaviridae family, may cause serious water blister diseases in pregnant sows and acute death in newborn piglets, which have resulted in economic losses in pig production. The 3C protease is a vital enzyme for SVV maturation and is capable of regulating protein cleavage and RNA replication of the virus. Additionally, this protease can impede the host's innate immune response by targeting the interferon pathway's principal factor and enhance virus replication by modulating the host's RNA metabolism while simultaneously triggering programmed cell death. This article reviews recent studies on SVV 3C functions, which include viral replication promotion, cell apoptosis modulation and host immune response evasion, and provides a theoretical basis for research on preventing and controlling SVV infection.
Collapse
Affiliation(s)
- Xin-yu Zhang
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Yu-ying Li
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Hai-xin Huang
- Institute of Virology, Wenzhou University, Wenzhou, China
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Chen-chen Zhao
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Xiao-xiao Lei
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Bao-peng Zhao
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Jing-yi Lu
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Tian Lan
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Wen-chao Sun
- Institute of Virology, Wenzhou University, Wenzhou, China
| |
Collapse
|
3
|
Touil Y, Latreche-Carton C, Bouazzati HE, Nugues AL, Jouy N, Thuru X, Laine W, Lepretre F, Figeac M, Tardivel M, Kluza J, Idziorek T, Quesnel B. p65/RelA NF-κB fragments generated by RIPK3 activity regulate tumorigenicity, cell metabolism, and stemness characteristics. J Cell Biochem 2021; 123:543-556. [PMID: 34927768 PMCID: PMC9299825 DOI: 10.1002/jcb.30198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/17/2021] [Accepted: 12/10/2021] [Indexed: 12/16/2022]
Abstract
Receptor‐interacting protein kinase 3 (RIPK3) can induce necroptosis, apoptosis, or cell proliferation and is silenced in several hematological malignancies. We previously reported that RIPK3 activity independent of its kinase domain induces caspase‐mediated p65/RelA cleavage, resulting in N‐terminal 1‐361 and C‐terminal 362‐549 fragments. We show here that a noncleavable p65/RelA D361E mutant expressed in DA1‐3b leukemia cells decreases mouse survival times and that coexpression of p65/RelA fragments increases the tumorigenicity of B16F1 melanoma cells. This aggressiveness in vivo did not correlate with NF‐κB activity measured in vitro. The fragments and p65/RelA D361E mutant induced different expression profiles in DA1‐3b and B16F1 cells. Stemness markers were affected: p65/RelA D361E increased ALDH activity in DA1‐3b cells, and fragment expression increased melanoma sphere formation in B16/F1 cells. p65/RelA fragments and the D361E noncleavable mutant decreased oxidative or glycolytic cell metabolism, with differences observed between models. Thus, p65/RelA cleavage initiated by kinase‐independent RIPK3 activity in cancer cells is not neutral and induces pleiotropic effects in vitro and in vivo that may vary across tumor types.
Collapse
Affiliation(s)
- Yasmine Touil
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Céline Latreche-Carton
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Hassiba El Bouazzati
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Anne-Lucie Nugues
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Nathalie Jouy
- UMS 2014 CNRS/US 41 Inserm, University of Lille, Lille, France
| | - Xavier Thuru
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - William Laine
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | | | - Martin Figeac
- UMS 2014 CNRS/US 41 Inserm, University of Lille, Lille, France
| | - Meryem Tardivel
- UMS 2014 CNRS/US 41 Inserm, University of Lille, Lille, France
| | - Jérôme Kluza
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Thierry Idziorek
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Bruno Quesnel
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France.,Service des Maladies du Sang, CHU Lille, Lille, France
| |
Collapse
|
4
|
Kıyga E, Şengelen A, Adıgüzel Z, Önay Uçar E. Investigation of the role of quercetin as a heat shock protein inhibitor on apoptosis in human breast cancer cells. Mol Biol Rep 2020; 47:4957-4967. [PMID: 32638319 DOI: 10.1007/s11033-020-05641-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
High expression of heat shock proteins (Hsp) in breast cancer has been closely associated with tumor cell proliferation and thus a poor clinical outcome. Quercetin, a good Hsp inhibitor as a dietary flavonoid, possesses anticarcinogenic properties. Although there are many studies on the effects of quercetin on Hsp levels in human breast cancer cells, research on elucidation of its molecular mechanism continues. Herein, we aimed to investigate the effect of quercetin on Hsp levels and whether quercetin is a suitable therapeutic for two breast cancer cell lines (MCF-7 and MDA-MB-231) representing breast tumors which differed in hormone receptor, aggressiveness and treatment responses. To examine the response to high and low doses of quercetin, the cells were treated with three doses of quercetin (10, 25 and 100 μM) determined by MTT. The effects of quercetin on Hsp levels, apoptosis and DNA damage were examined by western blot analysis, caspase activity assay, comet assay and microscopy in human breast cancer cells. Compared to MDA-MB231 cells, MCF-7 cells were more affected by quercetin treatments. Quercetin effectively suppressed the expression of Hsp27, Hsp70 and Hsp90. While quercetin did not induce DNA damage, it triggered apoptosis at high levels. Although an increase in NF-κB levels is observed in the cells exposed to quercetin, the net result is the anticancer effect in case of Hsp depletion and apoptosis induction. Taken together our findings suggested that quercetin can be an effective therapeutic agent for breast cancer therapy regardless of the presence or absence of hormone receptors.
Collapse
Affiliation(s)
- Ezgi Kıyga
- Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkey
| | - Aslıhan Şengelen
- Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkey
| | - Zelal Adıgüzel
- Basic Medical Sciences Department of Molecular Biology and Genetics, School of Medicine, Koç University, Istanbul, Turkey
| | - Evren Önay Uçar
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Turkey.
| |
Collapse
|
5
|
Fernandes MHV, Maggioli MF, Otta J, Joshi LR, Lawson S, Diel DG. Senecavirus A 3C Protease Mediates Host Cell Apoptosis Late in Infection. Front Immunol 2019; 10:363. [PMID: 30918505 PMCID: PMC6424860 DOI: 10.3389/fimmu.2019.00363] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/12/2019] [Indexed: 12/22/2022] Open
Abstract
Senecavirus A (SVA), an oncolytic picornavirus used for cancer treatment in humans, has recently emerged as a vesicular disease (VD)-causing agent in swine worldwide. Notably, SVA-induced VD is indistinguishable from foot-and-mouth disease (FMD) and other high-consequence VDs of pigs. Here we investigated the role of apoptosis on infection and replication of SVA. Given the critical role of the nuclear factor-kappa B (NF-κB) signaling pathway on modulation of cell death, we first assessed activation of NF-κB during SVA infection. Results here show that while early during infection SVA induces activation of NF-κB, as evidenced by nuclear translocation of NF-κB-p65 and NF-κB-mediated transcription, late in infection a cleaved product corresponding to the C-terminus of NF-κB-p65 is detected in infected cells, resulting in lower NF-κB transcriptional activity. Additionally, we assessed the potential role of SVA 3C protease (3Cpro) in SVA-induced host-cell apoptosis and cleavage of NF-κB-p65. Transient expression of SVA 3Cpro was associated with cleavage of NF-κB-p65 and Poly (ADP-ribose) polymerase (PARP), suggesting its involvement in virus-induced apoptosis. Most importantly, we showed that while cleavage of NF-κB-p65 is secondary to caspase activation, the proteolytic activity of SVA 3Cpro is essential for induction of apoptosis. Experiments using the pan-caspase inhibitor Z-VAD-FMK confirmed the relevance of late apoptosis for SVA infection, indicating that SVA induces apoptosis, presumably, as a mechanism to facilitate virus release and/or spread from infected cells. Together, these results suggest an important role of apoptosis for SVA infection biology.
Collapse
Affiliation(s)
| | | | | | | | | | - Diego G. Diel
- Animal Disease Research And Diagnostic Laboratory, Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD, United States
| |
Collapse
|
6
|
Niell N, Larriba MJ, Ferrer‐Mayorga G, Sánchez‐Pérez I, Cantero R, Real FX, del Peso L, Muñoz A, González‐Sancho JM. The human PKP2/plakophilin-2 gene is induced by Wnt/β-catenin in normal and colon cancer-associated fibroblasts. Int J Cancer 2018; 142:792-804. [PMID: 29044515 PMCID: PMC5765413 DOI: 10.1002/ijc.31104] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/24/2017] [Accepted: 10/04/2017] [Indexed: 12/15/2022]
Abstract
Colorectal cancer results from the malignant transformation of colonic epithelial cells. Stromal fibroblasts are the main component of the tumour microenvironment, and play an important role in the progression of this and other neoplasias. Wnt/β-catenin signalling is essential for colon homeostasis, but aberrant, constitutive activation of this pathway is a hallmark of colorectal cancer. Here we present the first transcriptomic study on the effect of a Wnt factor on human colonic myofibroblasts. Wnt3A regulates the expression of 1,136 genes, of which 662 are upregulated and 474 are downregulated in CCD-18Co cells. A set of genes encoding inhibitors of the Wnt/β-catenin pathway stand out among those induced by Wnt3A, which suggests that there is a feedback inhibitory mechanism. We also show that the PKP2 gene encoding the desmosomal protein Plakophilin-2 is a novel direct transcriptional target of Wnt/β-catenin in normal and colon cancer-associated fibroblasts. PKP2 is induced by β-catenin/TCF through three binding sites in the gene promoter and one additional binding site located in an enhancer 20 kb upstream from the transcription start site. Moreover, Plakophilin-2 antagonizes Wnt/β-catenin transcriptional activity in HEK-293T cells, which suggests that it may act as an intracellular inhibitor of the Wnt/β-catenin pathway. Our results demonstrate that stromal fibroblasts respond to canonical Wnt signalling and that Plakophilin-2 plays a role in the feedback control of this effect suggesting that the response to Wnt factors in the stroma may modulate Wnt activity in the tumour cells.
Collapse
Affiliation(s)
- Núria Niell
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) –Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Departamento de BioquímicaFacultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
| | - María Jesús Larriba
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) –Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ)MadridE‐28046Spain
- Instituto de Salud Carlos IIICIBER de Cáncer (CIBERONC)MadridSpain
| | - Gemma Ferrer‐Mayorga
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) –Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ)MadridE‐28046Spain
- Instituto de Salud Carlos IIICIBER de Cáncer (CIBERONC)MadridSpain
- Fundación de Investigación HM HospitalesMadridE‐28015Spain
| | - Isabel Sánchez‐Pérez
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) –Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Departamento de BioquímicaFacultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ)MadridE‐28046Spain
- Unidad asociada de Biomedicina UCLM‐CSICMadridSpain
- Instituto de Salud Carlos IIICIBER de Enfermedades Raras (CIBERER)MadridSpain
| | - Ramón Cantero
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ)MadridE‐28046Spain
- Department of Surgery, La Paz University HospitalColorectal UnitMadridE‐28046Spain
| | - Francisco X. Real
- Instituto de Salud Carlos IIICIBER de Cáncer (CIBERONC)MadridSpain
- Cancer Cell Biology Programme, Spanish National Cancer Research CentreEpithelial Carcinogenesis GroupMadridE‐28029Spain
- Departament de Ciències Experimentals i de la SalutUniversitat Pompeu FabraBarcelonaE‐08003Spain
| | - Luis del Peso
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) –Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Departamento de BioquímicaFacultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ)MadridE‐28046Spain
- Instituto de Salud Carlos IIICIBER de Enfermedades Respiratorias (CIBERES)MadridSpain
| | - Alberto Muñoz
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) –Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ)MadridE‐28046Spain
- Instituto de Salud Carlos IIICIBER de Cáncer (CIBERONC)MadridSpain
| | - José Manuel González‐Sancho
- Instituto de Investigaciones Biomédicas “Alberto Sols”, Consejo Superior de Investigaciones Científicas (CSIC) –Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Departamento de BioquímicaFacultad de Medicina, Universidad Autónoma de Madrid (UAM)MadridE‐28029Spain
- Instituto de Salud Carlos IIICIBER de Cáncer (CIBERONC)MadridSpain
| |
Collapse
|
7
|
LRRC25 Functions as an Inhibitor of NF-κB Signaling Pathway by Promoting p65/RelA for Autophagic Degradation. Sci Rep 2017; 7:13448. [PMID: 29044191 PMCID: PMC5647368 DOI: 10.1038/s41598-017-12573-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 08/17/2017] [Indexed: 01/05/2023] Open
Abstract
Nuclear factor κB (NF-κB) is a family of critical transcription factors that play a critical role in innate immune responses and inflammation, yet the molecular mechanisms responsible for its tight regulation is not fully understood. In this study, we identified LRRC25, a member of leucine-rich repeat (LRR)-containing protein family, as a negative regulator in the NF-κB signaling pathway. Ectopic expression of LRRC25 impaired NF-κB activation, whereas knockout of LRRC25 potentiated NF-κB activation and enhanced the production of inflammatory cytokines. Further study demonstrated that the LRR domain of LRRC25 interacted with the Rel Homology domain (RHD) of p65/RelA and promotes the degradation of p65/RelA. Furthermore, LRRC25 enhanced the interaction between p65/RelA and cargo receptor p62, thus facilitating the degradation of p65/RelA through autophagy pathway. Our study has not only identified LRRC25 as a novel inhibitor of NF-κB signaling pathway, but also uncovers a new mechanism of crosstalk between NF-κB signaling and autophagy pathways.
Collapse
|
8
|
Rice KM, Manne ND, Arvapalli R, Ginjupalli GK, Blough ER. Diabetes alters vascular mechanotransduction data: Pressure-induced regulation of Nf-kapa-B p65 and translational associated signaling in the rat inferior vena cava. Data Brief 2017; 14:676-685. [PMID: 28932772 PMCID: PMC5596213 DOI: 10.1016/j.dib.2017.08.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/14/2017] [Accepted: 08/24/2017] [Indexed: 01/21/2023] Open
Abstract
Diabetic patients have a high rate of vein graft failure due to attrition or vessel occlusion that cause recurrent ischemic events or vein graft. Veins grafted into a high-pressure arterial environment must undergo vascular remodeling to better handle the altered hemodynamics and intravascular increased pressure. Multiple cellular and molecular events are purported to be associated with vascular remodeling of veins. Understanding the effect diabetes has on vascular mechano-transductive response is critical to decreasing graft failure rates. This article represents data regarding a study published in Cardiovascular Diabetology [1] and Open Journal of Endocrine and Metabolic Diseases [2] with the purpose of evaluating the effect of pressurization on rat inferior venae cavae (IVC). Here we provide the information about the method and processing of raw data related to our prior publish work and Data in Brief articles [3], [4]. The data contained in this article evaluates the contribution of NF-kB signaling and associated proteins. IVC from lean and obese animals were exposed to a 30 min of perfusion at 120 mm Hg pressure and evaluated for changes in expression and (IkB-alpha, NF-kB p50, NF-kB p105, NF-kB p65, Traf2, caspase 12), phosphorylation of (IkB-alpha (ser 32), Fox01 (ser 256), and Fox04 (ser 193)) proteins thought to be involved in the regulation of vascular mechanotransduction.
Collapse
Affiliation(s)
- Kevin M. Rice
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA
- Department of Internal Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
- Biotechnology Graduate Program West Virginia State University, Institute, WV, USA
- Department of Health and Human Service, School of Kinesiology, Marshall University, Huntington, WV, USA
| | | | | | | | - Eric R. Blough
- Center for Diagnostic Nanosystems, Marshall University, Huntington, WV, USA
- Biotechnology Graduate Program West Virginia State University, Institute, WV, USA
- Department of Pharmaceutical Sciences and Research, School of Pharmacy, Marshall University, Huntington, WV, USA
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| |
Collapse
|
9
|
Liu Q, Zhang Z, Zheng Z, Zheng C, Liu Y, Hu Q, Ke X, Wang H. Human Bocavirus NS1 and NS1-70 Proteins Inhibit TNF-α-Mediated Activation of NF-κB by targeting p65. Sci Rep 2016; 6:28481. [PMID: 27329558 PMCID: PMC4916443 DOI: 10.1038/srep28481] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/25/2022] Open
Abstract
Human bocavirus (HBoV), a parvovirus, is a single-stranded DNA etiologic agent causing lower respiratory tract infections in young children worldwide. Nuclear factor kappa B (NF-κB) transcription factors play crucial roles in clearance of invading viruses through activation of many physiological processes. Previous investigation showed that HBoV infection could significantly upregulate the level of TNF-α which is a strong NF-κB stimulator. Here we investigated whether HBoV proteins modulate TNF-α–mediated activation of the NF-κB signaling pathway. We showed that HBoV NS1 and NS1-70 proteins blocked NF-κB activation in response to TNF-α. Overexpression of TNF receptor-associated factor 2 (TRAF2)-, IκB kinase alpha (IKKα)-, IκB kinase beta (IKKβ)-, constitutively active mutant of IKKβ (IKKβ SS/EE)-, or p65-induced NF-κB activation was inhibited by NS1 and NS1-70. Furthermore, NS1 and NS1-70 didn’t interfere with TNF-α-mediated IκBα phosphorylation and degradation, nor p65 nuclear translocation. Coimmunoprecipitation assays confirmed the interaction of both NS1 and NS1-70 with p65. Of note, NS1 but not NS1-70 inhibited TNF-α-mediated p65 phosphorylation at ser536. Our findings together indicate that HBoV NS1 and NS1-70 inhibit NF-κB activation. This is the first time that HBoV has been shown to inhibit NF-κB activation, revealing a potential immune-evasion mechanism that is likely important for HBoV pathogenesis.
Collapse
Affiliation(s)
- Qingshi Liu
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenfeng Zhang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zhenhua Zheng
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Caishang Zheng
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yan Liu
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Qinxue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xianliang Ke
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Hanzhong Wang
- Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
10
|
Hodgson A, Wan F. Interference with nuclear factor kappaB signaling pathway by pathogen-encoded proteases: global and selective inhibition. Mol Microbiol 2016; 99:439-52. [PMID: 26449378 PMCID: PMC5003429 DOI: 10.1111/mmi.13245] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2015] [Indexed: 01/26/2023]
Abstract
Pathogens have evolved a myriad of ways to abrogate and manipulate the host response to infections. Of the various mechanisms involved, pathogen-encoded and sometimes host-encoded proteases are an important category of virulence factors that cause robust changes on the host response by targeting key proteins along signaling cascades. The nuclear factor kappaB (NF-κB) signaling pathway is a crucial regulatory mechanism for the cell, controlling the expression of survival, immune and proliferation genes. Proteases from pathogens of almost all types have been demonstrated to target and cleave members of the NF-κB signaling pathway at nearly every level. This review provides discussion of proteases targeting the most abundant NF-κB subunit, p65, and the impact of protease-mediated p65 cleavage on the immune responses and survival of the infected host cell. After examining various examples of protease interference, it becomes evident that the cleavage fragments produced by pathogen-driven proteolytic processing should be further characterized to determine whether they have novel and unique functions within the cell. The selective targeting of p65 and its effect on gene transcription reveals unique mechanisms by which pathogens acutely alter their microenvironment, and further research may open new opportunities for novel therapeutics to combat pathogens.
Collapse
Affiliation(s)
- Andrea Hodgson
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21025, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21025, USA
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21025, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
11
|
Wier EM, Fu K, Hodgson A, Sun X, Wan F. Caspase-3 cleaved p65 fragment dampens NF-κB-mediated anti-apoptotic transcription by interfering with the p65/RPS3 interaction. FEBS Lett 2015; 589:3581-7. [PMID: 26526615 PMCID: PMC4655178 DOI: 10.1016/j.febslet.2015.10.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 10/11/2015] [Accepted: 10/16/2015] [Indexed: 12/17/2022]
Abstract
Caspase-3-mediated p65 cleavage is believed to suppress nuclear factor-kappa B (NF-κB)-mediated anti-apoptotic transactivation in cells undergoing apoptosis. However, only a small percentage of p65 is cleaved during apoptosis, not in proportion to the dramatic reduction in NF-κB transactivation. Here we show that the p65(1-97) fragment generated by Caspase-3 cleavage interferes with ribosomal protein S3 (RPS3), an NF-κB "specifier" subunit, and selectively retards the nuclear translocation of RPS3, thus dampening the RPS3/NF-κB-dependent anti-apoptotic gene expression. Our findings reveal a novel cell fate determination mechanism to ensure cells undergo programed cell death through interfering with RPS3/NF-κB-conferred anti-apoptotic transcription by the fragment from partial p65 cleavage by activated Caspase-3.
Collapse
Affiliation(s)
- Eric M Wier
- Department of Biochemistry and Molecular Biology, USA
| | - Kai Fu
- Department of Biochemistry and Molecular Biology, USA
| | - Andrea Hodgson
- Department of Biochemistry and Molecular Biology, USA; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21025, USA
| | - Xin Sun
- Department of Biochemistry and Molecular Biology, USA
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.
| |
Collapse
|
12
|
Hodgson A, Wier EM, Fu K, Sun X, Yu H, Zheng W, Sham HP, Johnson K, Bailey S, Vallance BA, Wan F. Metalloprotease NleC suppresses host NF-κB/inflammatory responses by cleaving p65 and interfering with the p65/RPS3 interaction. PLoS Pathog 2015; 11:e1004705. [PMID: 25756944 PMCID: PMC4355070 DOI: 10.1371/journal.ppat.1004705] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/26/2015] [Indexed: 11/18/2022] Open
Abstract
Attaching/Effacing (A/E) pathogens including enteropathogenic Escherichia coli (EPEC), enterohemorrhagic E. coli (EHEC) and the rodent equivalent Citrobacter rodentium are important causative agents of foodborne diseases. Upon infection, a myriad of virulence proteins (effectors) encoded by A/E pathogens are injected through their conserved type III secretion systems (T3SS) into host cells where they interfere with cell signaling cascades, in particular the nuclear factor kappaB (NF-κB) signaling pathway that orchestrates both innate and adaptive immune responses for host defense. Among the T3SS-secreted non-LEE-encoded (Nle) effectors, NleC, a metalloprotease, has been recently elucidated to modulate host NF-κB signaling by cleaving NF-κB Rel subunits. However, it remains elusive how NleC recognizes NF-κB Rel subunits and how the NleC-mediated cleavage impacts on host immune responses in infected cells and animals. In this study, we show that NleC specifically targets p65/RelA through an interaction with a unique N-terminal sequence in p65. NleC cleaves p65 in intestinal epithelial cells, albeit a small percentage of the molecule, to generate the p65¹⁻³⁸ fragment during C. rodentium infection in cultured cells. Moreover, the NleC-mediated p65 cleavage substantially affects the expression of a subset of NF-κB target genes encoding proinflammatory cytokines/chemokines, immune cell infiltration in the colon, and tissue injury in C. rodentium-infected mice. Mechanistically, the NleC cleavage-generated p65¹⁻³⁸ fragment interferes with the interaction between p65 and ribosomal protein S3 (RPS3), a 'specifier' subunit of NF-κB that confers a subset of proinflammatory gene transcription, which amplifies the effect of cleaving only a small percentage of p65 to modulate NF-κB-mediated gene expression. Thus, our results reveal a novel mechanism for A/E pathogens to specifically block NF-κB signaling and inflammatory responses by cleaving a small percentage of p65 and targeting the p65/RPS3 interaction in host cells, thus providing novel insights into the pathogenic mechanisms of foodborne diseases.
Collapse
Affiliation(s)
- Andrea Hodgson
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Eric M. Wier
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Kai Fu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Xin Sun
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Hongbing Yu
- Division of Gastroenterology, Department of Pediatrics, BC’s Children’s Hospital and Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Wenxin Zheng
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Ho Pan Sham
- Division of Gastroenterology, Department of Pediatrics, BC’s Children’s Hospital and Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Kaitlin Johnson
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Scott Bailey
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Bruce A. Vallance
- Division of Gastroenterology, Department of Pediatrics, BC’s Children’s Hospital and Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
13
|
Activation of apoptosis by caspase-3-dependent specific RelB cleavage in anticancer agent-treated cancer cells: involvement of positive feedback mechanism. Biochem Biophys Res Commun 2014; 456:810-4. [PMID: 25511695 DOI: 10.1016/j.bbrc.2014.12.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/05/2014] [Indexed: 11/22/2022]
Abstract
DTCM-glutarimide (DTCM-G) is a newly found anti-inflammatory agent. In the course of experiments with lymphoma cells, we found that DTCM-G induced specific RelB cleavage. Anticancer agent vinblastine also induced the specific RelB cleavage in human fibrosarcoma HT1080 cells. The site-directed mutagenesis analysis revealed that the Asp205 site in RelB was specifically cleaved possibly by caspase-3 in vinblastine-treated HT1080 cells. Moreover, the cells stably overexpressing RelB Asp205Ala were resistant to vinblastine-induced apoptosis. Thus, the specific Asp205 cleavage of RelB by caspase-3 would be involved in the apoptosis induction by anticancer agents, which would provide the positive feedback mechanism.
Collapse
|
14
|
RIP3 is downregulated in human myeloid leukemia cells and modulates apoptosis and caspase-mediated p65/RelA cleavage. Cell Death Dis 2014; 5:e1384. [PMID: 25144719 PMCID: PMC4454320 DOI: 10.1038/cddis.2014.347] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/08/2014] [Accepted: 07/10/2014] [Indexed: 11/08/2022]
Abstract
The receptor-interacting protein kinase 3 (RIP3) associates with RIP1 in a
necrosome complex that can induce necroptosis, apoptosis, or cell proliferation.
We analyzed the expression of RIP1 and RIP3 in CD34+ leukemia cells from a
cohort of patients with acute myeloid leukemia (AML) and CD34+ cells from
healthy donors. RIP3 expression was significantly reduced in most AML samples,
whereas the expression of RIP1 did not differ significantly. When re-expressed
in the mouse DA1-3b leukemia cell line, RIP3 induced apoptosis and necroptosis
in the presence of caspase inhibitors. Transfection of RIP3 in the WEHI-3b
leukemia cell line or in the mouse embryonic fibroblasts also resulted in
increased cell death. Surprisingly, re-expression of a RIP3 mutant with an
inactive kinase domain (RIP3-kinase dead (RIP3-KD)) induced significantly more
and earlier apoptosis than wild-type RIP3 (RIP3-WT), indicating that the RIP3
kinase domain is an essential regulator of apoptosis/necroptosis in leukemia
cells. The induced in vivo expression of RIP3-KD but not RIP3-WT
prolonged the survival of mice injected with leukemia cells. The expression of
RIP3-KD induced p65/RelA nuclear factor-κB
(NF-κB) subunit caspase-dependent cleavage, and a
non-cleavable p65/RelA D361E mutant rescued these cells from apoptosis.
p65/RelA cleavage appears to be at least partially mediated by caspase-6.
These data indicate that RIP3 silencing in leukemia cells results in suppression
of the complex regulation of the apoptosis/necroptosis switch and
NF-κB activity.
Collapse
|
15
|
Zhou LY, Chen FY, Shen LJ, Wan HX, Zhong JH. Arsenic trioxide induces apoptosis in the THP1 cell line by downregulating EVI-1. Exp Ther Med 2014; 8:85-90. [PMID: 24944602 PMCID: PMC4061239 DOI: 10.3892/etm.2014.1716] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 04/29/2014] [Indexed: 01/12/2023] Open
Abstract
Acute leukemia is a malignant clonal hematopoietic stem cell disease. In the current study, the effects of arsenic trioxide (ATO) on the ecotropic viral integration site-1 (EVI-1) gene were investigated in the THP1 cell line. THP-1 cells were treated with different concentrations of ATO (0, 1, 3 and 5 μM) for 24, 48 or 72 h, then tested for cell viability by CCK-8 kit, cell morphology by cytospin smear, cell apoptosis by flow cytometry, EVI-1 mRNA expression by reverse transcription polymerase chain reaction (RT-PCR) and protein quantity by western blot. ATO treatment was shown to inhibit proliferation and induce apoptosis in THP1 cells in a dose- and time-dependent manner. ATO downregulated the mRNA and protein expression of EVI-1 in the THP1 cell line. In addition, ATO significantly decreased the expression of antiapoptotic proteins, B-cell lymphoma 2 (Bcl-2) and B cell lymphoma-extra large (Bcl-xL), but markedly increased the expression of proapoptotic proteins, including c-Jun N-terminal kinase (JNK), phosphorylated-JNK, Bax, full length caspase-3 and cleaved caspase-3. These results indicated that ATO inhibited the proliferation and induced apoptosis in THP1 cells partially via blocking the inhibitory effects of EVI-1 on the JNK signaling pathway with the involvement of apoptosis-associated proteins, including Bax, Bcl-2, Bcl-xL and caspase-3. These novel observations may be used to elucidate the mechanism by which ATO induces apoptosis in acute leukemia cells, and provide rationales to develop a personalized medicine strategy for ATO via targeting EVI-1 positive neoplasm.
Collapse
Affiliation(s)
- Ling-Yun Zhou
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Fang-Yuan Chen
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Li-Jing Shen
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Hai-Xia Wan
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Ji-Hua Zhong
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
16
|
Caperna TJ, Shannon AE, Garrett WM, Ramsay TG, Blomberg LA, Elsasser TH. Identification and characterization of a nuclear factor-κ B-p65 proteolytic fragment in nuclei of porcine hepatocytes in monolayer culture. Domest Anim Endocrinol 2013; 45:154-62. [PMID: 24011531 DOI: 10.1016/j.domaniend.2013.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 08/12/2013] [Accepted: 08/12/2013] [Indexed: 01/12/2023]
Abstract
Hepatic responses to proinflammatory signals are controlled by the activation of several transcription factors, including, nuclear factor-κ B (NF-κB). In this study, hepatocytes prepared from suckling pigs and maintained in serum-free monolayer culture were used to define a novel proinflammatory cytokine-specific NF-κB subunit modification. The immunoreactive p65 protein was detected by Western blot analysis at the appropriate molecular weight in the cytosol of control cultures and those incubated with tumor necrosis factor-α (TNF). However, in nuclei, the p65 antisera cross-reacted with a protein of approximately 38 kDa (termed p38) after TNF addition, which was not observed in the cytosol of control or cytokine-treated cells. Specifically, incubation with TNF also resulted in phosphorylation (P < 0.05) of the inhibitor complex protein (IκB), whereas incubation with other cytokines, IL-6, IL-17a, or oncostatin M was not associated with either phosphorylation of IκB or nuclear translocation of p65. Intracellular endothelial nitric oxide synthase was deceased (P < 0.05) and plasminogen activator inhibitor-1 secretion was increased (P < 0.05) after TNF incubation. The TNF-induced p38 protein was purified from hepatocyte nuclei by immunoprecipitation, concentrated by electrophoresis, and subsequently analyzed by mass spectrometry. Ten unique NF-κB p65 peptides were identified after digestion with trypsin and chymotrypsin; however, all were mapped to the N-terminus and within the first 310 amino acid residues of the intact p65 protein. Although low molecular weight immunoreactive p65 molecules were previously observed in various human and rodent systems, this is the first report to positively identify the p38 fragment within hepatocyte nuclei or after specific cytokine (TNF) induction.
Collapse
Affiliation(s)
- T J Caperna
- Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, USDA, Agricultural Research Service, Bldg 200, Rm 202, BARC-East, Beltsville, MD 20705, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Wang Y, Zhang J, Wang Q, Zhang T, Yang Y, Yi Y, Gao G, Dong H, Zhu H, Li Y, Lin H, Tang H, Chen X. Bryostatin 5 induces apoptosis in acute monocytic leukemia cells by activating PUMA and caspases. Eur J Pharmacol 2013; 718:340-9. [PMID: 24036350 DOI: 10.1016/j.ejphar.2013.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/19/2013] [Accepted: 08/24/2013] [Indexed: 12/14/2022]
Abstract
Acute leukemia is a malignant clonal hematopoietic stem cell disease. In the current study, we examined the effects of bryostatin 5 on acute monocytic leukemia cells in vitro and in vivo. We also explored the mechanisms and pathways underlying the increase in apoptosis induced by bryostatin 5. Bryostatin 5 inhibited the growth of primary acute monocytic leukemia cells and U937 cells in a dose- and time-dependent manners. Bryostatin 5 also induced an increase in apoptosis and a decrease in the mitochondrial membrane potential (MMP) in U937 cells. Transmission electron microscopy (TEM) revealed that bryostatin 5-treated cells displayed typical apoptotic characteristics (chromatin condensation, karyopyknosis and formation of crescents and apoptotic bodies). In addition, bryostatin 5 increased the expression of P53 upregulated modulator of apoptosis (PUMA) and slightly increased P53 expression. Bryostatin 5 also significantly decreased Bcl-XL expression and significantly increased the expression levels of Bak, Bax, cleaved caspase 9 and cleaved caspase 3. The pro-apoptotic activity of bryostatin 5 in U937 cells was inhibited by PUMA siRNA and z-LEHD-fmk (a specific caspase 9 inhibitor). In addition, the PUMA siRNA significantly affected the expression of cleaved caspase 9, whereas z-LEHD-fmk had little effect on the expression of PUMA. The results suggest that PUMA is located upstream of caspase 9 in this apoptotic signaling pathway. These novel findings provide mechanistic insight into the induction of apoptosis by bryostatin 5 and might facilitate the development of clinical strategies to enhance the therapeutic efficacy of treatments for acute monocytic leukemia.
Collapse
Affiliation(s)
- Yiwei Wang
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Silva DS, Pereira LMG, Moreira AR, Ferreira-da-Silva F, Brito RM, Faria TQ, Zornetta I, Montecucco C, Oliveira P, Azevedo JE, Pereira PJB, Macedo-Ribeiro S, do Vale A, dos Santos NMS. The apoptogenic toxin AIP56 is a metalloprotease A-B toxin that cleaves NF-κb P65. PLoS Pathog 2013; 9:e1003128. [PMID: 23468618 PMCID: PMC3585134 DOI: 10.1371/journal.ppat.1003128] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 11/28/2012] [Indexed: 12/15/2022] Open
Abstract
AIP56 (apoptosis-inducing protein of 56 kDa) is a major virulence factor of Photobacterium damselae piscicida (Phdp), a Gram-negative pathogen that causes septicemic infections, which are among the most threatening diseases in mariculture. The toxin triggers apoptosis of host macrophages and neutrophils through a process that, in vivo, culminates with secondary necrosis of the apoptotic cells contributing to the necrotic lesions observed in the diseased animals. Here, we show that AIP56 is a NF-κB p65-cleaving zinc-metalloprotease whose catalytic activity is required for the apoptogenic effect. Most of the bacterial effectors known to target NF-κB are type III secreted effectors. In contrast, we demonstrate that AIP56 is an A-B toxin capable of acting at distance, without requiring contact of the bacteria with the target cell. We also show that the N-terminal domain cleaves NF-κB at the Cys39-Glu40 peptide bond and that the C-terminal domain is involved in binding and internalization into the cytosol. The apoptosis inducing protein of 56 kDa (AIP56) is a key virulence factor secreted by Photobacterium damselae piscicida (Phdp), a Gram-negative bacterium that causes septicaemic infections in economically important marine fish species. It is known that AIP56 induces massive destruction of the phagocytic cells of the infected host, allowing the extracellular multiplication of the bacteria and contributing to the genesis of the pathology. Here we show that AIP56 acts by cleaving NF-κB p65. The NF-κB family of transcription factors is evolutionarily conserved and plays a central role in the host responses to microbial pathogen invasion, regulating the expression of inflammatory and anti-apoptotic genes. Pathogenic bacteria have evolved complex strategies to interfere with NF-κB signalling, usually by injecting protein effectors directly into the cell's cytosol through bacterial secretion machineries that require contact with host cells. In contrast, AIP56 acts at distance and has an intrinsic ability to reach the cytosol due to the presence of a C-terminal domain that functions as “delivery module.”
Collapse
Affiliation(s)
- Daniela S. Silva
- Fish Immunology and Vaccinology, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Liliana M. G. Pereira
- Fish Immunology and Vaccinology, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Ana R. Moreira
- Fish Immunology and Vaccinology, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Frederico Ferreira-da-Silva
- Protein Production and Purification, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Rui M. Brito
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Chemistry Department, Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | - Tiago Q. Faria
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Irene Zornetta
- Dipartimento di Scienze Biomediche dell'Università di Padova and Instituto di Neuroscienze del CNR, Padova, Italy
| | - Cesare Montecucco
- Dipartimento di Scienze Biomediche dell'Università di Padova and Instituto di Neuroscienze del CNR, Padova, Italy
| | - Pedro Oliveira
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Jorge E. Azevedo
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
- Organelle Biogenesis and Function, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Pedro J. B. Pereira
- Biomolecular Structure, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Sandra Macedo-Ribeiro
- Protein Crystallography, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Ana do Vale
- Fish Immunology and Vaccinology, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Nuno M. S. dos Santos
- Fish Immunology and Vaccinology, Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
19
|
Wier EM, Neighoff J, Sun X, Fu K, Wan F. Identification of an N-terminal truncation of the NF-κB p65 subunit that specifically modulates ribosomal protein S3-dependent NF-κB gene expression. J Biol Chem 2012; 287:43019-29. [PMID: 23115242 DOI: 10.1074/jbc.m112.388694] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
NF-κB is a pleiotrophic transcription factor that plays a prominent regulatory role in various cellular processes. Although previous efforts have focused on its activation, how NF-κB selects specific target genes in response to discrete signals remains puzzling. In addition to the well defined Rel protein components of NF-κB, the ribosomal protein S3 (RPS3) was identified to be an essential component of specific NF-κB complexes. RPS3 synergistically interacts with the NF-κB p65 subunit to achieve optimal binding and transactivation of a subset of NF-κB target genes, thus providing regulatory specificity. Emerging evidence suggests an important role for the RPS3-p65 interaction in context-specific NF-κB gene transcription. The food-borne pathogen Escherichia coli O157:H7 impacts the transcription of a subset of NF-κB target genes encoding proinflammatory cytokines and chemokines in host cells by preventing the nuclear translocation of RPS3, but not p65. The N terminus of p65 is crucial for RPS3 binding. Although several p65 N-terminal fragments are generated by either protease cleavage or alternative mRNA splicing under certain pathophysiological conditions, the role of these fragments in modulating NF-κB signaling, in particular RPS3-dependent selective gene transcription, has not been fully characterized. Here we report that an N-terminal fragment of p65 (amino acids 21-186) can selectively modulate NF-κB gene transcription by competing for RPS3 binding to p65. This 21-186 fragment preferentially localizes in the cytoplasm where it delays stimuli-induced RPS3 nuclear translocation, without affecting the nuclear translocation of p65. Our findings thus uncover a new cytoplasmic function for the N-terminal domain of p65 and provide a novel strategy for selective inhibition of NF-κB gene transcription.
Collapse
Affiliation(s)
- Eric M Wier
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
20
|
Chacón PJ, Rodríguez-Tébar A. Increased expression of the homologue of enhancer-of-split 1 protects neurons from beta amyloid neurotoxicity and hints at an alternative role for transforming growth factor beta1 as a neuroprotector. ALZHEIMERS RESEARCH & THERAPY 2012; 4:31. [PMID: 22849569 PMCID: PMC3506945 DOI: 10.1186/alzrt134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 06/11/2012] [Accepted: 07/31/2012] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the deposition of β-amyloid (Aβ) in the brain, which produces progressive neuronal loss and dementia. We recently demonstrated that the noxious effects of Aβ on cultured hippocampal neurons are in part provoked by the antagonism of nerve growth factor (NGF) signalling, which impairs the activation of nuclear factor κB (NF-κB) by impeding the tyrosine phosphorylation of I-κBα. As a result, the expression of the homologue of Enhancer-of split 1 (Hes1) gene is downregulated and ultimately, gamma-aminobutyric acid (GABA)-ergic connectivity is lost. METHODS Hes1 activity was promoted in cultured hippocampal neurons by overexpressing a Hes1-encoding plasmid or by upregulating this gene by activating NF-κB through different approaches (overexpressing either the I-κB kinaseβ, or p65/RelA/NF-κB). Alternatively neurons were exposed to TGFβ1. Dendrite patterning, GABAergic connectivity and cell survival were analyzed by immunofluorescence microscopy. Hes1 expression was determined by real-time PCR. NF-κB activation was measured using the dual-luciferase reporter assay. RESULTS The expression of Hes1 abolished the effects of Aβ on dendritic patterning and GABAergic input, and it prevented the death of the cultured neurons. TGFβ1, a known neuroprotector, could counteract the deleterious effects of Aβ by inducing NF-κB activation following the serine phosphorylation of I-κBα. Indeed, the number of GABAergic terminals generated by inducing Hes1 expression was doubled. CONCLUSION Our data define some of the mechanisms involved in Aβ-mediated cell death and they point to potential means to counteract this noxious activity.
Collapse
Affiliation(s)
- Pedro J Chacón
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Americo Vespucio s/n, Isla de la Cartuja, 41092 Seville, Spain.
| | | |
Collapse
|
21
|
Sánchez-Jiménez C, Olivares I, de Ávila Lucas AI, Toledano V, Gutiérrez-Rivas M, Lorenzo-Redondo R, Grande-Pérez A, Domingo E, López-Galíndez C. Mutagen-mediated enhancement of HIV-1 replication in persistently infected cells. Virology 2012; 424:147-53. [PMID: 22265575 DOI: 10.1016/j.virol.2011.12.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 11/28/2011] [Accepted: 12/22/2011] [Indexed: 12/23/2022]
Abstract
Lethal mutagenesis, a new antiviral strategy to extinguish virus through elevated mutation rates, was explored in H61-D cells an HIV-1 persistently infected lymphoid cell line. Three mutagenic agents: 5-hydroxy-2(')-deoxycytidine (5-OHdC), 5-fluorouracil (5-FU) and 2,2(')-difluoro-2(')-deoxycytidine (gemcitabine) were used. After 54 passages, treatments with 5-FU and gemcitabine reduced virus infectivity, p24 and RT activity. Treatment with the pyrimidine analog 5-OHdC resulted in increases of p24 production, RT activity and infectivity. Rise in viral replication by 5-OHdC during HIV-1 persistence is in contrast with its inhibitory effect in acute infections. Viral replication enhancement by 5-OHdC was associated with an increase in intracellular HIV-1 RNA mutations. Mechanisms of HIV-1 replication enhancement by 5-OHdC are unknown but some potential factors are discussed. Increase of HIV-1 replication by 5-OHdC cautions against the use, without previous analyses, of mutagenic nucleoside analogs for AIDS treatment.
Collapse
Affiliation(s)
- Carmen Sánchez-Jiménez
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Reuter S, Gupta SC, Kannappan R, Aggarwal BB. WITHDRAWN: Evidence for the critical roles of NF-κB p65 and specificity proteins in the apoptosis-inducing activity of proteasome inhibitors in leukemia cells. BIOCHIMICA ET BIOPHYSICA ACTA 2012:S0925-4439(12)00013-0. [PMID: 22265847 PMCID: PMC3870997 DOI: 10.1016/j.bbadis.2012.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Revised: 12/30/2011] [Accepted: 01/04/2012] [Indexed: 10/14/2022]
Abstract
This article has been withdrawn at the request of editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
|
23
|
NleC, a type III secretion protease, compromises NF-κB activation by targeting p65/RelA. PLoS Pathog 2010; 6:e1001231. [PMID: 21187904 PMCID: PMC3002990 DOI: 10.1371/journal.ppat.1001231] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 11/15/2010] [Indexed: 02/06/2023] Open
Abstract
The NF-κB signaling pathway is central to the innate and adaptive immune responses. Upon their detection of pathogen-associated molecular patterns, Toll-like receptors on the cell surface initiate signal transduction and activate the NF-κB pathway, leading to the production of a wide array of inflammatory cytokines, in attempt to eradicate the invaders. As a countermeasure, pathogens have evolved ways to subvert and manipulate this system to their advantage. Enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC) are closely related bacteria responsible for major food-borne diseases worldwide. Via a needle-like protein complex called the type three secretion system (T3SS), these pathogens deliver virulence factors directly to host cells and modify cellular functions, including by suppressing the inflammatory response. Using gain- and loss-of-function screenings, we identified two bacterial effectors, NleC and NleE, that down-regulate the NF-κB signal upon being injected into a host cell via the T3SS. A recent report showed that NleE inhibits NF-κB activation, although an NleE-deficient pathogen was still immune-suppressive, indicating that other anti-inflammatory effectors are involved. In agreement, our present results showed that NleC was also required to inhibit inflammation. We found that NleC is a zinc protease that disrupts NF-κB activation by the direct cleavage of NF-κB's p65 subunit in the cytoplasm, thereby decreasing the available p65 and reducing the total nuclear entry of active p65. More importantly, we showed that a mutant EPEC/EHEC lacking both NleC and NleE (ΔnleC ΔnleE) caused greater inflammatory response than bacteria carrying ΔnleC or ΔnleE alone. This effect was similar to that of a T3SS-defective mutant. In conclusion, we found that NleC is an anti-inflammatory bacterial zinc protease, and that the cooperative function of NleE and NleC disrupts the NF-κB pathway and accounts for most of the immune suppression caused by EHEC/EPEC. Enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) cause food-borne diseases, including watery diarrhea or severe bloody diarrhea and life-threatening kidney disease (hemolytic uremic syndrome). Upon ingestion, EPEC/EHEC colonize the cells of the epithelial lining in the intestinal tract. In response, the affected cells initiate an immune response by secreting cytokines that attract immune cells. To prevent their early elimination by the host, these bacteria have developed strategies to prevent the host immune response. They do this by injecting bacterial effectors into the host cells to disrupt the NF-κB pathway, an essential effector of the host cell immune response. In the current study, we report the discovery of an NF-κB suppressive effector in EPEC/EHEC called NleC, and its novel mechanism. We found that NleC is a zinc protease that can digest p65, a critical component of the NF-κB pathway, thus dampening the host inflammatory response. NleE is another recently identified anti-inflammatory effector. We show here that an EPEC/EHEC mutant deficient in both NleC and NleE loses most of its ability to suppress the host inflammatory response. Our findings show how two different bacterial effectors can function in cooperation to modify the host immune response.
Collapse
|
24
|
Bedoya LM, Abad MJ, Calonge E, Saavedra LA, Gutierrez C M, Kouznetsov VV, Alcami J, Bermejo P. Quinoline-based compounds as modulators of HIV transcription through NF-kappaB and Sp1 inhibition. Antiviral Res 2010; 87:338-44. [PMID: 20600334 DOI: 10.1016/j.antiviral.2010.06.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 05/06/2010] [Accepted: 06/14/2010] [Indexed: 01/27/2023]
Abstract
18 quinoline-based compounds were tested for antiviral properties against human immunodeficiency syndrome (HIV). The compounds tested here contain quinoline or tetrahydroquinoline rings and can be divided into two main groups: group 1 includes 4-(2-oxopyrrolidinyl-1)-1,2,3,4-tetrahydroquinolines with 2-(3-nitrophenyl) substituent (N-series) or 2-(3-aminophenyl) moiety (H-series), and group 2 includes 2-(3-nitrophenyl)- or 2-(3-aminophenyl)-substituted quinolines (S-series). Two different antiviral assays were performed in order to test the anti-HIV activity of compounds: 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and recombinant virus assay (RVA). Results showed that the most active compounds were 2-aryl quinolines, particularly those containing methoxy substituents or no substituents in the quinoline skeleton. HIV transcription inhibition appears to be their target in both resting and phorbol myristate acetate (PMA) activated primary lymphocytes, and nuclear factor-kappaB (NF-kappaB) and specificity protein-1 (SP1) seems to be the most important transcription factors involved in their action.
Collapse
Affiliation(s)
- Luis Miguel Bedoya
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Crt. Majadahonda a Pozuelo, 28220 Majadahonda, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|