1
|
Luo Y, Yang Y, Wang W, Gao Q, Gong T, Feng Y, Wu D, Zheng X, Zhang G, Wang H. Aloe-emodin inhibits African swine fever virus replication by promoting apoptosis via regulating NF-κB signaling pathway. Virol J 2023; 20:158. [PMID: 37468960 PMCID: PMC10357809 DOI: 10.1186/s12985-023-02126-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
African swine fever (ASF) is an acute infectious haemorrhagic fever of pigs caused by African swine fever virus (ASFV). Aloe-emodin (Ae) is an active ingredient of Chinese herbs with antiviral, anticancer, and anti-inflammatory effects. We investigated the antiviral activity and mechanism of action of Ae against ASFV using Real-time quantitative PCR (qPCR), western blotting, and indirect immunofluorescence assays. Ae significantly inhibited ASFV replication. Furthermore, transcriptomic analysis revealed that ASFV infection activated the NF-κB signaling pathway in the early stage and the apoptosis pathway in the late stage. Ae significantly downregulated the expression levels of MyD88, phosphor-NF-κB p65, and pIκB proteins as well as the mRNA levels of IL-1β and IL-8 in porcine alveolar macrophages (PAMs) infected with ASFV, thereby inhibiting the activation of the NF-κB signaling pathway induced by ASFV. Flow cytometry and western blot analysis revealed that Ae significantly increased the percentage of ASFV-induced apoptotic cells. Additionally, Ae promoted apoptosis by upregulating the expression levels of cleaved-caspase3 and Bax proteins and downregulating the expression levels of Bcl-2 proteins. This suggests that Ae promotes apoptosis by inhibiting the NF-κB pathway, resulting in inhibition of ASFV replication. These findings have further improved therapeutic reserves for the prevention and treatment of ASF.
Collapse
Affiliation(s)
- Yizhuo Luo
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, 510642, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, PR China
| | - Yunlong Yang
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, 510642, China
| | - Wenru Wang
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China
| | - Qi Gao
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Ting Gong
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China
- Research Center for African Swine Fever Prevention and Control, South China Agricultural University, Guangzhou, 510642, China
| | - Yongzhi Feng
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Dongdong Wu
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China
- Research Center for African Swine Fever Prevention and Control, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoyu Zheng
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Guihong Zhang
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, 510642, China
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, PR China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
- Research Center for African Swine Fever Prevention and Control, South China Agricultural University, Guangzhou, 510642, China
| | - Heng Wang
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510462, China.
- African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou, 510642, China.
- Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou, PR China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China.
- Research Center for African Swine Fever Prevention and Control, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
2
|
Yu X, Liu Y, Wang Y, Mao X, Zhang Y, Xia J. Baicalein induces cervical cancer apoptosis through the NF-κB signaling pathway. Mol Med Rep 2018; 17:5088-5094. [PMID: 29393414 PMCID: PMC5865972 DOI: 10.3892/mmr.2018.8493] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 07/25/2017] [Indexed: 01/03/2023] Open
Abstract
To investigate the mechanism of baicalein in inducing human cervical cancer cell line C33A apoptosis. Baicalein (200 µM) was used to treat C33A cells. Cell proliferation was tested by the MTT assay. Cell apoptosis was detected by the TUNEL assay and caspase‑3 activity measurement. Cell cycle was determined by flow cytometry and associated gene expression at mRNA and protein levels. Nuclear factor (NF)‑κB activity was assessed by luciferase assay and western blotting. Baicalein suppressed cervical cancer cell C33A proliferation and induced cell apoptosis by activating caspase‑3 activity. Baicalein blocked cell cycle in G0/G1 phase through regulating the expression of associated genes. Baicalein inhibited NF‑κB activity by repressing nuclear translocation. Baicalein suppressed C33A proliferation and promoted cellular apoptosis by inhibiting NF‑κB signaling pathway. In conclusion, the results indicate that baicalein can inhibit cervical cancer cell proliferation and promote cell apoptosis by affecting NF-κB activity.
Collapse
Affiliation(s)
- Xiaolan Yu
- Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yuqing Liu
- School of Medical Information and Engineering, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yongzhou Wang
- Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xiguan Mao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yujiao Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jiyi Xia
- School of Medical Information and Engineering, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
3
|
Eosinophil-specific deletion of IκBα in mice reveals a critical role of NF-κB-induced Bcl-xL for inhibition of apoptosis. Blood 2015; 125:3896-904. [PMID: 25862560 DOI: 10.1182/blood-2014-10-607788] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 04/04/2015] [Indexed: 12/17/2022] Open
Abstract
Eosinophils are associated with type 2 immune responses to allergens and helminths. They release various proinflammatory mediators and toxic proteins on activation and are therefore considered proinflammatory effector cells. Eosinophilia is promoted by the cytokines interleukin (IL)-3, IL-5, and granulocyte macrophage-colony-stimulating factor (GM-CSF) and can result from enhanced de novo production or reduced apoptosis. In this study, we show that only IL-5 induces differentiation of eosinophils from bone marrow precursors, whereas IL-5, GM-CSF, and to a lesser extent IL-3 promote survival of mature eosinophils. The receptors for these cytokines use the common β chain, which serves as the main signaling unit linked to signal transducer and activator of transcription 5, p38 mitogen-activated protein kinase, and nuclear factor (NF)-κB pathways. Inhibition of NF-κB induced apoptosis of in vitro cultured eosinophils. Selective deletion of IκBα in vivo resulted in enhanced expression of Bcl-xL and reduced apoptosis during helminth infection. Retroviral overexpression of Bcl-xL promoted survival, whereas pharmacologic inhibition of Bcl-xL in murine or human eosinophils induced rapid apoptosis. These results suggest that therapeutic strategies targeting Bcl-xL in eosinophils could improve health conditions in allergic inflammatory diseases.
Collapse
|
4
|
NF-κB regulates caspase-4 expression and sensitizes neuroblastoma cells to Fas-induced apoptosis. PLoS One 2015; 10:e0117953. [PMID: 25695505 PMCID: PMC4335045 DOI: 10.1371/journal.pone.0117953] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/06/2015] [Indexed: 12/01/2022] Open
Abstract
Found in neurons and neuroblastoma cells, Fas-induced apoptosis and accompanied activation of NF-κB signaling were thought to be associated with neurodegenerative diseases. However, the detailed functions of NF-κB activation in Fas killing and the effect of NF-κB activation on its downstream events remain unclear. Here, we demonstrated that agonistic Fas antibody induces cell death in a dose-dependent way and NF-κB signaling is activated as well, in neuroblastoma cells SH-EP1. Unexpectedly, NF-κB activation was shown to be pro-apoptotic, as suggested by the reduction of Fas-induced cell death with either a dominant negative form of IκBα (DN-IκBα) or an IκB kinase-specific inhibitor. To our interest, when analyzing downstream events of NF-κB signaling, we found that DN-IκBα only suppressed the expression of caspase-4, but not other caspases. Vice versa, enhancement of NF-κB activity by p65 (RelA) overexpression increased the expression of caspase-4 at both mRNA and protein levels. More directly, results from dual luciferase reporter assay demonstrated the regulation of caspase-4 promoter activity by NF-κB. When caspase-4 activity was blocked by its dominant negative (DN) form, Fas-induced cell death was substantially reduced. Consistently, the cleavage of PARP and caspase-3 induced by Fas was also reduced. In contrast, the cleavage of caspase-8 remained unaffected in caspase-4 DN cells, although caspase-8 inhibitor could rescue Fas-induced cell death. Collectively, these data suggest that caspase-4 activity is required for Fas-induced cell apoptosis and caspase-4 may act upstream of PARP and caspase-3 and downstream of caspase-8. Overall, we demonstrate that NF-κB can mediate Fas-induced apoptosis through caspase-4 protease, indicating that caspase-4 is a new mediator of NF-κB pro-apoptotic pathway in neuroblastoma cells.
Collapse
|
5
|
Physiological functions of TNF family receptor/ligand interactions in hematopoiesis and transplantation. Blood 2014; 124:176-83. [PMID: 24859365 DOI: 10.1182/blood-2014-03-559641] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Secretion of ligands of the tumor necrosis factor (TNF) superfamily is a conserved response of parenchymal tissues to injury and inflammation that commonly perpetuates elimination of dysfunctional cellular components by apoptosis. The same signals of tissue injury that induce apoptosis in somatic cells activate stem cells and initiate the process of tissue regeneration as a coupling mechanism of injury and recovery. Hematopoietic stem and progenitor cells upregulate the TNF family receptors under stress conditions and are transduced with trophic signals. The progeny gradually acquires sensitivity to receptor-mediated apoptosis along the differentiation process, which becomes the major mechanism of negative regulation of mature proliferating hematopoietic lineages and immune homeostasis. Receptor/ligand interactions of the TNF family are physiological mechanisms transducing the need for repair, which may be harnessed in pathological conditions and transplantation. Because these interactions are physiological mechanisms of injury, neutralization of these pathways has to be carefully considered in disorders that do not involve intrinsic aberrations of excessive susceptibility to apoptosis.
Collapse
|
6
|
Mott R, Yuan W, Kaisaki P, Gan X, Cleak J, Edwards A, Baud A, Flint J. The architecture of parent-of-origin effects in mice. Cell 2014; 156:332-42. [PMID: 24439386 PMCID: PMC3898482 DOI: 10.1016/j.cell.2013.11.043] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 08/02/2013] [Accepted: 11/05/2013] [Indexed: 01/14/2023]
Abstract
The number of imprinted genes in the mammalian genome is predicted to be small, yet we show here, in a survey of 97 traits measured in outbred mice, that most phenotypes display parent-of-origin effects that are partially confounded with family structure. To address this contradiction, using reciprocal F1 crosses, we investigated the effects of knocking out two nonimprinted candidate genes, Man1a2 and H2-ab1, that reside at nonimprinted loci but that show parent-of-origin effects. We show that expression of multiple genes becomes dysregulated in a sex-, tissue-, and parent-of-origin-dependent manner. We provide evidence that nonimprinted genes can generate parent-of-origin effects by interaction with imprinted loci and deduce that the importance of the number of imprinted genes is secondary to their interactions. We propose that this gene network effect may account for some of the missing heritability seen when comparing sibling-based to population-based studies of the phenotypic effects of genetic variants.
Collapse
Affiliation(s)
- Richard Mott
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK.
| | - Wei Yuan
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Pamela Kaisaki
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Xiangchao Gan
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - James Cleak
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Andrew Edwards
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Amelie Baud
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Jonathan Flint
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
7
|
Kou X, Jing Y, Deng W, Sun K, Han Z, Ye F, Yu G, Fan Q, Gao L, Zhao Q, Zhao X, Li R, Wei L, Wu M. Tumor necrosis factor-α attenuates starvation-induced apoptosis through upregulation of ferritin heavy chain in hepatocellular carcinoma cells. BMC Cancer 2013; 13:438. [PMID: 24066693 PMCID: PMC3849379 DOI: 10.1186/1471-2407-13-438] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 09/19/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tumor microenviroment is characteristic of inflammation, ischemia and starvation of nutrient. TNF-α, which is an extraordinarily pleiotropic cytokine, could be an endogenous tumor promoter in some tumor types. The basic objective of this study was to investigate the effects of TNF-α on the cell viability and apoptosis of hepatocellular carcinoma cells under serum starvation, and to identify the molecular mechanisms involved. METHODS For this purpose, five different concentrations of TNF-α and two different serum settings (serum-cultured and serum-deprived) were used to investigate the effects of TNF-α on the cell viability and apoptosis of Hep3B and SMMC-7721 cells. RESULTS TNF-α (10 ng/ml) attenuated serum starvation-induced apoptosis of hepatocellular carcinoma cells, and autophagy conferred this process. BAY11-7082, a specific inhibitor of NF-κB, reversed the suppression of serum starvation-induced apoptosis by TNF-α. Moreover, TNF-α-induced NF-κB transactivation was suppressed by autophagy inhibitor 3-MA. In addition, TNF-α up-regulated Ferritin heavy chain (FHC) transiently by NF-κB activation and FHC levels were correlated with the TNF-α-induced protection against serum starvation-mediated apoptosis of hepatocellular carcinoma cells. Furthermore, FHC-mediated inhibition of apoptosis depended on suppressing ROS accumulation. CONCLUSIONS Our findings suggested that autophagy conferred the TNF-α protection against serum starvation-mediated apoptosis of hepatocellular carcinoma cells, the mechanism involved with the activation of the TNF-α/ NF-κB /FHC signaling pathway.
Collapse
Affiliation(s)
- Xingrui Kou
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, P, R China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Oh J, Kim SH, Ahn S, Lee CE. Suppressors of cytokine signaling promote Fas-induced apoptosis through downregulation of NF-κB and mitochondrial Bfl-1 in leukemic T cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:5561-71. [PMID: 23152563 DOI: 10.4049/jimmunol.1103415] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Suppressors of cytokine signaling (SOCS) are known as negative regulators of cytokine- and growth factor-induced signal transduction. Recently they have emerged as multifunctional proteins with regulatory roles in inflammation, autoimmunity, and cancer. We have recently reported that SOCS1 has antiapoptotic functions against the TNF-α- and the hydrogen peroxide-induced T cell apoptosis through the induction of thioredoxin, which protects protein tyrosine phosphatases and attenuates Jaks. In this study, we report that SOCS, on the contrary, promote death receptor Fas-mediated T cell apoptosis. The proapoptotic effect of SOCS1 was manifested with increases in Fas-induced caspase-8 activation, truncated Bid production, and mitochondrial dysfunctions. Both caspase-8 inhibitor c-Flip and mitochondrial antiapoptotic factor Bfl-1 were significantly reduced by SOCS1. These proapoptotic responses were not associated with changes in Jak or p38/Jnk activities but were accompanied with downregulation of NF-κB and NF-κB-dependent reporter gene expression. Indeed, p65 degradation via ubiquitination was accelerated in SOCS1 overexpressing cells, whereas it was attenuated in SOCS1 knockdown cells. With high NF-κB levels, the SOCS1-ablated cells displayed resistance against Fas-induced apoptosis, which was abrogated upon siBfl-1 transfection. The results indicate that the suppression of NF-κB-dependent induction of prosurvival factors, such as Bfl-1 and c-Flip, may serve as a mechanism for SOCS action to promote Fas-mediated T cell apoptosis. SOCS3 exhibited a similar proapoptotic function. Because both SOCS1 and SOCS3 are induced upon TCR stimulation, SOCS would play a role in activation-induced cell death by sensitizing activated T cells toward Fas-mediated apoptosis to maintain T cell homeostasis.
Collapse
Affiliation(s)
- Jiyoung Oh
- Laboratory of Immunology, Department of Biological Science, Sungkyunkwan University, Suwon 440-746, Korea
| | | | | | | |
Collapse
|
9
|
Gupta P, Dixit D, Sen E. Oncrasin targets the JNK-NF-κB axis to sensitize glioma cells to TNFα-induced apoptosis. Carcinogenesis 2012; 34:388-96. [DOI: 10.1093/carcin/bgs352] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
10
|
Yang D, Torres CM, Bardhan K, Zimmerman M, McGaha TL, Liu K. Decitabine and vorinostat cooperate to sensitize colon carcinoma cells to Fas ligand-induced apoptosis in vitro and tumor suppression in vivo. THE JOURNAL OF IMMUNOLOGY 2012; 188:4441-9. [PMID: 22461695 DOI: 10.4049/jimmunol.1103035] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The death receptor Fas and its physiological ligand (FasL) regulate apoptosis of cancerous cells, thereby functioning as a critical component of the host cancer immunosurveillance system. To evade Fas-mediated apoptosis, cancer cells often downregulate Fas to acquire an apoptosis-resistant phenotype, which is a hallmark of metastatic human colorectal cancer. Therefore, targeting Fas resistance is of critical importance in Fas-based cancer therapy and immunotherapy. In this study, we demonstrated that epigenetic inhibitors decitabine and vorinostat cooperate to upregulate Fas expression in metastatic human colon carcinoma cells. Decitabine also upregulates BNIP3 and Bik expression, whereas vorinostat decreased Bcl-x(L) expression. Altered expression of Fas, BNIP3, Bik, and Bcl-x(L) resulted in effective sensitization of the metastatic human colon carcinoma cells to FasL-induced apoptosis. Using an experimental metastasis mouse model, we further demonstrated that decitabine and vorinostat cooperate to suppress colon carcinoma metastasis. Analysis of tumor-bearing lung tissues revealed that a large portion of tumor-infiltrating CD8(+) T cells are FasL(+), and decitabine and vorinostat-mediated tumor-suppression efficacy was significantly decreased in Fas(gld) mice compared with wild-type mice, suggesting a critical role for FasL in decitabine and vorinostat-mediated tumor suppression in vivo. Consistent with their function in apoptosis sensitization, decitabine and vorinostat significantly increased the efficacy of CTL adoptive transfer immunotherapy in an experimental metastasis mouse model. Thus, our data suggest that combined modalities of chemotherapy to sensitize the tumor cell to Fas-mediated apoptosis and CTL immunotherapy is an effective approach for the suppression of colon cancer metastasis.
Collapse
Affiliation(s)
- Dafeng Yang
- Department of Biochemistry and Molecular Biology, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | |
Collapse
|
11
|
Abstract
The airway epithelium functions as a barrier and front line of host defense in the lung. Apoptosis or programmed cell death can be elicited in the epithelium as a response to viral infection, exposure to allergen or to environmental toxins, or to drugs. While apoptosis can be induced via activation of death receptors on the cell surface or by disruption of mitochondrial polarity, epithelial cells compared to inflammatory cells are more resistant to apoptotic stimuli. This paper focuses on the response of airway epithelium to apoptosis in the normal state, apoptosis as a potential regulator of the number and types of epithelial cells in the airway, and the contribution of epithelial cell apoptosis in important airways diseases.
Collapse
|
12
|
Tsuda M, Kawaida R, Kobayashi K, Shinagawa A, Sawada T, Yamada R, Yamamoto K, Aigaki T. POSH promotes cell survival inDrosophilaand in human RASF cells. FEBS Lett 2010; 584:4689-94. [DOI: 10.1016/j.febslet.2010.10.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/20/2010] [Indexed: 10/18/2022]
|
13
|
Wang L, Zhao S, Wang HX, Zou P. Inhibition of NF-kappa B can enhance Fas-mediated apoptosis in leukemia cell line HL-60. ACTA ACUST UNITED AC 2010; 4:323-8. [DOI: 10.1007/s11684-010-0026-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Accepted: 11/28/2009] [Indexed: 01/17/2023]
|
14
|
Sharma V, Tewari R, Sk UH, Joseph C, Sen E. Ebselen sensitizes glioblastoma cells to Tumor Necrosis Factor (TNFα)-induced apoptosis through two distinct pathways involving NF-κB downregulation and Fas-mediated formation of death inducing signaling complex. Int J Cancer 2008; 123:2204-12. [DOI: 10.1002/ijc.23771] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
15
|
Kunigal S, Lakka SS, Joseph P, Estes N, Rao JS. Matrix metalloproteinase-9 inhibition down-regulates radiation-induced nuclear factor-kappa B activity leading to apoptosis in breast tumors. Clin Cancer Res 2008; 14:3617-26. [PMID: 18519796 DOI: 10.1158/1078-0432.ccr-07-2060] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE Novel strategies are needed to prevent the high mortality rates of several types of cancer. These high rates stem from tumor resistance to radiation therapy, which is thought to result from the induction of matrix metalloproteinases (MMP) and plasminogen activators. In the present study, we show that the modulation of MMP-9 expression, using adenoviral-mediated transfer of the antisense MMP-9 gene (MMP-9 adenoviral construct, Ad-MMP-9), affects breast cancer sensitivity to radiation. EXPERIMENTAL DESIGN In the present study, we used antisense Ad-MMP-9 to down-regulate the expression of MMP-9 in MDA MB 231 breast cancer cell lines in vitro before irradiation and subsequently incubated cells in hypoxic condition. In vivo studies were done with orthotopic breast tumors, and radiosensitivity was evaluated both in vitro and in vivo. RESULTS Ad-MMP-9 infection resulted in down-regulation of radiation-induced levels of hypoxia-inducible factor 1 alpha and MMP-9 under hypoxic conditions in MDA MB 231 breast cancer cells. In addition, Ad-MMP-9, in combination with radiation, decreased levels of the transcription factors nuclear factor-kappaB and activator protein 1, both of which contribute to the radioresistance of breast tumors. Finally, the triggering of the Fas-Fas ligand apoptotic cascade, which resulted in the cleavage of PARP-1 and caspase-10, caspase-3, and caspase-7, signifies the efficiency of combined treatment of Ad-MMP-9 and radiation. Treatment with Ad-MMP-9 plus radiation completely regressed tumor growth in orthotopic breast cancer model. CONCLUSIONS In summary, integrating gene therapy (adenovirus-mediated inhibition of MMP-9) with radiotherapy could have a synergistic effect, thereby improving the survival of patients with breast cancer.
Collapse
Affiliation(s)
- Sateesh Kunigal
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Drive, Peoria, IL 61605, USA
| | | | | | | | | |
Collapse
|
16
|
Lee KH, Feig C, Tchikov V, Schickel R, Hallas C, Schütze S, Peter ME, Chan AC. The role of receptor internalization in CD95 signaling. EMBO J 2006; 25:1009-23. [PMID: 16498403 PMCID: PMC1409734 DOI: 10.1038/sj.emboj.7601016] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Accepted: 01/30/2006] [Indexed: 11/08/2022] Open
Abstract
Activation of the cell surface CD95 receptor triggers a cascade of signaling events, including assembly of the death-inducing signaling complex (DISC), that culminate in cellular apoptosis. In this study, we demonstrate a general requirement of receptor internalization for CD95 ligand-mediated DISC amplification, caspase activation and apoptosis in type I cells. Recruitment of DISC components to the activated receptor predominantly occurs after the receptor has moved into an endosomal compartment and blockade of CD95 internalization impairs DISC formation and apoptosis. In contrast, CD95 ligand stimulation of cells unable to internalize CD95 results in activation of proliferative Erk and NF-kappaB signaling pathways. Hence, the subcellular localization and internalization pathways of CD95 play important roles in controlling activation of distinct signaling cascades to determine divergent cellular fates.
Collapse
Affiliation(s)
- Kyeong-Hee Lee
- Department of Immunology, Genentech Inc., South San Francisco, CA, USA
| | - Christine Feig
- The Ben May Institute for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Vladimir Tchikov
- Institute of Immunology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Robert Schickel
- The Ben May Institute for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Cora Hallas
- Institute of Immunology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Stefan Schütze
- Institute of Immunology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Marcus E Peter
- The Ben May Institute for Cancer Research, University of Chicago, Chicago, IL, USA
- These authors shared senior authorship
| | - Andrew C Chan
- Department of Immunology, Genentech Inc., South San Francisco, CA, USA
- These authors shared senior authorship
- Department of Immunology, Genentech Inc., MS-34, Bldg. 12-281, 1 DNA Way, South San Francisco, CA 94080, USA. Tel.: +1 650 225 8104; Fax: +1 650 225 8136; E-mail: or
| |
Collapse
|
17
|
Xiao Q, Hsu C, Chen H, Ma X, Xu J, Lee JM. Characterization of cis-regulatory elements of the vascular endothelial growth inhibitor gene promoter. Biochem J 2005; 388:913-20. [PMID: 15702971 PMCID: PMC1183472 DOI: 10.1042/bj20041739] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
VEGI (vascular endothelial growth inhibitor), a member of the tumour necrosis factor superfamily, has been reported to inhibit endothelial cell proliferation, angiogenesis and tumour growth. We identified and cloned approx. 2.2 kb of the VEGI promoter from mouse cerebral endothelial cells. The promoter contained an atypical TATA-box-binding protein sequence TAAAAAA residing at -32/-26 relative to the transcription initiation site (+1), 83 bp upstream from the ATG start codon. To investigate critical sequences in the VEGI promoter, a series of deleted and truncated segments were constructed from a 2300 bp promoter construct (-2201/+96) linked to a luciferase reporter gene. Transient transfection of cerebral microvascular cells (bEND.3) and rat C6 glioma cells demonstrated that a 1700 bp deletion from the -2201 to -501 did not significantly affect promoter activity; however, a truncated construct (-501/+96) lacking the region between -312 and -57 resulted in nearly 90% loss of promoter activity. A consensus NF-kappaB (nuclear factor kappaB) and several SP1 (specificity protein-1)-binding sequences were identified within the deleted segment. Supershift analysis revealed that NF-kappaB subunits, p50 and p65, interacted with the VEGI promoter. Exposure of cerebral endothermic cells to the pro-inflammatory cytokine, tumour necrosis factor-alpha, increased VEGI mRNA levels and DNA-binding activities, whereas an NF-kappaB inhibitor attenuated this increase. In addition, p65 overexpression enhanced, whereas p50 overexpression decreased, the luciferase activity. Furthermore, mutation of the NF-kappaB DNA binding site blocked this p65- and tumour necrosis factor-alpha-induced luciferase activity. These findings suggest that the transcription factor NF-kappaB plays an important role in the regulation of VEGI expression.
Collapse
Affiliation(s)
- Qingli Xiao
- *The Hope Center for Neurological Disorders, Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO 63110, U.S.A
| | - Chung Y. Hsu
- *The Hope Center for Neurological Disorders, Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO 63110, U.S.A
- †Taipei Medical University, Taipei, Taiwan
| | - Hong Chen
- *The Hope Center for Neurological Disorders, Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO 63110, U.S.A
| | - Xiucui Ma
- *The Hope Center for Neurological Disorders, Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO 63110, U.S.A
| | - Jan Xu
- *The Hope Center for Neurological Disorders, Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO 63110, U.S.A
| | - Jin-Moo Lee
- *The Hope Center for Neurological Disorders, Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO 63110, U.S.A
- To whom correspondence should be addressed (email )
| |
Collapse
|
18
|
Park H, Jung YK, Park OJ, Lee YJ, Choi JY, Choi Y. Interaction of Fas Ligand and Fas Expressed on Osteoclast Precursors Increases Osteoclastogenesis. THE JOURNAL OF IMMUNOLOGY 2005; 175:7193-201. [PMID: 16301623 DOI: 10.4049/jimmunol.175.11.7193] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We incidentally found that osteoclast precursors and mature osteoclasts express Fas ligand (FasL) as well as Fas, which was confirmed by flow cytometry, immunofluorescent staining, and RT-PCR. The aim of this study was to determine the role of FasL in differentiation and cell death of osteoclasts. To study the role of FasL in osteoclastogenesis, neutralizing anti-FasL mAb or rFasL was added during receptor activator of NF-kappaB ligand (RANKL)-induced osteoclastogenesis using bone marrow-derived macrophages. Neutralization of endogenous FasL by anti-FasL mAb decreased osteoclastogenesis, whereas rFasL enhanced osteoclast differentiation in a dose-dependent manner. In addition, rFasL up-regulated the secretion of osteoclastogenic cytokines, such as IL-1beta and TNF-alpha, and the activation of NF-kappaB. Functional blocking of IL-1beta and TNF-alpha using IL-1 receptor antagonist and soluble TNFR confirmed that those cytokines mediated the effect of FasL on osteoclastogenesis. The osteoclast precursors were relatively resistant to rFasL-induced apoptosis especially before RANKL treatment, resulting in minimal cell loss by rFasL treatment during osteoclastogenesis. Although rFasL increased the cell death of mature osteoclasts, growth factor withdrawal induced much more cell death. However, anti-FasL mAb did not affect the survival of mature osteoclasts, suggesting that the endogenous FasL does not have a role in the apoptosis of osteoclasts. Finally, in contrast to the effect on apoptosis, rFasL-assisted osteoclastogenesis was not mediated by caspases. In conclusion, FasL has a novel function in bone homeostasis by enhancing the differentiation of osteoclasts, which was not considered previously.
Collapse
Affiliation(s)
- Hyewon Park
- Department of Oromaxillofacial Infection & Immunity and Dental Research Institute, School of Dentistry, Seoul National University, Jongro-gu, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
19
|
Lavastre V, Chiasson S, Cavalli H, Girard D. Viscum album agglutinin-I induces apoptosis and degradation of cytoskeletal proteins via caspases in human leukaemia eosinophil AML14.3D10 cells: differences with purified human eosinophils. Br J Haematol 2005; 130:527-35. [PMID: 16098066 DOI: 10.1111/j.1365-2141.2005.05633.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Although there are several agents that induce neutrophil apoptosis, few are known as inducers of eosinophil apoptosis. As eosinophils are potent effector cells contributing to allergic inflammation and asthma, we investigated whether the pro-apoptotic agent Viscum album agglutinin-I (VAA-I) could induce eosinophil apoptosis. VAA-I was found to induce apoptosis in eosinophilic AML14.3D10 (3D10) cells and that these cells expressed caspases-1, -2, -3, -4, -7, -8, -9 and -10. VAA-I-induced gelsolin degradation was reversed by the pan-caspase inhibitor N-benzyloxycarbonyl-V-A-D-O-methylfluoromethyl ketone (z-VAD). Also, paxillin, vimentin and lamin B1 were cleaved by caspases in VAA-I-induced 3D10 cells. VAA-I activated caspase-3 and -8 in 3D10 cells but, unlike z-VAD, treatment with a caspase-8 inhibitor slightly reversed apoptosis. Treatment of purified human eosinophils with VAA-I was found to induce apoptosis, degradation of gelsolin and lamin B1, but unlike 3D10 cells, cleavage of lamin B1 and cell apoptosis was not reversed by z-VAD. We conclude that VAA-I is a potent inducer of eosinophil apoptosis and that proteases other than those inhibited by z-VAD in 3D10 cells are involved in VAA-I-induced peripheral blood eosinophil apoptosis and lamin B1 cleavage. Thus, VAA-I represents a potential candidate for the reduction of the number of eosinophils in diseases where they play important roles.
Collapse
Affiliation(s)
- Valérie Lavastre
- INRS-Institut Armand-Frappier, Université du Québec, Pointe-Claire, Canada
| | | | | | | |
Collapse
|
20
|
Matalova E, Tucker AS, Misek I. Apoptosis-related factors (Fas receptor, Fas ligand, FADD) in early tooth development of the field vole (Microtus agrestis). Arch Oral Biol 2005; 50:165-9. [PMID: 15721145 DOI: 10.1016/j.archoralbio.2004.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2004] [Accepted: 10/12/2004] [Indexed: 11/24/2022]
Abstract
Fas (CD95/APO-1) belongs to the TNF receptor (TNFR) family. Fas ligand binding followed by Fas-receptor oligomerisation leads to formation of a death-inducing signal complex starting with recruitment of the Fas-adapter protein (FADD). Components of this initiation complex (Fas, Fas-L, FADD) were correlated with apoptotic cells, detected by specific DNA fragmentation and morphological criteria. Apoptotic cells can be detected throughout the embryonic development of molar teeth. Restricted temporospatial distribution suggests several important roles for apoptosis in tooth morphogenesis. However, the mechanisms employed in dental apoptosis remain unclear. Frontal sections of the field vole at stage 13.5-15.5 of embryonic development were exploited to investigate and correlate location of Fas, Fas-ligand, FADD molecules and apoptosis in developing first molars by immunohistochemistry. During these stages the primary enamel knot appears and is gradually terminated by apoptosis. Initially, apoptotic cells were demonstrated in the most superficial layer of the dental lamina. The number of TUNEL-positive cells expanded from late bud to cap stages. Restricted areas of apoptotic cells were found in the stalk and primary enamel knot. Fas, Fas-L and FADD were co-localised, particularly in the primary enamel knot, and the stalk, correlating with the occurrence of apoptosis in these areas. Fas-L, however, was also found in proliferating parts of the developing tooth germ, such as in the cervical loops. Interestingly, FADD molecules were also observed in areas, where Fas protein was not detected. According to the immunohistochemical data, Fas-mediated signalling may have a triggering or enhancing role in dental apoptosis. This remains to be functionally confirmed.
Collapse
Affiliation(s)
- E Matalova
- Laboratory of Genetics and Embryology, IAPG, Czech Academy of Sciences, Veveri 97, 602 00 Brno, Czech Republic.
| | | | | |
Collapse
|
21
|
Ivory K, Martin R, Hughes DA. Significant presence of terminally differentiated T cells and altered NF-κB and I-κBα interactions in healthy ageing. Exp Gerontol 2004; 39:567-76. [PMID: 15050292 DOI: 10.1016/j.exger.2003.09.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2003] [Revised: 09/10/2003] [Accepted: 09/16/2003] [Indexed: 10/26/2022]
Abstract
The risk of infection and cancer increases dramatically beyond middle age, when T-cell function is noticeably altered. Nevertheless, many elderly people remain in apparently good health. To identify immunological adaptations favouring longevity, a pilot study was undertaken to compare peripheral blood T cells from healthy volunteers aged 18-25 years with those >65 years. Instead of preserved immune function in the elderly, there was an emergence of haematopoietic space particularly affecting T- and B-cell numbers, together with early signs of immunoglobulin dysregulation. Age-associated proliferative defects were present irrespective of the stimuli used. A higher constitutive expression of NF-kappaB and I-kappaBalpha in the nuclei of peripheral lymphocytes from the elderly remained unaltered by activation stimuli, despite the presence of exogenous cytokines. Nevertheless, activation resulted in their higher CD95 upregulation and more intracellular bcl-2 (suggesting a survival advantage), but lower CD27, CD28 and CD45Rb expression. The presence of CD45RO(+) CD45Rb(-) populations was unique to the elderly and their lower replicative potential was not due to the presence of CD25(+) regulatory T cells. These data collectively suggest altered gene regulation and the accumulation of terminally differentiated T cells during healthy ageing.
Collapse
Affiliation(s)
- Kamal Ivory
- Immunology Group, Nutrition Division, Institute of Food Research, Norwich Research Park, Norwich, East Anglia, UK.
| | | | | |
Collapse
|
22
|
Park SM, Park HY, Lee TH. Functional effects of TNF-alpha on a human follicular dendritic cell line: persistent NF-kappa B activation and sensitization for Fas-mediated apoptosis. THE JOURNAL OF IMMUNOLOGY 2004; 171:3955-62. [PMID: 14530313 DOI: 10.4049/jimmunol.171.8.3955] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Follicular dendritic cells (FDC) play crucial roles in germinal center (GC) formation and differentiation of GC B cells. FDC functions are influenced by cytokines produced in the GC. Among the GC cytokines, TNF is known to be essential for the formation and maintenance of the FDC network in the GC. We found that TNF is a mitogenic growth factor to an established FDC-like cell line, HK cells. Differing from most cell types which become desensitized to TNF action, HK cells exhibited persistent TNF signaling, as demonstrated by prolonged and biphasic NF-kappaB activation even after 3 days of TNF treatment. As a result, antiapoptotic genes including TNFR-associated factors 1 and 2, and cellular inhibitor of apoptosis proteins 1 and 2 were persistently induced by TNF, leading to the protection against TNF-mediated cell death. However, TNF pretreatment enhanced Fas-mediated apoptosis by up-regulating surface Fas expression in an NF-kappaB-dependent pathway. During the GC responses, proliferation followed by FDC death has not been documented. However, our in vitro results suggest that FDCs proliferate in response to TNF, and die by Fas-mediated apoptosis whose susceptibility is enhanced by TNF, representing a mode of action for TNF in the maintenance of FDC networks by regulating the survival or death of FDC.
Collapse
Affiliation(s)
- Sun-Mi Park
- Department of Biology and Protein Network Research Center, Yonsei University, Seodaemoon, Seoul, Korea
| | | | | |
Collapse
|
23
|
Mandelker L. The natural activities of cells, the role of reactive oxygen species, and their relation to antioxidants, nutraceuticals, botanicals, and other biologic therapies. Vet Clin North Am Small Anim Pract 2004; 34:39-66. [PMID: 15032125 DOI: 10.1016/j.cvsm.2003.09.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
There have been remarkable advances in molecular and cell biology that define the mechanisms of how various supplements function in and around cells. Current evidence strongly supports the probability that cellular functions and cellular responses that pertain to inflammation, disease, and life and death activity can be modulated with supplementation; however, the complexity of each individual's reaction and the vast differences in physiologic influences makes clinical research difficult in regard to clinical studies using antioxidant and biologic therapies. Not enough is known specifically about each supplement and its interactions with cells, nor is enough understood about how the body compensates or reacts to such applications. What works well in one individual or species might work differently in another. In addition, not all antioxidants are created equally, and discrepancies in purity and absorption can occur. It must also be determined whether or not less than optimum levels or infrequent usage will produce the same physiological effects. Not everyone--nor every species of animal--responds in the same manner to supplements, which might account for the variations in clinical research. The cellular effects of antioxidants and other supplements are well defined and meaningful, and their clinical application looks promising despite individual variations. Combinations of antioxidants are synergistic and support cellular functions, effects that are often not apparent with individual agents. Such combinations offer a variety of mechanisms for reducing oxygen metabolites in tissues, altering signaling pathways, and modulating transcription factors, and they might play key roles in reducing the damage afforded by ROS. It is the author's opinion that combinations of antioxidants are best suited for clinical application in modulating disease and reducing premature aging when caused by excessive free radical accumulation. Clinicians should approach clinical application of these supplements based on the best available scientific research and species-specific information available.
Collapse
Affiliation(s)
- Lester Mandelker
- Community Veterinary Hospital, 1631 W. Bay Drive, Largo, FL 33770, USA.
| |
Collapse
|