1
|
Sutter SO, Tobler K, Seyffert M, Lkharrazi A, Zöllig J, Schraner EM, Vogt B, Büning H, Fraefel C. Interferon-γ inducible factor 16 (IFI16) restricts adeno-associated virus type 2 (AAV2) transduction in an immune-modulatory independent way. J Virol 2024; 98:e0011024. [PMID: 38837381 PMCID: PMC11338077 DOI: 10.1128/jvi.00110-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/28/2024] [Indexed: 06/07/2024] Open
Abstract
We determined the transcription profile of adeno-associated virus type 2 (AAV2)-infected primary human fibroblasts. Subsequent analysis revealed that cells respond to AAV infection through changes in several significantly affected pathways, including cell cycle regulation, chromatin modulation, and innate immune responses. Various assays were performed to validate selected differentially expressed genes and to confirm not only the quality but also the robustness of the raw data. One of the genes upregulated in AAV2-infected cells was interferon-γ inducible factor 16 (IFI16). IFI16 is known as a multifunctional cytosolic and nuclear innate immune sensor for double-stranded as well as single-stranded DNA, exerting its effects through various mechanisms, such as interferon response, epigenetic modifications, or transcriptional regulation. IFI16 thereby constitutes a restriction factor for many different viruses among them, as shown here, AAV2 and thereof derived vectors. Indeed, the post-transcriptional silencing of IFI16 significantly increased AAV2 transduction efficiency, independent of the structure of the virus/vector genome. We also show that IFI16 exerts its inhibitory effect on AAV2 transduction in an immune-modulatory independent way by interfering with Sp1-dependent transactivation of wild-type AAV2 and AAV2 vector promoters. IMPORTANCE Adeno-associated virus (AAV) vectors are among the most frequently used viral vectors for gene therapy. The lack of pathogenicity of the parental virus, the long-term persistence as episomes in non-proliferating cells, and the availability of a variety of AAV serotypes differing in their cellular tropism are advantageous features of this biological nanoparticle. To deepen our understanding of virus-host interactions, especially in terms of antiviral responses, we present here the first transcriptome analysis of AAV serotype 2 (AAV2)-infected human primary fibroblasts. Our findings indicate that interferon-γ inducible factor 16 acts as an antiviral factor in AAV2 infection and AAV2 vector-mediated cell transduction in an immune-modulatory independent way by interrupting the Sp1-dependent gene expression from viral or vector genomes.
Collapse
Affiliation(s)
| | - Kurt Tobler
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | - Michael Seyffert
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | - Anouk Lkharrazi
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | - Joël Zöllig
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | | | - Bernd Vogt
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| | - Hildegard Büning
- Institute of
Experimental Hematology, Hannover Medical
School, Hannover,
Germany
| | - Cornel Fraefel
- Institute of Virology,
University of Zurich,
Zurich, Switzerland
| |
Collapse
|
2
|
Lin W, Zhao Z, Du W, Ni Z, Pan C, Fang P, Li J, ZhuGe L, Jin S. Interferon-Gamma-Inducible Protein 16 Inhibits Hepatocellular Carcinoma via Interferon Regulatory Factor 3 on Chemosensitivity. Dig Dis Sci 2024; 69:491-501. [PMID: 38170337 DOI: 10.1007/s10620-023-08175-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/29/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND AND AIM Previous reports have suggested IFI16 as a tumor suppressor in hepatocellular carcinoma (HC). Nonetheless, the biological significance of IFI16 and its mechanism concerning resistance to cisplatin (DDP) in HC requires further exploration. METHODS Samples of tumor and corresponding para-carcinoma tissues were acquired from patients with HC. Furthermore, DDP-resistant cell lines of HC, specifically HCC, Huh7 and Hepatoblastoma, HepG3, were generated by gradually increasing the concentration of DDP. Cell apoptosis and DNA damage were evaluated by utilizing flow cytometry assay and TUNEL staining. The interaction between IFI16 and interferon regulatory factor 3 (IRF3) proteins were analyzed using Co-Immunoprecipitation (Co-IP) assay. In vivo assays were conducted by establishing HC subcutaneous xenograft tumor models. RESULTS The study found a reduction in IFI16 expression in both HC tissues and DDP-resistant HC cell lines. The binding of IFI16 to IRF3 regulated DNA damage-associated markers in vitro. Overexpression of IFI16 heightened the susceptibility of DDP-induced apoptosis and DNA damage, which was counteracted by IRF3 knockdown, while strengthened by IRF3 overexpression. Moreover, overexpression of IFI16 diminished in vivo DDP-resistant HC tumorigenicity. CONCLUSION In summary, our findings suggest that IFI16 serves as a tumor suppressor in HC by promoting DNA damage via its interaction with IRF3, thereby reversing DDP resistance.
Collapse
Affiliation(s)
- Wei Lin
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China.
| | - Zhiguang Zhao
- Department of Pathology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenjun Du
- Department of Liver Diseases, Shandong Public Health Clinical Center, Shangdong University, Jinan, Shangdong, China
| | - Zhonglin Ni
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenwei Pan
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| | - Peipei Fang
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| | - Jie Li
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| | - Lu ZhuGe
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| | - Shuanghong Jin
- Department of Infectious Diseases, The Second Affiliated Hospital of Wenzhou Medical University, #1111 of Wenzhou Wenzhou Avenue, Longwan District, Wenzhou, Zhejiang, China
| |
Collapse
|
3
|
Azumi Y, Koma YI, Tsukamoto S, Kitamura Y, Ishihara N, Yamanaka K, Nakanishi T, Miyako S, Urakami S, Tanigawa K, Kodama T, Nishio M, Shigeoka M, Kakeji Y, Yokozaki H. IFI16 Induced by Direct Interaction between Esophageal Squamous Cell Carcinomas and Macrophages Promotes Tumor Progression via Secretion of IL-1α. Cells 2023; 12:2603. [PMID: 37998338 PMCID: PMC10670642 DOI: 10.3390/cells12222603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
Tumor-associated macrophages (TAMs), one of the major components of the tumor microenvironment, contribute to the progression of esophageal squamous cell carcinoma (ESCC). We previously established a direct co-culture system of human ESCC cells and macrophages and reported the promotion of malignant phenotypes, such as survival, growth, and migration, in ESCC cells. These findings suggested that direct interactions between cancer cells and macrophages contribute to the malignancy of ESCC, but its underlying mechanisms remain unclear. In this study, we compared the expression levels of the interferon-induced genes between mono- and co-cultured ESCC cells using a cDNA microarray and found that interferon-inducible protein 16 (IFI16) was most significantly upregulated in co-cultured ESCC cells. IFI16 knockdown suppressed malignant phenotypes and also decreased the secretion of interleukin-1α (IL-1α) from ESCC cells. Additionally, recombinant IL-1α enhanced malignant phenotypes of ESCC cells through the Erk and NF-κB signaling. Immunohistochemistry revealed that high IFI16 expression in human ESCC tissues tended to be associated with disease-free survival and was significantly associated with tumor depth, lymph node metastasis, and macrophage infiltration. The results of this study reveal that IFI16 is involved in ESCC progression via IL-1α and imply the potential of IFI16 as a novel prognostic factor for ESCC.
Collapse
Affiliation(s)
- Yuki Azumi
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.K.); (K.T.); (Y.K.)
| | - Yu-ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Shuichi Tsukamoto
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Yu Kitamura
- Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.K.); (K.T.); (Y.K.)
| | - Nobuaki Ishihara
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Keitaro Yamanaka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takashi Nakanishi
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.K.); (K.T.); (Y.K.)
| | - Shoji Miyako
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.K.); (K.T.); (Y.K.)
| | - Satoshi Urakami
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Kohei Tanigawa
- Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.K.); (K.T.); (Y.K.)
| | - Takayuki Kodama
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Mari Nishio
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Manabu Shigeoka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Yoshihiro Kakeji
- Division of Gastro-Intestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.K.); (K.T.); (Y.K.)
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (Y.A.); (S.T.); (N.I.); (K.Y.); (T.N.); (S.M.); (S.U.); (T.K.); (M.N.); (M.S.); (H.Y.)
| |
Collapse
|
4
|
Justice JL, Cristea IM. Nuclear antiviral innate responses at the intersection of DNA sensing and DNA repair. Trends Microbiol 2022; 30:1056-1071. [PMID: 35641341 PMCID: PMC9560981 DOI: 10.1016/j.tim.2022.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 01/13/2023]
Abstract
The coevolution of vertebrate and mammalian hosts with DNA viruses has driven the ability of host cells to distinguish viral from cellular DNA in the nucleus to induce intrinsic immune responses. Concomitant viral mechanisms have arisen to inhibit DNA sensing. At this virus-host interface, emerging evidence links cytokine responses and cellular homeostasis pathways, particularly the DNA damage response (DDR). Nuclear DNA sensors, such as the interferon (IFN)-γ inducible protein 16 (IFI16), functionally intersect with the DDR regulators ataxia telangiectasia mutated (ATM) and DNA-dependent protein kinase (DNA-PK). Here, we discuss accumulating knowledge for the DDR-innate immunity signaling axis. Through the lens of this infection-driven signaling axis, we present host and viral molecular strategies acquired to regulate autoinflammation and antiviral responses.
Collapse
Affiliation(s)
- Joshua L Justice
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA.
| |
Collapse
|
5
|
Nav1.6 promotes the progression of human follicular thyroid carcinoma cells via JAK-STAT signaling pathway. Pathol Res Pract 2022; 236:153984. [PMID: 35753135 DOI: 10.1016/j.prp.2022.153984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022]
Abstract
Follicular thyroid carcinoma (FTC) is one of the most common malignant tumors of the endocrine system. Recent studies have shown that voltage-gated sodium channels (VGSCs) affect the proliferation, migration, and invasion of tumor cells. However, the expression and functions of VGSCs, and the molecular pathways activated by VGSCs in FTC cells remain unclear. Our studies revealed that the expression of Nav1.6, encoded by SCN8A, was the predominantly upregulated subtype of VGSCs in FTC tissues. Knockdown of Nav1.6 significantly inhibited the proliferation, epithelial-mesenchymal transition and invasiveness of FTC cells. Using gene set enrichment analysis and Kyoto Encyclopedia of Genes and Genomics, SCN8A was predicted to be related to the JAK-STAT signaling pathway. Hence, we targeted the JAK-STAT pathway and demonstrated that Nav1.6 enhanced FTC cell proliferation, epithelial-mesenchymal transition, and invasion by phosphorylating JAK2 to activate STAT3. Furthermore, downregulating the expression of Nav1.6 improve the susceptibility of FTC cells to ubenimex in vitro. These results suggest Nav1.6 accelerates FTC progression through JAK/STAT signaling and may be a potential target for FTC therapy.
Collapse
|
6
|
Borucka J, Sterzyńska K, Kaźmierczak D, Świerczewska M, Nowacka M, Wojtowicz K, Klejewski A, Nowicki M, Zabel M, Ramlau R, Januchowski R. The significance of interferon gamma inducible protein 16 (IFI16) expression in drug resistant ovarian cancer cell lines. Biomed Pharmacother 2022; 150:113036. [PMID: 35489285 DOI: 10.1016/j.biopha.2022.113036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Inherent or developed during treatment drug resistance is the main reason for the low effectiveness of chemotherapy in ovarian cancer. IFI16 is a cytoplasmic/nuclear protein involved in response to virus's infection and cell cycle arrest associated with the cellular senescence. METHODS Here we performed a detailed IFI16 expression analysis in ovarian cancer cell lines sensitive (A2780) and resistant to doxorubicin (DOX) (A2780DR1 and A2780DR2) and paclitaxel (PAC) (A2780PR1). IFI16 mRNA level, protein level in the nuclear and cytoplasmic fraction (Western blot analysis), the protein expression in cancer cells and nuclei (immunofluorescence analysis) and cancer patient lesions (immunohistochemistry) were performed in this study. RESULTS We observed upregulation of IFI16 expression in drug resistant cell lines with dominant cytoplasmic localization in DOX-resistant cell lines and nuclear one in the PAC-resistant cell line. The most abundantly overexpressed isoforms of IFI16 were IFI16A and IFI16C. Finally, an analysis of a histological type of ovarian cancer (immunohistochemistry) showed expression in serous ovarian cancer. CONCLUSIONS Expression of IFI16 in drug-resistant cell lines suggests its role in drug resistance development in ovarian cancer. Expression in serous ovarian cancer suggests its role in the pathogenesis of this histological type.
Collapse
Affiliation(s)
- Justyna Borucka
- Department of Oncology, Poznan University of Medical Sciences, Szamarzewskiego 82/84, 60-569 Poznań, Poland
| | - Karolina Sterzyńska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Dominika Kaźmierczak
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Monika Świerczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Marta Nowacka
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Karolina Wojtowicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Andrzej Klejewski
- Department of Nursing, Poznan University of Medical Sciences, Smoluchowskiego 11 St., 60-179 Poznań, Poland; Department of Obstetrics and Women's Diseases, Poznan University of Medical Sciences, Polna 33 St, 60-535 Poznań, Poland
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland; Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, Zyty 28 St, 65-046 Zielona Gora, Poland
| | - Rodryg Ramlau
- Department of Oncology, Poznan University of Medical Sciences, Szamarzewskiego 82/84, 60-569 Poznań, Poland
| | - Radosław Januchowski
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, Zyty 28 St, 65-046 Zielona Gora, Poland.
| |
Collapse
|
7
|
Zou Y, Zhang J, Zhang L, Yan X. Interferon-induced protein 16 expression in colorectal cancer and its correlation with proliferation and immune signature markers. Oncol Lett 2021; 22:687. [PMID: 34434286 PMCID: PMC8335744 DOI: 10.3892/ol.2021.12948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Interferon-induced protein 16 (IFI16) is important for innate immune recognition of foreign/damaged DNA. Abnormal IFI16 expression is closely related to the occurrence of multiple malignant tumours, but its expression pattern in colorectal cancer (CRC) remains unclear. The present study aimed to investigated IFI16 expression and association with cell proliferation in CRC tissues and adjacent normal tissues. A multiplex immunofluorescence panel of antibodies against IFI16, Ki-67 and phosphorylated (p)-ERK1/2 was applied to assess a tissue microarray (TMA). The TMA included 77 CRC samples and 74 normal adjacent tissue samples which were collected from The First People's Hospital of Yunnan Province (Kunming, China) (3 paracancerous tissues were lost because of repeated cutting). Immunohistochemistry was used to detect CD8+ tumour-infiltrating lymphocyte (TIL) abundance and programmed death-ligand 1 (PD-L1) expression in cancer tissues. The present study demonstrated that IFI16 localized to the nucleus of CRC cells. Although IFI16 was weakly expressed in normal mucosal epithelial cells, absent to strong expression was detectable in different patients with CRC. Typically, IFI16 was not co-localized with Ki-67 within CRC cells. The multiplex immunofluorescence data demonstrated that the proportion of IFI16-/Ki-67+ cells from CRC tissues was 57.13%; however, that of IFI16+/Ki-67+ cells was 1.50%. The IFI16-/Ki-67+ phenotype was significantly positively associated with the tumor-node-metastasis stage and was marginally significantly correlated with lymph node metastasis. p-ERK1/2 protein was primarily localized to the cytoplasm and cell membrane of CRC cells and sometimes to the nucleus. Although, IFI16 demonstrated a strong correlation with p-ERK1/2, IFI16 did not co-localize with p-ERK1/2 and the proportion of IFI16 and p-ERK1/2 double-negative CRC cells was 84.95%. IFI16 expression displayed no significant association with CD8+ TILs or PD-L1. However, a strong positive correlation between CD8+ TILs and PD-L1 was observed. High CD8+ TIL infiltration in CRC tissue was associated with lower lymph node metastasis and tumor-node-metastasis stage. In summary, the results of the present study provided a novel insight for the role of IFI16 in CRC occurrence via the regulation of cancer cell proliferation.
Collapse
Affiliation(s)
- Yunlian Zou
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Jinping Zhang
- Institute of Medical Sciences, Yunnan Blood Disease Clinical Medical Center, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Lichen Zhang
- Medical Faculty, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Xinmin Yan
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
8
|
Zhu M, Nan Y, Zhai M, Wang M, Shao Y, Blair HT, Morris ST, Kenyon PR, Zhao Z, Zhang H. Comparative profiling of the resistance of different genotypes of mannose-binding lectin to Mycoplasma pneumoniae infection in Chinese Merino sheep based on high-throughput sequencing technology. Vet Immunol Immunopathol 2021; 233:110183. [PMID: 33476923 DOI: 10.1016/j.vetimm.2021.110183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 12/28/2020] [Accepted: 12/31/2020] [Indexed: 10/22/2022]
Abstract
Mannose-binding lectin (MBL) glycoproteins in blood can selectively recognise lectins on the surface of bacteria, and play an important role in natural immunity. Micro RNAs (miRNAs) are key molecules that regulate gene expression at the post-transcriptional level in vivo, and their pathways are specific and effective. Previous studies indicate that small RNAs such as miRNAs perform regulatory roles in immunology. Herein, we investigated differential expression of miRNAs during MBL protein immunotherapy in sheep following treatment with different MBL genotypes (resistant and susceptible), and identified miRNAs linked to different target genes and pathways. RNA was extracted from liver tissue of resistant and susceptible sheep, miRNAs were identified by high-throughput sequencing, and differentially expressed miRNAs were analysed by SOAP to predict target genes and biological pathways. Results: Some miRNAs (oar-mir-143, oar-mir-10b, oar-mir-382, oar-mir-432 and oar-mir-379) were up-regulated, while others were down-regulated. GPATCH3 and DNAJC5 were predicted target genes of oar-mir-379, DMRT1 and GATA4 were linked to oar-mir-382, and oar-mir-432 was associated with STAT2, DMRT1 and ATG16L1. Identification of miRNAs differentially expressed in resistant and susceptible sheep may expand our understanding of miRNAs in immune regulation, and the role of MBL in innate immunity.
Collapse
Affiliation(s)
- Mengting Zhu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, PR China; State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, 832000, PR China
| | - Ying Nan
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, PR China
| | - Mengting Zhai
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, PR China
| | - Mingyuan Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, PR China
| | - Yanyan Shao
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, PR China
| | - Hugh T Blair
- Institute Veterinary, Animal & Biomedical Sciences, Massey University, Auckland, Palmerston North, New Zealand
| | - Stephen Todd Morris
- Institute Veterinary, Animal & Biomedical Sciences, Massey University, Auckland, Palmerston North, New Zealand
| | - Paul Richard Kenyon
- Institute Veterinary, Animal & Biomedical Sciences, Massey University, Auckland, Palmerston North, New Zealand
| | - Zongsheng Zhao
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, PR China.
| | - Hongmei Zhang
- First Affiliated Hospital, School of Medical College, Shihezi University, Shihezi, Xinjiang 832008, PR China.
| |
Collapse
|
9
|
IFI16 promotes human embryonic stem cell trilineage specification through interaction with p53. NPJ Regen Med 2020; 5:18. [PMID: 33298947 PMCID: PMC7596047 DOI: 10.1038/s41536-020-00104-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/25/2020] [Indexed: 11/08/2022] Open
Abstract
Transcriptional regulation plays an essential role in the self-renewal and differentiation of human embryonic stem cells (hESCs). However, how external signals disrupt the self-renewal regulatory network and further drive hESC differentiation remains largely unknown. Here, we found the immune regulative protein, gamma-interferon-inducible protein 16 (IFI16) was involved in the regulation of both self-renewal and differentiation gene expression during hESC trilineage specification through interaction with p53. IFI16 expression levels were upregulated through JNK activation. IFI16 knockdown delayed the downregulation of self-renewal gene expression and suppressed the upregulation of differentiation gene expression, while IFI16 overexpression accelerated trilineage specification. Furthermore, IFI16 stabilized p53-binding in the genome through IFI16-p53 interaction and differentially regulated self-renewal and differentiation gene expression. Together, our results suggest a particular role of IFI16 in differential gene expression regulation during trilineage specification of hESCs in a manner that is dependent on the genome-wide profile of p53-binding directed by IFI16-p53 interaction.
Collapse
|
10
|
Shi X, Li S, Wang L, Li H, Li Z, Wang W, Bai J, Sun Y, Li J, Li X. RalB degradation by dihydroartemisinin induces autophagy and IFI16/caspase-1 inflammasome depression in the human laryngeal squamous cell carcinoma. Chin Med 2020; 15:64. [PMID: 32577124 PMCID: PMC7304197 DOI: 10.1186/s13020-020-00340-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/26/2020] [Indexed: 02/04/2023] Open
Abstract
Background Interferon-inducible 16 (IFI16)/caspase-1 inflammasome activates and secretes IL-1β. However, it is still unclear whether the IFI16 inflammasome is involved in human laryngeal squamous cell carcinoma. Autophagy directly removed inflammasome components and limited early IL-1β production. RalB is required for the crosstalk between inflammasome and autophagy in macrophages. Dihydroartemisinin (DHA), the main derived ingredient of artemisinin, has a variety of biological activities. The mechanism of DHA in regulating the crosstalk between IFI16 inflammasome and autophagy by inhibiting RalB expression was analyzed in order to provide clues for new therapeutic methods in laryngeal cancer. Methods The expression of IFI16 was analyzed by Oncomine and GEPIA databases and detected by Western blot and immunohistochemistry. The relationship between IFI16 inflammasome and autophagy was investigated by transmission electron microscopy, immunofluorescence assay, etc. in Hep-2, Cal-27 and HeLa cells treated with DHA. The xenograft tumor of hep-2 cell in nude mice were used to assess the effect of DHA on laryngeal cancer. Results It was reported for the first time in this study that IFI16 was overexpressed and positively correlated with caspase-1 in laryngeal carcinoma tissues. DHA significantly inhibited the activation of inflammasome and reduced IL-1β production in the microenvironment of Hep-2 cell xenograft tumor in nude mice. Mechanistically, we found that DHA degraded RalB, inhibited USP33 expression, and triggered autophagy. Meanwhile, enhanced autophagy can reduce the expression of RalB and USP33. Furthermore, DHA promotes autophagy, which suppresses the activation of IFI16/caspase-1 inflammasome and IL-1β production. Conclusions Therefore, our findings demonstrate that DHA may act as a RalB inhibitor to regulate the crosstalk between autophagy and IFI16/caspase-1 inflammasome, which inhibits IL-1β production in tumor microenvironment.
Collapse
Affiliation(s)
- Xinli Shi
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, 050081 China.,Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200 China
| | - Shenghao Li
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200 China
| | - Li Wang
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Hui Li
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, 050081 China
| | - Zhen Li
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, 050081 China
| | - Weiyi Wang
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, 050081 China.,Department of Neurology, Children's Hospital of Hebei Province, Shijiazhuang, 050000 China
| | - Jing Bai
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, 050081 China
| | - Yajing Sun
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, 050081 China
| | - Jianchun Li
- Laboratory of Organ Fibrosis Prophylaxis and Treatment by Combine Traditional Chinese and Western Medicine, Research Center of Combine Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Xiaoming Li
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, 050081 China
| |
Collapse
|
11
|
Riva G, Biolatti M, Pecorari G, Dell’Oste V, Landolfo S. PYHIN Proteins and HPV: Role in the Pathogenesis of Head and Neck Squamous Cell Carcinoma. Microorganisms 2019; 8:microorganisms8010014. [PMID: 31861809 PMCID: PMC7023031 DOI: 10.3390/microorganisms8010014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/11/2019] [Accepted: 12/18/2019] [Indexed: 12/16/2022] Open
Abstract
In the last decades, the human papillomavirus (HPV) emerged as an etiological cause of head and neck squamous cell carcinoma (HNSCC), especially in the oropharynx. The role of two intracellular DNA sensors, which belong to the PYHIN family (interferon-inducible protein 16 (IFI16) and absent in melanoma 2 protein (AIM2)), has been analyzed in relation to HPV infection and head and neck carcinogenesis. In particular, IFI16 and AIM2 expression depends on HPV infection in HNSCC. They represent viral restriction factors and are key components of the intrinsic immunity activated against different viruses, including HPV. This review analyzed and summarized the recent findings about the role of PYHIN proteins in HPV+ and HPV− HNSCC.
Collapse
Affiliation(s)
- Giuseppe Riva
- Otorhinolaryngology Division, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (G.P.)
| | - Matteo Biolatti
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatrics, School of Medicine, University of Turin, 10126 Turin, Italy; (M.B.); (V.D.)
| | - Giancarlo Pecorari
- Otorhinolaryngology Division, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (G.P.)
| | - Valentina Dell’Oste
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatrics, School of Medicine, University of Turin, 10126 Turin, Italy; (M.B.); (V.D.)
| | - Santo Landolfo
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatrics, School of Medicine, University of Turin, 10126 Turin, Italy; (M.B.); (V.D.)
- Correspondence: ; Tel.: +39-011-670-5636
| |
Collapse
|
12
|
Cardenas H, Jiang G, Thomes Pepin J, Parker JB, Condello S, Nephew KP, Nakshatri H, Chakravarti D, Liu Y, Matei D. Interferon-γ signaling is associated with BRCA1 loss-of-function mutations in high grade serous ovarian cancer. NPJ Precis Oncol 2019; 3:32. [PMID: 31840082 PMCID: PMC6897992 DOI: 10.1038/s41698-019-0103-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/29/2019] [Indexed: 12/17/2022] Open
Abstract
Loss-of-function mutations of the breast cancer type 1 susceptibility protein (BRCA1) are associated with breast (BC) and ovarian cancer (OC). To identify gene signatures regulated by epigenetic mechanisms in OC cells carrying BRCA1 mutations, we assessed cellular responses to epigenome modifiers and performed genome-wide RNA- and chromatin immunoprecipitation-sequencing in isogenic OC cells UWB1.289 (carrying a BRCA1 mutation, BRCA1-null) and UWB1.289 transduced with wild-type BRCA1 (BRCA1+). Increased sensitivity to histone deacetylase inhibitors (HDACi) was observed in BRCA1-null vs. BRCA1+ cells. Gene expression profiles of BRCA1-null vs. BRCA1+ cells and treated with HDACi were integrated with chromatin mapping of histone H3 lysine 9 or 27 acetylation. Gene networks activated in BRCA1-null vs. BRCA1 + OC cells related to cellular movement, cellular development, cellular growth and proliferation, and activated upstream regulators included TGFβ1, TNF, and IFN-γ. The IFN-γ pathway was altered by HDACi in BRCA1+ vs. BRCA1-null cells, and in BRCA1-mutated/or low vs. BRCA1-normal OC tumors profiled in the TCGA. Key IFN-γ-induced genes upregulated at baseline in BRCA1-null vs. BRCA1+OC and BC cells included CXCL10, CXCL11, and IFI16. Increased localization of STAT1 in the promoters of these genes occurred in BRCA1-null OC cells, resulting in diminished responses to IFN-γ or to STAT1 knockdown. The IFN-γ signature was associated with improved survival among OC patients profiled in the TCGA. In all, our results support that changes affecting IFN-γ responses are associated with inactivating BRCA1 mutations in OC. This signature may contribute to altered responses to anti-tumor immunity in BRCA1-mutated cells or tumors.
Collapse
Affiliation(s)
- Horacio Cardenas
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
| | - Guanglong Jiang
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN USA
- Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN USA
| | - Jessica Thomes Pepin
- Department of Obstetrics and Gynecology, Indiana University, Indianapolis, IN USA
| | - J. Brandon Parker
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
| | - Salvatore Condello
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
| | - Kenneth P. Nephew
- Department of Obstetrics and Gynecology, Indiana University, Indianapolis, IN USA
- Melvin and Bren Simon Cancer Center, Indianapolis, IN USA
- Medical Sciences, Indiana University School of Medicine, Bloomington, IN USA
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Harikrishna Nakshatri
- Melvin and Bren Simon Cancer Center, Indianapolis, IN USA
- Departments of Surgery, Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN USA
| | - Debabrata Chakravarti
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
- Robert H Lurie Comprehensive Cancer Center, Chicago, IL USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN USA
- Melvin and Bren Simon Cancer Center, Indianapolis, IN USA
| | - Daniela Matei
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL USA
- Robert H Lurie Comprehensive Cancer Center, Chicago, IL USA
- Jesse Brown VA Medical Center, Chicago, IL USA
| |
Collapse
|
13
|
Piccaluga PP, Navari M, Visani A, Rigotti F, Agostinelli C, Righi S, Diani E, Ligozzi M, Carelli M, Ponti C, Bon I, Zipeto D, Landolfo S, Gibellini D. Interferon gamma inducible protein 16 (IFI16) expression is reduced in mantle cell lymphoma. Heliyon 2019; 5:e02643. [PMID: 31840115 PMCID: PMC6893061 DOI: 10.1016/j.heliyon.2019.e02643] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 07/12/2019] [Accepted: 10/09/2019] [Indexed: 11/05/2022] Open
Abstract
IFI16, member of the IFN-inducible PYHIN-200 gene family, modulates proliferation, survival and differentiation of different cell lineages. In particular, IFI16 expression, which is regulated during the differentiation of B cells, was recently studied in B-CLL as well. Here, we compared IFI16 expression in several lymphomas including Burkitt lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, marginal zone lymphoma and mantle cell lymphoma with respect to normal cell counterparts. We observed that IFI16 expression was significantly deregulated only in mantle cell lymphoma (p < 0.05). Notably, IFI16 was associated with the expression of genes involved in interferon response, cell cycle, cell death and proliferation and, interestingly, lipid and glucose metabolism, suggesting that IFI16 deregulation might be associated with relevant changes in cell biology. In our group of mantle cell lymphoma samples a correlation between patient survival and IFI16 expression was not detected even though mantle cell lymphoma prognosis is known to be associated with cell proliferation. Altogether, these results suggest a complex relationship between IFI16 expression and MCL which needs to be analyzed in further studies.
Collapse
Affiliation(s)
- Pier Paolo Piccaluga
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy.,Istituto Euro-Mediterraneo di Scienza e Tecnologia (IEMEST) Palermo, Italy.,Department of Pathology, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Mohsen Navari
- Department of Medical Biotechnology, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Axel Visani
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Flavia Rigotti
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Claudio Agostinelli
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Simona Righi
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Erica Diani
- Department of Diagnostic and Public Health, Unit of Microbiology, University of Verona, Verona, Italy
| | - Marco Ligozzi
- Department of Diagnostic and Public Health, Unit of Microbiology, University of Verona, Verona, Italy
| | - Maria Carelli
- Department of Diagnostic and Public Health, Unit of Microbiology, University of Verona, Verona, Italy
| | - Cristina Ponti
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Isabella Bon
- Department of Experimental, Diagnostic, and Specialty Medicine, Microbiology Unit, University of Bologna, Bologna, Italy
| | - Donato Zipeto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Santo Landolfo
- Department of Public Health and Microbiology, University of Turin, Turin, Italy
| | - Davide Gibellini
- Department of Diagnostic and Public Health, Unit of Microbiology, University of Verona, Verona, Italy
| |
Collapse
|
14
|
Caneparo V, Landolfo S, Gariglio M, De Andrea M. The Absent in Melanoma 2-Like Receptor IFN-Inducible Protein 16 as an Inflammasome Regulator in Systemic Lupus Erythematosus: The Dark Side of Sensing Microbes. Front Immunol 2018; 9:1180. [PMID: 29892303 PMCID: PMC5985366 DOI: 10.3389/fimmu.2018.01180] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/11/2018] [Indexed: 12/22/2022] Open
Abstract
Absent in melanoma 2 (AIM2)-like receptors (ALRs) are a newly characterized class of pathogen recognition receptors (PRRs) involved in cytosolic and nuclear pathogen DNA recognition. In recent years, two ALR family members, the interferon (IFN)-inducible protein 16 (IFI16) and AIM2, have been linked to the pathogenesis of various autoimmune diseases, among which systemic lupus erythematosus (SLE) has recently gained increasing attention. SLE patients are indeed often characterized by constitutively high serum IFN levels and increased expression of IFN-stimulated genes due to an abnormal response to pathogens and/or incorrect self-DNA recognition process. Consistently, we and others have shown that IFI16 is overexpressed in a wide range of autoimmune diseases where it triggers production of specific autoantibodies. In addition, evidence from mouse models supports a model whereby ALRs are required for IFN-mediated host response to both exogenous and endogenous DNA. Following interaction with cytoplasmic or nuclear nucleic acids, ALRs can form a functional inflammasome through association with the adaptor ASC [apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD)] and with procaspase-1. Importantly, inflammasome-mediated upregulation of IL-1β and IL-18 production positively correlates with SLE disease severity. Therefore, targeting ALR sensors and their downstream pathways represents a promising alternative therapeutic approach for SLE and other systemic autoimmune diseases.
Collapse
Affiliation(s)
- Valeria Caneparo
- Viral Pathogenesis Unit, Department of Public Health and Pediatric Sciences, Turin Medical School, Turin, Italy.,Virology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Department of Translational Medicine, Novara Medical School, Novara, Italy.,Intrinsic Immunity Unit, CAAD - Center for Translational Research on Autoimmune and Allergic Disease, University of Piemonte Orientale, Novara, Italy
| | - Santo Landolfo
- Viral Pathogenesis Unit, Department of Public Health and Pediatric Sciences, Turin Medical School, Turin, Italy
| | - Marisa Gariglio
- Virology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Department of Translational Medicine, Novara Medical School, Novara, Italy.,Intrinsic Immunity Unit, CAAD - Center for Translational Research on Autoimmune and Allergic Disease, University of Piemonte Orientale, Novara, Italy
| | - Marco De Andrea
- Viral Pathogenesis Unit, Department of Public Health and Pediatric Sciences, Turin Medical School, Turin, Italy.,Virology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Department of Translational Medicine, Novara Medical School, Novara, Italy.,Intrinsic Immunity Unit, CAAD - Center for Translational Research on Autoimmune and Allergic Disease, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
15
|
Pushkarev VM, Guda BB, Pushkarev VV, Tronko ND. Oncogene toxicity in thyroid carcinomas and other types of tumors. CYTOL GENET+ 2018. [DOI: 10.3103/s0095452718010103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
16
|
Hao B, Xiao Y, Song F, Long X, Huang J, Tian M, Deng S, Wu Q. Metformin-induced activation of AMPK inhibits the proliferation and migration of human aortic smooth muscle cells through upregulation of p53 and IFI16. Int J Mol Med 2017; 41:1365-1376. [PMID: 29286156 PMCID: PMC5819901 DOI: 10.3892/ijmm.2017.3346] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/07/2017] [Indexed: 11/28/2022] Open
Abstract
The proliferation and migration of vascular smooth muscle cells are significant in the development and progression of atherosclerosis and plaque rupture. Metformin is a widely used antidiabetic drug, which has been reported to inhibit cell growth and migration. The antiproliferative and antimigratory effects of metformin have been attributed to 5′ adenosine monophosphate-activated protein kinase (AMPK) activation. The purpose of the present study was to investigate the effects of metformin on primary human aortic muscle cells (HASMCs) in vitro and to clarify the underlying mechanism. We investigated the effectiveness of metformin in inhibiting the proliferation and migration of HASMCs in vitro using RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR), cell number counting, cell viability assay, cell cycle assay and cell migration assay. Through transfection with small interfering (si)RNA targeting p53 and interferon-inducible protein 16 (IFI16), the roles of p53 and IFI16 in these processes were evaluated. The present study demonstrated that p53, IFI16 and AMPK were upregulated in senescent primary HASMCs, which exhibited a decrease in proliferation and migration. In addition, metformin was able to activate p53, IFI16 and AMPK, in order to inhibit proliferation and migration of HASMCs. Furthermore, siRNA-mediated knockdown of p53 and IFI16 attenuated AMPK activation and reversed the suppressive effects of metformin. Notably, in response to metformin, the activation of AMPK was not observed in p53- and IFI16-silenced HASMCs. These results indicated that metformin-induced activation of AMPK suppresses the proliferation and migration of HASMCs by upregulating p53 and IFI16. These findings suggested that metformin may have potential use in the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Biao Hao
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Yan Xiao
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Fang Song
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Xiangshu Long
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Jing Huang
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Maobo Tian
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Shiyan Deng
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| | - Qiang Wu
- Department of Cardiology, The Affiliated People's Hospital of Guizhou Medical University, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
17
|
Landolfo S, De Andrea M, Dell’Oste V, Gugliesi F. Intrinsic host restriction factors of human cytomegalovirus replication and mechanisms of viral escape. World J Virol 2016; 5:87-96. [PMID: 27563536 PMCID: PMC4981826 DOI: 10.5501/wjv.v5.i3.87] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/03/2016] [Accepted: 07/13/2016] [Indexed: 02/05/2023] Open
Abstract
Before a pathogen even enters a cell, intrinsic immune defenses are active. This first-line defense is mediated by a variety of constitutively expressed cell proteins collectively termed “restriction factors” (RFs), and they form a vital element of the immune response to virus infections. Over time, however, viruses have evolved in a variety ways so that they are able to overcome these RF defenses via mechanisms that are specific for each virus. This review provides a summary of the universal characteristics of RFs, and goes on to focus on the strategies employed by some of the most important RFs in their attempt to control human cytomegalovirus (HCMV) infection. This is followed by a discussion of the counter-restriction mechanisms evolved by viruses to circumvent the host cell’s intrinsic immune defenses. RFs include nuclear proteins IFN-γ inducible protein 16 (IFI16) (a Pyrin/HIN domain protein), Sp100, promyelocytic leukemia, and hDaxx; the latter three being the keys elements of nuclear domain 10 (ND10). IFI16 inhibits the synthesis of virus DNA by down-regulating UL54 transcription - a gene encoding a CMV DNA polymerase; in response, the virus antagonizes IFI16 via a process involving viral proteins UL97 and pp65 (pUL83), which results in the mislocalizing of IFI16 into the cytoplasm. In contrast, viral regulatory proteins, including pp71 and IE1, seek to modify or disrupt the ND10 proteins and thus block or reverse their inhibitory effects upon virus replication. All in all, detailed knowledge of these HCMV counter-restriction mechanisms will be fundamental for the future development of new strategies for combating HCMV infection and for identifying novel therapeutic agents.
Collapse
|
18
|
Harwardt T, Lukas S, Zenger M, Reitberger T, Danzer D, Übner T, Munday DC, Nevels M, Paulus C. Human Cytomegalovirus Immediate-Early 1 Protein Rewires Upstream STAT3 to Downstream STAT1 Signaling Switching an IL6-Type to an IFNγ-Like Response. PLoS Pathog 2016; 12:e1005748. [PMID: 27387064 PMCID: PMC4936752 DOI: 10.1371/journal.ppat.1005748] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/16/2016] [Indexed: 12/24/2022] Open
Abstract
The human cytomegalovirus (hCMV) major immediate-early 1 protein (IE1) is best known for activating transcription to facilitate viral replication. Here we present transcriptome data indicating that IE1 is as significant a repressor as it is an activator of host gene expression. Human cells induced to express IE1 exhibit global repression of IL6- and oncostatin M-responsive STAT3 target genes. This repression is followed by STAT1 phosphorylation and activation of STAT1 target genes normally induced by IFNγ. The observed repression and subsequent activation are both mediated through the same region (amino acids 410 to 445) in the C-terminal domain of IE1, and this region serves as a binding site for STAT3. Depletion of STAT3 phenocopies the STAT1-dependent IFNγ-like response to IE1. In contrast, depletion of the IL6 receptor (IL6ST) or the STAT kinase JAK1 prevents this response. Accordingly, treatment with IL6 leads to prolonged STAT1 instead of STAT3 activation in wild-type IE1 expressing cells, but not in cells expressing a mutant protein (IE1dl410-420) deficient for STAT3 binding. A very similar STAT1-directed response to IL6 is also present in cells infected with a wild-type or revertant hCMV, but not an IE1dl410-420 mutant virus, and this response results in restricted viral replication. We conclude that IE1 is sufficient and necessary to rewire upstream IL6-type to downstream IFNγ-like signaling, two pathways linked to opposing actions, resulting in repressed STAT3- and activated STAT1-responsive genes. These findings relate transcriptional repressor and activator functions of IE1 and suggest unexpected outcomes relevant to viral pathogenesis in response to cytokines or growth factors that signal through the IL6ST-JAK1-STAT3 axis in hCMV-infected cells. Our results also reveal that IE1, a protein considered to be a key activator of the hCMV productive cycle, has an unanticipated role in tempering viral replication. Our previous work has shown that the human cytomegalovirus (hCMV) major immediate-early 1 protein (IE1) modulates host cell signaling pathways involving proteins of the signal transducer and activator of transcription (STAT) family. IE1 has also long been known to facilitate viral replication by activating transcription. In this report we demonstrate that IE1 is as significant a repressor as it is an activator of host gene expression. Many genes repressed by IE1 are normally induced via STAT3 signaling triggered by interleukin 6 (IL6) or related cytokines, whereas many genes activated by IE1 are normally induced via STAT1 signaling triggered by interferon gamma (IFNγ). Our results suggest that the repression of STAT3- and the activation of STAT1-responsive genes by IE1 are coupled. By targeting STAT3, IE1 rewires upstream STAT3 to downstream STAT1 signaling. Consequently, genes normally induced by IL6 are repressed while genes normally induced by IFNγ become responsive to IL6 in the presence of IE1. We also demonstrate that, by switching an IL6 to an IFNγ-like response, IE1 tempers viral replication. These results suggest an unanticipated dual role for IE1 in either promoting or limiting hCMV propagation and demonstrate how a key viral regulatory protein merges two central cellular signaling pathways to divert cytokine responses relevant to hCMV pathogenesis.
Collapse
Affiliation(s)
- Thomas Harwardt
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Simone Lukas
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Marion Zenger
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Tobias Reitberger
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Daniela Danzer
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Theresa Übner
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Diane C. Munday
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
| | - Michael Nevels
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
- * E-mail: (MN); (CP)
| | - Christina Paulus
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
- Biomedical Sciences Research Complex, University of St Andrews, St Andrews, United Kingdom
- * E-mail: (MN); (CP)
| |
Collapse
|
19
|
Ertosun MG, Hapil FZ, Osman Nidai O. E2F1 transcription factor and its impact on growth factor and cytokine signaling. Cytokine Growth Factor Rev 2016; 31:17-25. [PMID: 26947516 DOI: 10.1016/j.cytogfr.2016.02.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 02/27/2016] [Indexed: 12/13/2022]
Abstract
E2F1 is a transcription factor involved in cell cycle regulation and apoptosis. The transactivation capacity of E2F1 is regulated by pRb. In its hypophosphorylated form, pRb binds and inactivates DNA binding and transactivating functions of E2F1. The growth factor stimulation of cells leads to activation of CDKs (cyclin dependent kinases), which in turn phosphorylate Rb and hyperphosphorylated Rb is released from E2F1 or E2F1/DP complex, and free E2F1 can induce transcription of several genes involved in cell cycle entry, induction or inhibition of apoptosis. Thus, growth factors and cytokines generally utilize E2F1 to direct cells to either fate. Furthermore, E2F1 regulates expressions of various cytokines and growth factor receptors, establishing positive or negative feedback mechanisms. This review focuses on the relationship between E2F1 transcription factor and cytokines (IL-1, IL-2, IL-3, IL-6, TGF-beta, G-CSF, LIF), growth factors (EGF, KGF, VEGF, IGF, FGF, PDGF, HGF, NGF), and interferons (IFN-α, IFN-β and IFN-γ).
Collapse
Affiliation(s)
- Mustafa Gokhan Ertosun
- Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetic, Kampus, Antalya 07070, Turkey
| | - Fatma Zehra Hapil
- Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetic, Kampus, Antalya 07070, Turkey
| | - Ozes Osman Nidai
- Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetic, Kampus, Antalya 07070, Turkey.
| |
Collapse
|
20
|
IFI16 Expression Is Related to Selected Transcription Factors during B-Cell Differentiation. J Immunol Res 2015; 2015:747645. [PMID: 26185770 PMCID: PMC4491573 DOI: 10.1155/2015/747645] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/27/2015] [Accepted: 05/14/2015] [Indexed: 01/21/2023] Open
Abstract
The interferon-inducible DNA sensor IFI16 is involved in the modulation of cellular survival, proliferation, and differentiation. In the hematopoietic system, IFI16 is consistently expressed in the CD34+ stem cells and in peripheral blood lymphocytes; however, little is known regarding its regulation during maturation of B- and T-cells. We explored the role of IFI16 in normal B-cell subsets by analysing its expression and relationship with the major transcription factors involved in germinal center (GC) development and plasma-cell (PC) maturation. IFI16 mRNA was differentially expressed in B-cell subsets with significant decrease in IFI16 mRNA in GC and PCs with respect to naïve and memory subsets. IFI16 mRNA expression is inversely correlated with a few master regulators of B-cell differentiation such as BCL6, XBP1, POU2AF1, and BLIMP1. In contrast, IFI16 expression positively correlated with STAT3, REL, SPIB, RELA, RELB, IRF4, STAT5B, and STAT5A. ARACNE algorithm indicated a direct regulation of IFI16 by BCL6, STAT5B, and RELB, whereas the relationship between IFI16 and the other factors is modulated by intermediate factors. In addition, analysis of the CD40 signaling pathway showed that IFI16 gene expression directly correlated with NF-κB activation, indicating that IFI16 could be considered an upstream modulator of NF-κB in human B-cells.
Collapse
|
21
|
Plasmid-based Stat3-specific siRNA and GRIM-19 inhibit the growth of thyroid cancer cells in vitro and in vivo. Oncol Rep 2014; 32:573-80. [DOI: 10.3892/or.2014.3233] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/22/2014] [Indexed: 11/05/2022] Open
|
22
|
Abstract
The Raf/MEK/extracellular signal-regulated kinase (ERK) pathway has a pivotal role in facilitating cell proliferation, and its deregulated activation is a central signature of many epithelial cancers. However paradoxically, sustained activity of Raf/MEK/ERK can also result in growth arrest in many different cell types. This anti-proliferative Raf/MEK/ERK signaling also has physiological significance, as exemplified by its potential as a tumor suppressive mechanism. Therefore, significant questions include in which cell types and by what mechanisms this pathway can mediate such an opposing context of signaling. Particularly, our understating of the role of ERK1 and ERK2, the focal points of pathway signaling, in growth arrest signaling is still limited. This review discusses these aspects of Raf/MEK/ERK-mediated growth arrest signaling.
Collapse
|
23
|
Sosonkina N, Starenki D, Park JI. The Role of STAT3 in Thyroid Cancer. Cancers (Basel) 2014; 6:526-44. [PMID: 24662939 PMCID: PMC3980610 DOI: 10.3390/cancers6010526] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/15/2014] [Accepted: 02/27/2014] [Indexed: 12/16/2022] Open
Abstract
Thyroid cancer is the most common endocrine malignancy and its global incidence rates are rapidly increasing. Although the mortality of thyroid cancer is relatively low, its rate of recurrence or persistence is relatively high, contributing to incurability and morbidity of the disease. Thyroid cancer is mainly treated by surgery and radioiodine remnant ablation, which is effective only for non-metastasized primary tumors. Therefore, better understanding of the molecular targets available in this tumor is necessary. Similarly to many other tumor types, oncogenic molecular alterations in thyroid epithelium include aberrant signal transduction of the mitogen-activated protein kinase, phosphatidylinositol 3-kinase/AKT (also known as protein kinase B), NF-кB, and WNT/β-catenin pathways. However, the role of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT3) pathway, a well-known mediator of tumorigenesis in different tumor types, is relatively less understood in thyroid cancer. Intriguingly, recent studies have demonstrated that, in thyroid cancer, the JAK/STAT3 pathway may function in the context of tumor suppression rather than promoting tumorigenesis. In this review, we provide an update of STAT3 function in thyroid cancer and discuss some of the evidences that support this hypothesis.
Collapse
Affiliation(s)
- Nadiya Sosonkina
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| | - Dmytro Starenki
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
24
|
Unterholzner L. The interferon response to intracellular DNA: why so many receptors? Immunobiology 2013; 218:1312-21. [PMID: 23962476 DOI: 10.1016/j.imbio.2013.07.007] [Citation(s) in RCA: 192] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/11/2013] [Accepted: 07/17/2013] [Indexed: 12/22/2022]
Abstract
The detection of intracellular DNA has emerged to be a key event in the innate immune response to viruses and intracellular bacteria, and during conditions of sterile inflammation and autoimmunity. One of the consequences of the detection of DNA as a 'stranger' and a 'danger' signal is the production of type I interferons and pro-inflammatory cytokines. Much work has been dedicated to the elucidation of the signalling cascades that activate this DNA-induced gene expression programme. However, while many proteins have been proposed to act as sensors for intracellular DNA in recent years, none has been met with universal acceptance, and a theory linking all the recent observations is, as yet, lacking. This review presents the evidence for the various interferon-inducing DNA receptors proposed to date, and examines the hypotheses that might explain why so many different receptors appear to be involved in the innate immune recognition of intracellular DNA.
Collapse
Affiliation(s)
- Leonie Unterholzner
- Division of Cell Signalling and Immunology, College of Life Sciences, University of Dundee, DD1 5EH, UK.
| |
Collapse
|
25
|
Starenki D, Singh NK, Jensen DR, Peterson FC, Park JI. Recombinant leukemia inhibitory factor suppresses human medullary thyroid carcinoma cell line xenografts in mice. Cancer Lett 2013; 339:144-51. [PMID: 23856028 DOI: 10.1016/j.canlet.2013.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/02/2013] [Accepted: 07/08/2013] [Indexed: 01/10/2023]
Abstract
Medullary thyroid carcinoma (MTC) is a neoplasm of the endocrine system, which originates from parafollicular C-cells of the thyroid gland. For MTC therapy, the Food and Drug Administration recently approved vandetanib and cabozantinib, multi-kinase inhibitors targeting RET and other tyrosine kinase receptors of vascular endothelial growth factor, epidermal growth factor, or hepatocyte growth factor. Nevertheless, not all patients with the progressive MTC respond to these drugs, requiring the development of additional therapeutic modalities that have distinct activity. Previously, we reported that expression of activated Ras or Raf in the human MTC cell lines, TT and MZ-CRC-1, can induce growth arrest and RET downregulation via a leukemia inhibitory factor (LIF)-mediated autocrine/paracrine loop. In this study, we aimed to evaluate bacterially-produced recombinant human LIF for its efficacy to suppress human MTC xenografts in mice. Here, we report that, consistent with its effects in vitro, locally or systemically administered recombinant LIF effectively suppressed growth of TT and MZ-CRC-1 xenografts in mice. Further, as predicted from its effects in TT and MZ-CRC-1 cell cultures in vitro, recombinant LIF activated the JAK/STAT pathway and downregulated RET and E2F1 expression in tumors in mice. These results suggest that LIF is a potent cytostatic agent for MTC cells, which regulates unique mechanisms that are not targeted by currently available therapeutic agents.
Collapse
Affiliation(s)
- Dmytro Starenki
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | |
Collapse
|
26
|
Abstract
Although tyrosine-phosphorylated or activated STAT3 (pY-STAT3) is a well-described mediator of tumorigenesis, its role in thyroid cancer has not been investigated. We observed that 63 of 110 (57%) human primary papillary thyroid carcinoma (PTC) cases expressed nuclear pY-STAT3 in tumor cells, preferentially in association with the tumor stroma. An inverse relationship between pY-STAT3 expression with tumor size and the presence of distant metastases was observed. Using human thyroid cancer-derived cell lines [harboring rearranged during transfection (RET)/PTC, v-RAF murine sarcoma viral oncogene homolog B (BRAF), or rat sarcoma virus oncogene (RAS) alterations], we determined that IL-6/gp130/JAK signaling is responsible for STAT3 activation. STAT3 knockdown by shRNA in representative thyroid cancer cell lines that express high levels of pY-STAT3 had no effect on in vitro growth. However, xenografted short hairpin STAT3 cells generated larger tumors than control cells. Similarly, STAT3 deficiency in a murine model of BRAFV600E-induced PTC led to thyroid tumors that were more proliferative and larger than those tumors expressing STAT3wt. Genome expression analysis revealed that STAT3 knockdown resulted in the down-regulation of multiple transcripts, including the tumor suppressor insulin-like growth factor binding protein 7. Furthermore, STAT3 knockdown led to an increase in glucose consumption, lactate production, and expression of Hypoxia-inducible factor 1 (HIF1α) target genes, suggesting that STAT3 is a negative regulator of aerobic glycolysis. Our studies show that, in the context of thyroid cancer, STAT3 is paradoxically a negative regulator of tumor growth. These findings suggest that targeting STAT3 in these cancers could enhance tumor size and highlight the complexities of the role of STAT3 in tumorigenesis.
Collapse
|
27
|
Zhao Y, Zhang J, Xia H, Zhang B, Jiang T, Wang J, Chen X, Wang Y. Stat3 is involved in the motility, metastasis and prognosis in lingual squamous cell carcinoma. Cell Biochem Funct 2012; 30:340-6. [PMID: 22302289 DOI: 10.1002/cbf.2810] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 01/19/2012] [Accepted: 01/20/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Yan Zhao
- Key Laboratory for Oral Biomedical Engineering of the Ministry of Education, School & Hospital of Stomatology; Wuhan University; Wuhan; China
| | - Jiali Zhang
- Department of Pathology, School & Hospital of Stomatology; Wuhan University; Wuhan; China
| | - Haibin Xia
- Key Laboratory for Oral Biomedical Engineering of the Ministry of Education, School & Hospital of Stomatology; Wuhan University; Wuhan; China
| | - Bi Zhang
- Key Laboratory for Oral Biomedical Engineering of the Ministry of Education, School & Hospital of Stomatology; Wuhan University; Wuhan; China
| | - Tao Jiang
- Key Laboratory for Oral Biomedical Engineering of the Ministry of Education, School & Hospital of Stomatology; Wuhan University; Wuhan; China
| | - Jiawei Wang
- Key Laboratory for Oral Biomedical Engineering of the Ministry of Education, School & Hospital of Stomatology; Wuhan University; Wuhan; China
| | - Xinmin Chen
- Department of Pathology, School & Hospital of Stomatology; Wuhan University; Wuhan; China
| | - Yining Wang
- Key Laboratory for Oral Biomedical Engineering of the Ministry of Education, School & Hospital of Stomatology; Wuhan University; Wuhan; China
| |
Collapse
|
28
|
Ong PS, Chan SY, Ho PC. Microarray analysis revealed dysregulation of multiple genes associated with chemoresistance to As(2)O(3) and increased tumor aggressiveness in a newly established arsenic-resistant ovarian cancer cell line, OVCAR-3/AsR. Eur J Pharm Sci 2011; 45:367-78. [PMID: 22178533 DOI: 10.1016/j.ejps.2011.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 11/30/2011] [Accepted: 12/03/2011] [Indexed: 01/07/2023]
Abstract
The potential of arsenic trioxide (As(2)O(3)) for use as a novel therapy for ovarian cancer treatment has been increasingly recognized. In this study, we developed an arsenic-resistant OVCAR-3 subline (OVCAR-3/AsR) and aimed to identify the molecular mechanisms and signaling pathways contributing to the development of acquired arsenic chemoresistance in ovarian cancer. OVCAR-3/AsR cells were obtained following continual exposure of parental OVCAR-3 cells to low dose As(2)O(3) for 12months. Cytotoxicity of OVCAR-3/AsR cells to As(2)O(3), paclitaxel and cisplatin was investigated. Cell apoptosis and cell cycle distribution following As(2)O(3) treatment of OVCAR-3/AsR cells was also analyzed using flow cytometry. Subsequently, cDNA microarray analysis was performed from the RNA samples of OVCAR-3 and OVCAR-3/AsR cells in duplicate experiments. Microarray data were analyzed using Genespring® and Pathway Studio® Softwares. OVCAR-3/AsR cells showed 9-fold greater resistance to As(2)O(3) and lack of collateral resistance to cisplatin and paclitaxel. Compared with parental OVCAR-3 cells, OVCAR-3/AsR had significantly lower apoptotic rates following As(2)O(3) treatment. These cells were also arrested at both the S phase and G(2)/M phase of the cell cycle after exposure to high concentrations of As(2)O(3). Gene expression profiling revealed significant differences in expression levels of 397 genes between OVCAR-3/AsR and OVCAR-3 cells. The differentially regulated transcripts genes have functional ontologies related to continued cancer cell growth, cell survival, tumor metastasis and tumor aggressiveness. Additionally, numerous gene targets of the nuclear factor erythroid 2-related factor 2 (NRF2) transcription factor showed elevated expression in OVCAR-3/AsR cells. Subsequent pathway analysis further revealed a gene network involving interleukin 1-alpha (IL1A) in mediating the arsenic-resistant phenotype. These results showed that changes in multiple genes and an increased in tumor aggressiveness occurred during the development of acquired chemoresistance to As(2)O(3) in ovarian cancer cells. The functional relevance of these genetic changes should be validated in future studies.
Collapse
Affiliation(s)
- Pei-Shi Ong
- Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117 543, Singapore.
| | | | | |
Collapse
|
29
|
Dombrowski Y, Peric M, Koglin S, Kammerbauer C, Göss C, Anz D, Simanski M, Gläser R, Harder J, Hornung V, Gallo RL, Ruzicka T, Besch R, Schauber J. Cytosolic DNA triggers inflammasome activation in keratinocytes in psoriatic lesions. Sci Transl Med 2011; 3:82ra38. [PMID: 21562230 DOI: 10.1126/scitranslmed.3002001] [Citation(s) in RCA: 319] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The proinflammatory cytokine interleukin-1β (IL-1β) plays a central role in the pathogenesis and the course of inflammatory skin diseases, including psoriasis. Posttranscriptional activation of IL-1β is mediated by inflammasomes; however, the mechanisms triggering IL-1β processing remain unknown. Recently, cytosolic DNA has been identified as a danger signal that activates inflammasomes containing the DNA sensor AIM2. In this study, we detected abundant cytosolic DNA and increased AIM2 expression in keratinocytes in psoriatic lesions but not in healthy skin. In cultured keratinocytes, interferon-γ induced AIM2, and cytosolic DNA triggered the release of IL-1β via the AIM2 inflammasome. Moreover, the antimicrobial cathelicidin peptide LL-37, which can interact with DNA in psoriatic skin, neutralized cytosolic DNA in keratinocytes and blocked AIM2 inflammasome activation. Together, these data suggest that cytosolic DNA is an important disease-associated molecular pattern that can trigger AIM2 inflammasome and IL-1β activation in psoriasis. Furthermore, cathelicidin LL-37 interfered with DNA-sensing inflammasomes, which thereby suggests an anti-inflammatory function for this peptide. Thus, our data reveal a link between the AIM2 inflammasome, cathelicidin LL-37, and autoinflammation in psoriasis, providing new potential targets for the treatment of this chronic skin disease.
Collapse
Affiliation(s)
- Yvonne Dombrowski
- Department of Dermatology and Allergy, Ludwig-Maximilian-University, 80337 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Minisini R, Giarda P, Grossi G, Bitetto D, Toniutto P, Falleti E, Avellini C, Occhino G, Fabris C, Pirisi M. Early activation of interferon-stimulated genes in human liver allografts: relationship with acute rejection and histological outcome. J Gastroenterol 2011; 46:1307-15. [PMID: 21789480 DOI: 10.1007/s00535-011-0440-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Accepted: 06/19/2011] [Indexed: 02/04/2023]
Abstract
BACKGROUND Innate immunity mechanisms have been shown to play a paramount role in organ transplantation. Our aim was to investigate the hypothesis that activation of the interferon system may affect clinically relevant outcomes, such as acute rejection and/or early fibrosis progression, after liver transplantation. METHODS We studied 71 consecutive recipients (57 males; 25 with hepatitis C) who underwent two per protocol graft biopsies: the first, within 60 days after the transplant operation (median 24) and the second, after 1 year. The mRNA expression for five interferon-stimulated genes (Mx1, OAS2, PKR, IRF7A, IFI16) was measured on the first biopsy specimens. The main outcome measures were acute rejection during the first post-transplant year and fibrosis progression at the second biopsy. RESULTS On multivariate analysis, the independent predictors of gene expression were hepatitis C (Mx1, OAS2, PKR and IFI16), donor age (IFI16) and recipient gender (IRF7A) (P < .05 for all). During the first post-transplant year, 19/71 patients (27%) had acute cellular rejection. At multivariate analysis, acute cellular rejection was independently predicted by high IRF7A mRNA expression. At the end of follow-up, 25 patients had some degree of fibrosis (F2 or higher in seven cases). On multivariate analysis, hepatitis C etiology, recipient age, and OAS2 overexpression were independent predictors of early fibrosis progression. CONCLUSIONS In the early postoperative period of liver transplantation, interferon-stimulated gene activation is dependent on hepatitis C recurrence (the main factor responsible for early fibrosis progression) and donor age, and is related to the risk of acute cellular rejection.
Collapse
Affiliation(s)
- Rosalba Minisini
- Department of Clinical and Experimental Medicine, Università del Piemonte Orientale A. Avogadro, Via G. Solaroli 17, 28100 Novara, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hong SK, Kim JH, Lin MF, Park JI. The Raf/MEK/extracellular signal-regulated kinase 1/2 pathway can mediate growth inhibitory and differentiation signaling via androgen receptor downregulation in prostate cancer cells. Exp Cell Res 2011; 317:2671-82. [PMID: 21871886 DOI: 10.1016/j.yexcr.2011.08.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 08/08/2011] [Accepted: 08/09/2011] [Indexed: 12/18/2022]
Abstract
Upregulated ERK1/2 activity is correlated with androgen receptor (AR) downregulation in certain prostate cancer (PCa) that exhibits androgen deprivation-induced neuroendocrine differentiation, but its functional relevance requires elucidation. We found that sustained ERK1/2 activation using active Raf or MEK1/2 mutants is sufficient to induce AR downregulation at mRNA and protein levels in LNCaP. Downregulation of AR protein, but not mRNA, was blocked by proteasome inhibitors, MG132 and bortezomib, indicating that the pathway regulation is mediated at multiple points. Ectopic expression of a constitutively active AR inhibited Raf/MEK/ERK-mediated regulation of the differentiation markers, neuron-specific enolase and neutral endopeptidase, and the cyclin-dependent kinase inhibitors, p16(INK4A) and p21(CIP1), but not Rb phosphorylation and E2F1 expression, indicating that AR has a specific role in the pathway-mediated differentiation and growth inhibitory signaling. However, despite the sufficient role of Raf/MEK/ERK, its inhibition using U0126 or ERK1/2 knockdown could not block androgen deprivation-induced AR downregulation in an LNCaP neuroendocrine differentiation model, suggesting that additional signaling pathways are involved in the regulation. We additionally report that sustained Raf/MEK/ERK activity can downregulate full length as well as hormone binding domain-deficient AR isoforms in androgen-refractory C4-2 and CWR22Rv1, but not in LAPC4 and MDA-PCa-2b. Our study demonstrates a novel role of the Raf/MEK/ERK pathway in regulating AR expression in certain PCa types and provides an insight into PCa responses to its aberrant activation.
Collapse
Affiliation(s)
- Seung-Keun Hong
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | |
Collapse
|
32
|
Gariglio M, Mondini M, De Andrea M, Landolfo S. The multifaceted interferon-inducible p200 family proteins: from cell biology to human pathology. J Interferon Cytokine Res 2011; 31:159-72. [PMID: 21198352 DOI: 10.1089/jir.2010.0106] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The interferon-inducible p200 family proteins consist of a group of homologous human and mouse proteins that have an N-terminal Pyrin domain and 1 or 2 partially conserved 200 amino acid long C-terminal domains (designated the HIN domain or p200 X domain). These proteins display multifaceted activity due to their ability to bind to various target proteins (eg, transcription factors, signaling proteins, and tumor suppressor proteins) and modulate different cell functions. In addition to a role in interferon biology, increasing evidence supports a role for these proteins as regulators of various cell functions, including proliferation, differentiation, apoptosis, senescence, inflammasome assembly, and control of organ transplants. As a consequence, alterations in their expression and function may be of relevance in the pathogenesis of human diseases, such as systemic autoimmune syndromes, tumors, and degenerative diseases. This review summarizes the literature describing these data, highlights some of the important findings derived from recent studies, and speculates about future perspectives.
Collapse
Affiliation(s)
- Marisa Gariglio
- Department of Clinical and Experimental Medicine, Medical School of Novara, Novara, Italy
| | | | | | | |
Collapse
|
33
|
Pawlik A, Alibert O, Baulande S, Vaigot P, Tronik-Le Roux D. Transcriptome characterization uncovers the molecular response of hematopoietic cells to ionizing radiation. Radiat Res 2011; 175:66-82. [PMID: 21175349 DOI: 10.1667/rr2282.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ionizing radiation causes rapid and acute suppression of hematopoietic cells that manifests as the hematopoietic syndrome. However, the roles of molecules and regulatory pathways induced in vivo by irradiation of different hematopoietic cells have not been completely elaborated. Using a strategy that combined different microarray bioinformatics tools, we identified gene networks that might be involved in the early response of hematopoietic cells radiation response in vivo. The grouping of similar time-ordered gene expression profiles using quality threshold clustering enabled the successful identification of common binding sites for 56 transcription factors that may be involved in the regulation of the early radiation response. We also identified novel genes that are responsive to the transformation-related protein 53; all of these genes were biologically validated in p53-transgenic null mice. Extension of the analysis to purified bone marrow cells including highly purified long-term hematopoietic stem cells, combined with functional classification, provided evidence of gene expression modifications that were largely unknown in this primitive population. Our methodology proved particularly useful for analyzing the transcriptional regulation of the complex ionizing radiation response of hematopoietic cells. Our data may help to elucidate the molecular mechanisms involved in tissue radiosensitivity and to identify potential targets for improving treatment in radiation emergencies.
Collapse
|
34
|
Arthan D, Hong SK, Park JI. Leukemia inhibitory factor can mediate Ras/Raf/MEK/ERK-induced growth inhibitory signaling in medullary thyroid cancer cells. Cancer Lett 2010; 297:31-41. [PMID: 20570039 DOI: 10.1016/j.canlet.2010.04.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Revised: 04/14/2010] [Accepted: 04/27/2010] [Indexed: 11/19/2022]
Abstract
Medullary thyroid carcinoma (MTC) is a multiple endocrine neoplasia type 2 syndrome caused by mutations in extracellular receptor or intracellular kinase domains of the RET proto-oncogene. Activation of the Ras/Raf/MEK/ERK pathway can lead to growth arrest by secreting leukemia inhibitory factor (LIF) in MTC cells harboring a RET receptor domain mutation. Here, we report that Ras/Raf/MEK/ERK can also mediate, via LIF, growth inhibition in MTC cells harboring a RET kinase domain mutation. Ras/Raf/MEK/ERK activation was sufficient to induce growth inhibition and LIF expression in the human MTC line MZ-CRC-1. Presence of LIF-mediated signaling was determined by blocking the activity of culture medium conditioned by Raf-activated cells using anti-LIF neutralizing antibody. In addition, recombinant LIF effectively suppressed cell proliferation via cell cycle arrest in G0/G1 phase. Expression of dominant negative STAT3 abrogated LIF effects, indicating that LIF mediates its signaling through the JAK/STAT3 pathway. These results suggest that growth inhibition and activation of the autocrine/paracrine signaling through LIF/JAK/STAT may be a common response to Ras/Raf activation in different MTC types, and justify further evaluation of LIF as a potential anticancer agent for MTC.
Collapse
Affiliation(s)
- Dumrongkiet Arthan
- Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, 53226, USA
| | | | | |
Collapse
|
35
|
Interferon-inducible IFI16, a negative regulator of cell growth, down-regulates expression of human telomerase reverse transcriptase (hTERT) gene. PLoS One 2010; 5:e8569. [PMID: 20052289 PMCID: PMC2797294 DOI: 10.1371/journal.pone.0008569] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 12/11/2009] [Indexed: 01/07/2023] Open
Abstract
Background Increased levels of interferon (IFN)-inducible IFI16 protein (encoded by the IFI16 gene located at 1q22) in human normal prostate epithelial cells and diploid fibroblasts (HDFs) are associated with the onset of cellular senescence. However, the molecular mechanisms by which the IFI16 protein contributes to cellular senescence-associated cell growth arrest remain to be elucidated. Here, we report that increased levels of IFI16 protein in normal HDFs and in HeLa cells negatively regulate the expression of human telomerase reverse transcriptase (hTERT) gene. Methodology/Principal Findings We optimized conditions for real-time PCR, immunoblotting, and telomere repeat amplification protocol (TRAP) assays to detect relatively low levels of hTERT mRNA, protein, and telomerase activity that are found in HDFs. Using the optimized conditions, we report that treatment of HDFs with inhibitors of cell cycle progression, such as aphidicolin or CGK1026, which resulted in reduced steady-state levels of IFI16 mRNA and protein, was associated with increases in hTERT mRNA and protein levels and telomerase activity. In contrast, knockdown of IFI16 expression in cells increased the expression of c-Myc, a positive regulator of hTERT expression. Additionally, over-expression of IFI16 protein in cells inhibited the c-Myc-mediated stimulation of the activity of hTERT-luc-reporter and reduced the steady-state levels of c-Myc and hTERT. Conclusions/Significance These data demonstrated that increased levels of IFI16 protein in HDFs down-regulate the expression of hTERT gene. Our observations will serve basis to understand how increased cellular levels of the IFI16 protein may contribute to certain aging-dependent diseases.
Collapse
|
36
|
Hong SK, Yoon S, Moelling C, Arthan D, Park JI. Noncatalytic function of ERK1/2 can promote Raf/MEK/ERK-mediated growth arrest signaling. J Biol Chem 2009; 284:33006-18. [PMID: 19805545 PMCID: PMC2785141 DOI: 10.1074/jbc.m109.012591] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2009] [Revised: 09/09/2009] [Indexed: 10/20/2022] Open
Abstract
Kinase activity is known as the key biochemical property of MAPKs. Here, we report that ERK1/2 also utilizes its noncatalytic function to mediate certain signal transductions. Sustained activation of the Raf/MEK/ERK pathway induces growth arrest, accompanied by changes in cell cycle regulators (decreased retinoblastoma phosphorylation, E2F1 down-regulation, and/or p21(CIP1) up-regulation) and cell type-specific changes in morphology and expression of c-Myc or RET in the human tumor lines LNCaP, U251, and TT. Ablation of ERK1/2 by RNA interference abrogated all these effects. However, active site-disabled ERK mutants (ERK1-K71R, ERK2-K52R, and ERK2-D147A), which competitively inhibit activation of endogenous ERK1/2, could not block Raf/MEK-induced growth arrest as well as changes in the cell cycle regulators, although they effectively blocked phosphorylation of the ERK1/2 catalytic activity readouts, p90(RSK) and ELK1, as well as the cell type-specific changes. Because this indicated a potential noncatalytic ERK1/2 function, we generated stable lines of the tumor cells in which both ERK1 and ERK2 were significantly knocked down, and we further investigated the possibility using rat-derived kinase-deficient ERK mutants (ERK2-K52R and ERK2-T183A/Y185F) that were not targeted by human small hairpin RNA. Indeed, ERK2-K52R selectively restored Raf-induced growth inhibitory signaling in ERK1/2-depleted cells, as manifested by regained cellular ability to undergo growth arrest and to control the cell cycle regulators without affecting c-Myc and morphology. However, ERK2-T183A/Y185F was less effective, indicating the requirement of TEY site phosphorylation. Our study suggests that functions of ERK1/2 other than its "canonical" kinase activity are also involved in the pathway-mediated growth arrest signaling.
Collapse
Affiliation(s)
- Seung-Keun Hong
- From the Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Seunghee Yoon
- From the Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Cas Moelling
- From the Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Dumrongkiet Arthan
- From the Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Jong-In Park
- From the Department of Biochemistry, The Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
37
|
Borgogna C, Toniutto P, Smirne C, Azzimonti B, Rittà M, Avellini C, Fabris C, Landolfo S, Gariglio M, Pirisi M. Expression of the interferon-inducible proteins MxA and IFI16 in liver allografts. Histopathology 2009; 54:837-46. [PMID: 19635103 DOI: 10.1111/j.1365-2559.2009.03311.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
AIMS To test the hypothesis that the activation of the interferon (IFN) system pathways might link hepatitis C virus (HCV) recurrence in the liver allograft with acute cellular rejection. METHODS AND RESULTS In this retrospective study, allograft biopsy specimens from 28 adult patients (14 HCV+ and 14 HCV-) who had undergone their first liver transplantation were analysed. Eleven biopsy specimens showed acute cellular rejection (Banff rejection activity index score > or =3). Specimens were immunostained for two IFN-inducible proteins, MxA and IFI16, and for CD45. The predominant MxA reactivity pattern was hepatocytic, whereas IFI16 was expressed in both the hepatocellular and inflammatory compartments. Moderate to strong MxA expression in hepatocytes was associated positively with rejection score (P < 0.01), donor's age < or =45 years (P < 0.05) and aspartate aminotransferase levels >40 U/l on the day of biopsy (P < 0.05), and inversely with infiltration of portal triads by IFI16+/CD45+ cells (P < 0.005) and time to progression beyond Ishak stage 2 of recurrent hepatitis C (P < 0.01). On multivariate analysis, MxA expression in hepatocytes was independently associated with allograft rejection and donor's age. CONCLUSIONS Acute allograft rejection and recurrence of HCV infection in the liver allograft appear to intersect in the IFN system pathways.
Collapse
Affiliation(s)
- Cinzia Borgogna
- DPMSC, Medical Liver Transplantation Unit, University of Udine, Udine, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chadwick N, Fennessy C, Nostro MC, Baron M, Brady G, Buckle AM. Notch induces cell cycle arrest and apoptosis in human erythroleukaemic TF-1 cells. Blood Cells Mol Dis 2008; 41:270-7. [PMID: 18693120 DOI: 10.1016/j.bcmd.2008.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 05/13/2008] [Accepted: 06/17/2008] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Notch signalling is known to promote hematopoietic stem cell self-renewal and to influence the lineage commitment decisions of progenitor cells. The purpose of this study was to investigate the mechanism of Notch-induced apoptosis in the erythroleukaemic cell line TF-1, and in primary cord blood CD34+ cells. METHODS Retroviral constructs containing constitutively active forms of Notch as well as components of the Notch signalling pathway were used to transduce cells and their effect on cell cycle kinetics and apoptosis assayed by immunostaining for the S-phase marker Ki67 and Annexin V. RESULTS We found that TF-1 cells undergo cell cycle arrest followed by apoptosis in a cytokine-independent manner in response to active Notch. Transduction of TF-1 cells with known targets of Notch signalling, Deltex1, HES1 and HERP2, showed that Notch-induced cell cycle arrest was not mediated by these proteins. However, analysis of cell cycle gene expression revealed that Notch signalling was associated with an up-regulation of IFI16 expression in TF-1 cells and in primary cord blood CD34+ cells. CONCLUSION These data demonstrate that, in the context of TF-1 cells, Notch signalling can induce cell cycle arrest and apoptosis.
Collapse
Affiliation(s)
- Nicholas Chadwick
- Faculty of Life Sciences, Manchester Interdisciplinary Biocenter, University of Manchester, Manchester M1 7DN, UK
| | | | | | | | | | | |
Collapse
|
39
|
Ye F, Hu Y, Lu W, Zhou C, Xie X. Expression of leukaemia inhibitory factor in epithelial ovarian carcinoma: correlation with clinical characteristics. Histopathology 2008; 53:224-8. [DOI: 10.1111/j.1365-2559.2008.03068.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
40
|
A pathologic link between Wilms tumor suppressor gene, WT1, and IFI16. Neoplasia 2008; 10:69-78. [PMID: 18231640 DOI: 10.1593/neo.07869] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 11/07/2007] [Accepted: 11/08/2007] [Indexed: 11/18/2022] Open
Abstract
The Wilms tumor gene (WT1) is mutated or deleted in patients with heredofamilial syndromes associated with the development of Wilms tumors, but is infrequently mutated in sporadic Wilms tumors. By comparing the microarray profiles of syndromic versus sporadic Wilms tumors and WT1-inducible Saos-2 osteosarcoma cells, we identified interferon-inducible protein 16 (IFI16), a transcriptional modulator, as a differentially expressed gene and a candidate WT1 target gene. WT1 induction in Saos-2 osteosarcoma cells led to strong induction of IFI16 expression and its promoter activity was responsive to the WT1 protein. Immunohistochemical analysis showed that IFI16 and WT1 colocalized in WT1-replete Wilms tumors, but not in normal human midgestation fetal kidneys, suggesting that the ability of WT1 to regulate IFI16 in tumors represented an aberrant pathologic relationship. In addition, endogenous IFI16 and WT1 interacted in vivo in two Wilms tumor cell lines. Furthermore, IFI16 augmented the transcriptional activity of WT1 on both synthetic and physiological promoters. Strikingly, short hairpin RNA (shRNA)-mediated knockdown of either IFI16 or WT1 led to decreased growth of Wilms tumor cells. These data suggest that IFI16 and WT1, in certain cellular context including sporadic Wilms tumors, may support cell survival.
Collapse
|
41
|
STAT3 signaling is induced by intercellular adhesion in squamous cell carcinoma cells. Exp Cell Res 2007; 314:377-86. [PMID: 17961551 DOI: 10.1016/j.yexcr.2007.09.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Revised: 08/27/2007] [Accepted: 09/22/2007] [Indexed: 10/22/2022]
Abstract
The signal transducer and activator of transcription-3 (STAT3) frequently activated during tumor progression has been linked to enhanced cell growth. In squamous cell carcinoma of the head and neck (HNSCC), STAT3 signaling has been shown to inhibit apoptosis and induce a more aggressive phenotype through the activation of specific signaling pathways. In the present study, we have examined the potential mechanism by which cell-cell contact initiates STAT3 activation. Using a panel of HNSCC cell lines, Ca(+2)-dependent cell-cell adhesion and adherens junction formation in multicellular aggregates triggered phosphorylation of STAT3-Y705 and STAT1-Y701. This intercellular adhesion-induced STAT3 activation was mediated by JAK and Src signaling and partially by EGFR signaling. In addition, immunolocalization studies revealed initial formation of phosphorylated STAT3-Y705 at nascent E-cadherin cell junctions with eventual translocation to the nucleus in cell aggregates. Adhesion-mediated STAT activation in monolayer and cell aggregate cultures required functional E-cadherin. These results indicate that, in HNSCC cells, cadherin-mediated intercellular adhesion induces STAT signaling that may modulate cell survival and resistance to apoptosis during tumor progression.
Collapse
|
42
|
Mondini M, Vidali M, Airò P, De Andrea M, Riboldi P, Meroni PL, Gariglio M, Landolfo S. Role of the Interferon-Inducible Gene IFI16 in the Etiopathogenesis of Systemic Autoimmune Disorders. Ann N Y Acad Sci 2007; 1110:47-56. [PMID: 17911419 DOI: 10.1196/annals.1423.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Interferons (IFNs) are now known to exert a multitude of immunological functions on both the innate and adaptive immunity. Given their pleiotropic effects on the immune system, it is conceivable that excess type I IFN or aberrant regulation of its signaling could contribute to autoimmunity. Several lines of evidence link IFNs to autoimmune disorders, in particular to systemic lupus erythematosus (SLE) and systemic sclerosis (SSc). Expression of a spectrum of genes that constitutes an "IFN signature" is the most significant observation indicating that IFNs may be dominant among the pathogenic mediators involved in some autoimmune diseases. A family of IFN-inducible genes, designated HIN-200 in the human and IFI-200 in the murine species, encodes evolutionary related human (IFI16, MNDA, AIM2, IFIX) and murine proteins (Ifi202 a, Ifi202b, Ifi203, Ifi204, Ifi205/D3). Physiological IFI16 expression was found in cells of the immune system, in endothelial cells, and in stratified squamous epithelia, such as skin. The presence of anti-IFI16 antibodies was reported in SLE and primary/secondary Sjögren's syndrome. More recently, we reported that anti-IFI16 autoantibodies differentiate limited cutaneous systemic sclerosis (lcSSc) from diffuse systemic sclerosis (dcSSc). Molecular studies performed in primary endothelial cells overexpressing IFI16 demonstrated that it may be involved in the early steps of inflammation by modulating endothelial cell function, such as expression of adhesion molecules and chemokine production, cell growth, and apoptosis. Moreover, here we report that IFI16 expression is induced by proinflammatory cytokines. In this article the role of the IFI16 protein and its corresponding autoantibodies in the etiopathogenesis of systemic autoimmune diseases, in which chronic inflammation is involved, are discussed.
Collapse
Affiliation(s)
- Michele Mondini
- Department of Public Health and Microbiology, Medical School, University of Turin, V. Santena 9, 10126, Turin, Italy
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Parent R, Kolippakkam D, Booth G, Beretta L. Mammalian target of rapamycin activation impairs hepatocytic differentiation and targets genes moderating lipid homeostasis and hepatocellular growth. Cancer Res 2007; 67:4337-45. [PMID: 17483347 DOI: 10.1158/0008-5472.can-06-3640] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The mammalian target of rapamycin (mTOR) pathway, a major regulator of translation, is frequently activated in hepatocellular carcinomas. We investigated the effects of mTOR activation in the human HepaRG cells, which possess potent hepatocytic differentiation capability. Differentiation of HepaRG cells into functional and polarized hepatocyte-like cells correlated with a decrease in mTOR and Akt activities. Stable cell lines expressing an activated mutant of mTOR were generated. Sustained activation of mTOR impaired the hepatocytic differentiation capability of these cells as shown by impaired formation of bile canaliculi, absence of polarity, and reduced secretion of alpha1-antitrypsin. An inhibitor of mTOR, rapamycin, was able to revert this phenotype. Furthermore, increased mTOR activity in HepaRG cells resulted in their resistance to the antiproliferative effects of transforming growth factor-beta1. Profiling of polysome-bound transcripts indicated that activated mTOR specifically targeted genes posttranscriptionally regulated on hepatocytic differentiation. Three major biological networks targeted by activated mTOR were identified: (a) cell death associated with tumor necrosis factor superfamily members, IFNs and caspases; (b) lipid homeostasis associated with the transcription factors PPARalpha, PPARdelta, and retinoid X receptor beta; and (c) liver development associated with CCAAT/enhancer binding protein alpha and hepatic mitogens. In conclusion, increased mTOR activity conferred a preneoplastic phenotype to the HepaRG cells by altering the translation of genes vital for establishing normal hepatic energy homeostasis and moderating hepatocellular growth.
Collapse
Affiliation(s)
- Romain Parent
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | | | | | | |
Collapse
|
44
|
Torres A, Storey L, Anders M, Miller RL, Bulbulian BJ, Jin J, Raghavan S, Lee J, Slade HB, Birmachu W. Immune-mediated changes in actinic keratosis following topical treatment with imiquimod 5% cream. J Transl Med 2007; 5:7. [PMID: 17257431 PMCID: PMC1796543 DOI: 10.1186/1479-5876-5-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2006] [Accepted: 01/26/2007] [Indexed: 12/15/2022] Open
Abstract
Background The objective of this study was to identify the molecular processes responsible for the anti-lesional activity of imiquimod in subjects with actinic keratosis using global gene expression profiling. Methods A double-blind, placebo-controlled, randomized study was conducted to evaluate gene expression changes in actinic keratosis treated with imiquimod 5% cream. Male subjects (N = 17) with ≥ 5 actinic keratosis on the scalp applied placebo cream or imiquimod 3 times a week on nonconsecutive days for 4 weeks. To elucidate the molecular processes involved in actinic keratosis lesion regression by imiquimod, gene expression analysis using oligonucleotide arrays and real time reverse transcriptase polymerase chain reaction were performed on shave biopsies of lesions taken before and after treatment. Results Imiquimod modulated the expression of a large number of genes important in both the innate and adaptive immune response, including increased expression of interferon-inducible genes with known antiviral, anti-proliferative and immune modulatory activity, as well as various Toll-like receptors. In addition, imiquimod increased the expression of genes associated with activation of macrophages, dendritic cells, cytotoxic T cells, and natural killer cells, as well as activation of apoptotic pathways. Conclusion Data suggest that topical application of imiquimod stimulates cells in the skin to secrete cytokines and chemokines that lead to inflammatory cell influx into the lesions and subsequent apoptotic and immune cell-mediated destruction of lesions.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Adaptive Immunity/genetics
- Adjuvants, Immunologic/pharmacology
- Administration, Topical
- Aged
- Aged, 80 and over
- Aminoquinolines/administration & dosage
- Aminoquinolines/therapeutic use
- Apoptosis/drug effects
- Apoptosis/genetics
- Cell Proliferation/drug effects
- Chemokines/genetics
- Chemokines/metabolism
- Demography
- Dendritic Cells/drug effects
- Dendritic Cells/metabolism
- Dosage Forms
- Gene Expression Profiling
- Gene Expression Regulation/drug effects
- Humans
- Imiquimod
- Immunity, Innate/drug effects
- Immunity, Innate/genetics
- Interferon Type I/pharmacology
- Keratosis, Actinic/drug therapy
- Keratosis, Actinic/genetics
- Keratosis, Actinic/immunology
- Keratosis, Actinic/pathology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Macrophages/drug effects
- Macrophages/metabolism
- Male
- Middle Aged
- Oligonucleotide Array Sequence Analysis
- Receptors, Pattern Recognition/metabolism
- Reproducibility of Results
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Abel Torres
- Dermatology Office, Loma Linda University Medical Center, Loma Linda, California, USA
| | - Leslie Storey
- Dermatology Office, Loma Linda University Medical Center, Loma Linda, California, USA
| | - Makala Anders
- Dermatology Office, Loma Linda University Medical Center, Loma Linda, California, USA
| | | | | | - Jizhong Jin
- Pharmacology, 3M Pharmaceuticals, St Paul, Minnesota, USA
| | | | - James Lee
- Medical & Scientific Affairs, 3M Pharmaceuticals, St Paul, Minnesota, USA
| | - Herbert B Slade
- Medical & Scientific Affairs, 3M Pharmaceuticals, St Paul, Minnesota, USA
| | | |
Collapse
|
45
|
Rodrigues A. Perspectivas de novos tratamentos para o carcinoma tireoidiano avançado. Rev Col Bras Cir 2006. [DOI: 10.1590/s0100-69912006000300011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|