1
|
Daniels P, Cassoday S, Gupta K, Giurini E, Leifheit ME, Zloza A, Marzo AL. Intratumoral Influenza Vaccine Administration Attenuates Breast Cancer Growth and Restructures the Tumor Microenvironment through Sialic Acid Binding of Vaccine Hemagglutinin. Int J Mol Sci 2023; 25:225. [PMID: 38203396 PMCID: PMC10779129 DOI: 10.3390/ijms25010225] [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: 11/10/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Breast cancer continues to have a high disease burden worldwide and presents an urgent need for novel therapeutic strategies to improve outcomes. The influenza vaccine offers a unique approach to enhance the anti-tumor immune response in patients with breast cancer. Our study explores the intratumoral use of the influenza vaccine in a triple-negative 4T1 mouse model of breast cancer. We show that the influenza vaccine attenuated tumor growth using a three-dose intratumoral regimen. More importantly, prior vaccination did not alter this improved anti-tumor response. Furthermore, we characterized the effect that the influenza vaccine has on the tumor microenvironment and the underlying mechanisms of action. We established that the vaccine facilitated favorable shifts in restructuring the tumor microenvironment. Additionally, we show that the vaccine's ability to bind sialic acid residues, which have been implicated in having oncogenic functions, emerged as a key mechanism of action. Influenza hemagglutinin demonstrated binding ability to breast cancer cells through sialic acid expression. When administered intratumorally, the influenza vaccine offers a promising therapeutic strategy for breast cancer patients by reshaping the tumor microenvironment and modestly suppressing tumor growth. Its interaction with sialic acids has implications for effective therapeutic application and future research.
Collapse
Affiliation(s)
- Preston Daniels
- Department of Internal Medicine, Division of Hematology and Oncology, Rush University Medical Center, Chicago, IL 60612, USA; (P.D.); (M.E.L.); (A.Z.)
| | - Stefanie Cassoday
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Kajal Gupta
- Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA; (K.G.); (E.G.)
| | - Eileena Giurini
- Department of Surgery, Rush University Medical Center, Chicago, IL 60612, USA; (K.G.); (E.G.)
| | - Malia E. Leifheit
- Department of Internal Medicine, Division of Hematology and Oncology, Rush University Medical Center, Chicago, IL 60612, USA; (P.D.); (M.E.L.); (A.Z.)
| | - Andrew Zloza
- Department of Internal Medicine, Division of Hematology and Oncology, Rush University Medical Center, Chicago, IL 60612, USA; (P.D.); (M.E.L.); (A.Z.)
| | - Amanda L. Marzo
- Department of Internal Medicine, Division of Hematology and Oncology, Rush University Medical Center, Chicago, IL 60612, USA; (P.D.); (M.E.L.); (A.Z.)
| |
Collapse
|
2
|
WT1 Inhibits Human Renal Carcinoma Cell Proliferation and Induces G2/M Arrest by Upregulating IL-24 Expression. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1093945. [PMID: 35915803 PMCID: PMC9338855 DOI: 10.1155/2022/1093945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/26/2022] [Accepted: 07/08/2022] [Indexed: 11/17/2022]
Abstract
The transcription factor Wilms’ tumor 1 (WT1) is involved in development, tissue homeostasis, and disease. However, the exact roles and the mechanisms of WT1 in renal carcinoma are not well understood. Therefore, in this study, we evaluated the ability of WT1 to block proliferation in renal carcinoma cells in vitro. Experimental analysis showed that WT1 overexpression inhibited the proliferation of renal carcinoma A498 cells and promoted arrest at the G2/M checkpoint. RNA-Seq identified differentially expressed genes, including IL-24, related to both the cell proliferation and the cell cycle. WT1 overexpression upregulated IL-24 expression, and IL-24 overexpression induced G2/M arrest. ChIP-Seq identified JUN as a direct target of WT1 in A498 cells, in which positive regulation was shown by RT-qPCR. It has been shown that the transcription factor JUN can regulate IL-24 expression, and therefore, we hypothesize that WT1 might regulate the IL-24 through JUN. Furthermore, analysis based on TCGA datasets showed that the expression of WT1-regulated genes, including TXNIP and GADD45A, was significantly correlated with the stage and histological grade of tumors, with high levels linked to favorable prognoses. Our results demonstrated that the overexpression of WT1 upregulates IL-24, leading to G2/M checkpoint arrest to reduce proliferation. These results indicate that regulation of IL-24 by WT1 inhibits proliferation and may represent a potential target for treating renal carcinoma.
Collapse
|
3
|
Parnigoni A, Caon I, Teo WX, Hua SH, Moretto P, Bartolini B, Viola M, Karousou E, Yip GW, Götte M, Heldin P, Passi A, Vigetti D. The natural antisense transcript HAS2-AS1 regulates breast cancer cells aggressiveness independently from hyaluronan metabolism. Matrix Biol 2022; 109:140-161. [PMID: 35395387 DOI: 10.1016/j.matbio.2022.03.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022]
Abstract
Hyaluronan (HA) is a ubiquitous extracellular matrix component playing a crucial role in the regulation of cell behaviors, including cancer. Aggressive breast cancer cells tend to proliferate, migrate and metastatize. Notably, triple-negative breast cancer cells lacking the expression of estrogen receptor (ER) as well as progesterone receptor and HER2 are more aggressive than ER-positive ones. As currently no targeted therapy is available for triple-negative breast cancer, the identification of novel therapeutic targets has a high clinical priority. In ER-negative cells, tumoral behavior can be reduced by inhibiting HA synthesis or silencing the enzymes involved in its metabolism, such as HA synthase 2 (HAS2). HAS2-AS1 is a long non-coding RNA belonging to the natural antisense transcript family which is known to favor HAS2 gene expression and HA synthesis, thus bolstering malignant progression in brain, ovary, and lung tumors. As the role of HAS2-AS1 has not yet been investigated in breast cancer, in this work we report that ER-positive breast cancers had lower HAS2-AS1 expression compared to ER-negative tumors. Moreover, the survival of patients with ER-negative tumors was higher when the expression of HAS2-AS1 was elevated. Experiments with ER-negative cell lines as MDA-MB-231 and Hs 578T revealed that the overexpression of either the full-length HAS2-AS1 or its exon 2 long or short isoforms alone, strongly reduced cell viability, migration, and invasion, whereas HAS2-AS1 silencing increased cell aggressiveness. Unexpectedly, in these ER-negative cell lines, HAS2-AS1 is involved neither in the regulation of HAS2 nor in HA deposition. Finally, transcriptome analysis revealed that HAS2-AS1 modulation affected several pathways, including apoptosis, proliferation, motility, adhesion, epithelial to mesenchymal transition, and signaling, describing this long non-coding RNA as an important regulator of breast cancer cells aggressiveness.
Collapse
Affiliation(s)
- Arianna Parnigoni
- From the Department of Medicine and Surgery - University of Insubria - via J.H. Dunant 5, 21100, Varese, Italy
| | - Ilaria Caon
- From the Department of Medicine and Surgery - University of Insubria - via J.H. Dunant 5, 21100, Varese, Italy
| | - Wei Xuan Teo
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore 4 Medical Drive, Block MD10, Singapore, 117594, Singapore
| | - San Hue Hua
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore 4 Medical Drive, Block MD10, Singapore, 117594, Singapore
| | - Paola Moretto
- From the Department of Medicine and Surgery - University of Insubria - via J.H. Dunant 5, 21100, Varese, Italy
| | - Barbara Bartolini
- From the Department of Medicine and Surgery - University of Insubria - via J.H. Dunant 5, 21100, Varese, Italy
| | - Manuela Viola
- From the Department of Medicine and Surgery - University of Insubria - via J.H. Dunant 5, 21100, Varese, Italy
| | - Evgenia Karousou
- From the Department of Medicine and Surgery - University of Insubria - via J.H. Dunant 5, 21100, Varese, Italy
| | - George W Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore 4 Medical Drive, Block MD10, Singapore, 117594, Singapore
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital Münster, Albert-Schweitzer-Campus 1, D11, 48149, Münster, Germany
| | - Paraskevi Heldin
- Department Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Alberto Passi
- From the Department of Medicine and Surgery - University of Insubria - via J.H. Dunant 5, 21100, Varese, Italy
| | - Davide Vigetti
- From the Department of Medicine and Surgery - University of Insubria - via J.H. Dunant 5, 21100, Varese, Italy.
| |
Collapse
|
4
|
Mori H, Saeki K, Chang G, Wang J, Wu X, Hsu PY, Kanaya N, Wang X, Somlo G, Nakamura M, Bild A, Chen S. Influence of Estrogen Treatment on ESR1+ and ESR1- Cells in ER + Breast Cancer: Insights from Single-Cell Analysis of Patient-Derived Xenograft Models. Cancers (Basel) 2021; 13:cancers13246375. [PMID: 34944995 PMCID: PMC8699443 DOI: 10.3390/cancers13246375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 01/07/2023] Open
Abstract
Simple Summary The benefit of endocrine therapy is normally observed for cancers with 10% or more of cells positive for ER expression. We compared the gene expression profiles in both ESR1+ and ESR1– cells in ER+ tumors following estrogen treatment. Our single-cell RNA sequencing analysis of estrogen-stimulated (SC31) and estrogen-suppressed (GS3) patient-derived xenograft models offered an unprecedented opportunity to address the molecular and functional differences between ESR1+ and ESR1– cells. While estrogen should activate ERα and stimulate ESR1+ cells, our findings regarding ESR1– cells were important, indicating that the proliferation of ESR1– cells in ER+ cancer is also influenced by estrogen. Another valuable finding from our studies was that estrogen also upregulated a tumor-suppressor gene, IL-24, only in GS3. Estrogen increased the percentage of cells expressing IL-24, associated with the estrogen-dependent inhibition of GS3 tumor growth. Abstract A 100% ER positivity is not required for an endocrine therapy response. Furthermore, while estrogen typically promotes the progression of hormone-dependent breast cancer via the activation of estrogen receptor (ER)-α, estrogen-induced tumor suppression in ER+ breast cancer has been clinically observed. With the success in establishing estrogen-stimulated (SC31) and estrogen-suppressed (GS3) patient-derived xenograft (PDX) models, single-cell RNA sequencing analysis was performed to determine the impact of estrogen on ESR1+ and ESR1– tumor cells. We found that 17β-estradiol (E2)-induced suppression of GS3 transpired through wild-type and unamplified ERα. E2 upregulated the expression of estrogen-dependent genes in both SC31 and GS3; however, E2 induced cell cycle advance in SC31, while it resulted in cell cycle arrest in GS3. Importantly, these gene expression changes occurred in both ESR1+ and ESR1– cells within the same breast tumors, demonstrating for the first time a differential effect of estrogen on ESR1– cells. E2 also upregulated a tumor-suppressor gene, IL-24, in GS3. The apoptosis gene set was upregulated and the G2M checkpoint gene set was downregulated in most IL-24+ cells after E2 treatment. In summary, estrogen affected pathologically defined ER+ tumors differently, influencing both ESR1+ and ESR1– cells. Our results also suggest IL-24 to be a potential marker of estrogen-suppressed tumors.
Collapse
Affiliation(s)
- Hitomi Mori
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
- Department of Surgery and Oncology, Graduate School of Medicine, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Kohei Saeki
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - Gregory Chang
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - Jinhui Wang
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, 655 Huntington Drive, Monrovia, CA 91016, USA; (J.W.); (X.W.)
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, 655 Huntington Drive, Monrovia, CA 91016, USA; (J.W.); (X.W.)
| | - Pei-Yin Hsu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - Xiaoqiang Wang
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
| | - George Somlo
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA 91010, USA; (G.S.); (A.B.)
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medicine, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Andrea Bild
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA 91010, USA; (G.S.); (A.B.)
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, CA 91010, USA; (H.M.); (K.S.); (G.C.); (P.-Y.H.); (N.K.); (X.W.)
- Correspondence: ; Tel.: +1-626-218-3454; Fax: +1-626-301-8972
| |
Collapse
|
5
|
Interleukin (IL)-24: Reconfiguring the Tumor Microenvironment for Eliciting Antitumor Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1290:99-110. [PMID: 33559858 DOI: 10.1007/978-3-030-55617-4_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Interleukin (IL)-24 is a member of the IL-10 family of cytokines. Due to its unique ability to function as both a tumor suppressor and cytokine, IL-24-based cancer therapy has been developed for treating a broad spectrum of human cancers. Majority of the studies reported to date have focused on establishing IL-24 as a cancer therapeutic by primarily focusing on tumor cell killing. However, the ability of IL-24 treatment on modulating the tumor microenvironment and immune response is underinvestigated. In this article, we summarize the biological and functional properties of IL-24 and the benefits of applying IL-24-based therapy for cancer.
Collapse
|
6
|
Gao W, Wen H, Liang L, Dong X, Du R, Zhou W, Zhang X, Zhang C, Xiang R, Li N. IL20RA signaling enhances stemness and promotes the formation of an immunosuppressive microenvironment in breast cancer. Theranostics 2021; 11:2564-2580. [PMID: 33456560 PMCID: PMC7806486 DOI: 10.7150/thno.45280] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022] Open
Abstract
Rationale: Tumor microenvironment interacts with tumor cells to regulate their stemness properties through various cytokines and cytokine receptors. Previous studies revealed the possible role of interleukin 20 receptor subunit alpha (IL20RA) signaling in the progression of several types of tumors. However, its regulatory effects on the stemness and the microenvironment of breast cancer need to be studied. Methods: Immunohistochemical staining and western blot analysis were used to evaluate the association between IL20RA and SOX2 in breast tumors and noncancerous tissues. Enzyme-linked immunosorbent assay and TCGA dataset analysis were performed to determine the function of IL20RA signaling in breast cancer progression. Gain- and loss-of-function methods were performed to examine the effects of IL20RA on the stemness of breast cancer cells. The stemness features were analyzed by detecting the expression of core stemness genes, side population (SP), sphere formation ability, and aldehyde dehydrogenase (ALDH) activity. Flow cytometric analysis was applied to detect the changes of tumor-infiltration lymphocytes in tumor tissues in mice. Based on the relevant molecular mechanisms elucidated in this study, a novel IL20RA-targeted liposomal nanoparticle encapsulating the signal transducer and activator of transcription 3 (STAT3) inhibitor stattic (NP-Stattic-IL20RA) was synthesized. These NPs were combined with anti-programmed death ligand 1 (PD-L1) antibody and chemotherapy to inhibit the development of breast tumors in mice. Results: IL20RA is highly expressed in human breast cancers and is positively associated with the SOX2 expression. IL20RA increases the SP and ALDHbr proportions of breast cancer cells, enhances the sphere formation ability, and promotes the expression of core stemness genes, such as Sox2 and Oct4, as well as increases chemoresistance of breast cancer cells. IL20RA promotes the tumor-initiating ability and lung metastasis of breast cancer cells in vivo. In addition, IL20RA activates the Janus kinase 1 (JAK1)-STAT3-SOX2 signaling pathway, leading to increased expression of PD-L1 and reduced recruitment of anti-cancer lymphocytes, including CD8+ T cells and natural killer cells. Meanwhile, IL20RA signaling enhances the proportion of myeloid-derived suppressor cells. Combined with anti-PD-L1 antibody and NPs-Stattic-IL20RA, the chemotherapeutic efficacy was increased in breast cancer mouse models in vivo. Conclusion: Collectively, our results reveal that the IL20RA pathway is a novel signaling pathway involved in promoting the stemness features of breast cancer along with the formation of a tumor-favorable immune microenvironment. Targeting the IL20RAhi population with STAT3 signaling inhibition combined with anti-PD-L1 antibody can increase the therapeutic efficacy of chemotherapeutic agents for breast cancer. This study thus introduces a promising novel strategy for breast cancer therapy.
Collapse
|
7
|
Emdad L, Bhoopathi P, Talukdar S, Pradhan AK, Sarkar D, Wang XY, Das SK, Fisher PB. Recent insights into apoptosis and toxic autophagy: The roles of MDA-7/IL-24, a multidimensional anti-cancer therapeutic. Semin Cancer Biol 2019; 66:140-154. [PMID: 31356866 DOI: 10.1016/j.semcancer.2019.07.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 06/21/2019] [Accepted: 07/19/2019] [Indexed: 12/18/2022]
Abstract
Apoptosis and autophagy play seminal roles in maintaining organ homeostasis. Apoptosis represents canonical type I programmed cell death. Autophagy is viewed as pro-survival, however, excessive autophagy can promote type II cell death. Defective regulation of these two obligatory cellular pathways is linked to various diseases, including cancer. Biologic or chemotherapeutic agents, which can reprogram cancer cells to undergo apoptosis- or toxic autophagy-mediated cell death, are considered effective tools for treating cancer. Melanoma differentiation associated gene-7 (mda-7) selectively promotes these effects in cancer cells. mda-7 was identified more than two decades ago by subtraction hybridization showing elevated expression during induction of terminal differentiation of metastatic melanoma cells following treatment with recombinant fibroblast interferon and mezerein (a PKC activating agent). MDA-7 was classified as a member of the IL-10 gene family based on its chromosomal location, and the presence of an IL-10 signature motif and a secretory sequence, and re-named interleukin-24 (MDA-7/IL-24). Multiple studies have established MDA-7/IL-24 as a potent anti-cancer agent, which when administered at supra-physiological levels induces growth arrest and cell death through apoptosis and toxic autophagy in a wide variety of tumor cell types, but not in corresponding normal/non-transformed cells. Furthermore, in a phase I/II clinical trial, MDA-7/IL-24 administered by means of a non-replicating adenovirus was well tolerated and displayed significant clinical activity in patients with multiple advanced cancers. This review examines our current comprehension of the role of MDA-7/IL-24 in mediating cancer-specific cell death via apoptosis and toxic autophagy.
Collapse
Affiliation(s)
- Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.
| | - Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Sarmistha Talukdar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Anjan K Pradhan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA.
| |
Collapse
|
8
|
Ubiquitination and SUMOylation in the chronic inflammatory tumor microenvironment. Biochim Biophys Acta Rev Cancer 2018; 1870:165-175. [DOI: 10.1016/j.bbcan.2018.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/10/2018] [Accepted: 08/15/2018] [Indexed: 12/28/2022]
|
9
|
Mao LJ, Ding M, Xu K, Pan J, Yu H, Yang C. Oncolytic Adenovirus Harboring Interleukin-24 Improves Chemotherapy for Advanced Prostate Cancer. J Cancer 2018; 9:4391-4397. [PMID: 30519344 PMCID: PMC6277655 DOI: 10.7150/jca.26437] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/29/2018] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Oncolytic adenoviruses emerge as new agents for cancer therapy. This study aimed to investigate the synergistic anti-tumor activity of oncolytic adenovirus armed with IL-24 (ZD55-IL-24) and docetaxel (DTX) on advanced prostate cancer in vitro and in vivo. METHODS DU145 prostate cancer cells or nude mice xenografted with DU145 prostate cancer cells were treated by ZD55-IL-24 and DTX alone or in combination. RESULTS DTX did not affect ZD55-IL-24 replication and IL-24 expression in DU145 cells. In vitro, the combination of ZD55-IL-24 and DTX showed synergistic inhibitory effects on prostate cancer cell viability and invasion. In vivo, ZD55-IL-24 and DTX synergistically inhibited the growth and activated the apoptosis of DU145 xenografts, accompanied by significantly decreased PARP-1 levels and increased caspase-3 and caspase-8 levels as well as decreased CD31 expression. CONCLUSION We reported the synergistic anti-tumor efficacy of ZD55-IL-24 and DTX on prostate cancer. Our results suggest that chemotherapy combined with oncolytic adenovirus mediated gene therapy is a promising strategy for the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Li-Jun Mao
- Department of Urinary Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Meng Ding
- Department of Urinary Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Kai Xu
- Department of Urinary Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Jun Pan
- Department of Urinary Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Haiyuan Yu
- Department of Urinary Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Chunhua Yang
- Department of Urinary Surgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.,Radiotherapy Department, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| |
Collapse
|
10
|
Autieri MV. IL-19 and Other IL-20 Family Member Cytokines in Vascular Inflammatory Diseases. Front Immunol 2018; 9:700. [PMID: 29681905 PMCID: PMC5897441 DOI: 10.3389/fimmu.2018.00700] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/21/2018] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular disease remains a major medical and socioeconomic burden in developed and developing countries and will increase with an aging and increasingly sedentary society. Many vascular diseases and atherosclerotic vascular disease, in particular, are essentially inflammatory disorders, involving multiple cell types. Communication between these cells is initiated and sustained by a complex network of cytokines and their receptors. The interleukin (IL)-20 family members, IL-19, IL-20, IL-22, and IL-24, initiate, sustain, and drive the progression of vascular disease. They are important in vascular disease as they facilitate a bidirectional cross-talk between resident vascular cells with immune cells. These cytokines are grouped into the same family based on shared common receptor subunits and signaling pathways. This communication is varied and can result in exacerbation, attenuation, and even repair of the vasculature. We will briefly review what is known about IL-20, IL-22, and IL-24 in cardiovascular biology. Because IL-19 is the most studied member of this family in terms of its role in vascular pathophysiological processes, the major emphasis of this review will focus on the expression and atheroprotective roles of IL-19 in vascular inflammatory disease.
Collapse
Affiliation(s)
- Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
11
|
Dabitao D, Hedrich CM, Wang F, Vacharathit V, Bream JH. Cell-Specific Requirements for STAT Proteins and Type I IFN Receptor Signaling Discretely Regulate IL-24 and IL-10 Expression in NK Cells and Macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:2154-2164. [PMID: 29436412 PMCID: PMC5840025 DOI: 10.4049/jimmunol.1701340] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/15/2018] [Indexed: 12/11/2022]
Abstract
Il10 forms a cytokine cluster with Il19, Il20, and Il24 in a conserved region of chromosome 1. The latter genes are in the IL-20 subfamily of IL-10-related cytokines and, although they are not as well studied their biologic actions and expression patterns, seem to have little in common with IL-10. IL-24, like IL-10, however, is uniquely expressed in T cells and is a signature gene of the Th2 lineage, which suggests they could be coregulated in certain cell types. Little is known about other cellular sources of IL-24. We investigated IL-24 and IL-10 expression in murine macrophages and NK cells, and found that although they are coexpressed under most stimulation conditions, IL-24 and IL-10 are controlled by distinct, cell type-specific pathways. In bone marrow-derived macrophages, optimal IL-24 expression required LPS+IL-4 costimulation and STAT6 but was independent of type I IFN receptor signaling and STAT4. Conversely, LPS-induced IL-10 was independent of IL-4/STAT6 and STAT4 but, consistent with other reports, required type I IFN receptor signaling for optimal expression. Remarkably, NK-specific IL-24 (but not IL-10) expression was dependent on both type I IFN receptor signaling and STAT4. Induction of IL-24 expression was accompanied by cell-specific recruitment of STAT6 and STAT4 to multiple sites that we identified within Il24, which mediated STAT-dependent histone modifications across the gene. Collectively, our results indicate that despite being coexpressed, IL-10 and IL-24 are independently regulated by different type I IFN receptor signaling pathways in innate immune cells and provide insight into the mechanisms that fine-tune cell type-specific gene expression within the Il10 cluster.
Collapse
Affiliation(s)
- Djeneba Dabitao
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205; and
| | - Christian M Hedrich
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205; and
| | - Fengying Wang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205; and
| | - Vimvara Vacharathit
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205; and
- Graduate Program in Immunology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Jay H Bream
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205; and
- Graduate Program in Immunology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| |
Collapse
|
12
|
Abstract
Subtraction hybridization identified genes displaying differential expression as metastatic human melanoma cells terminally differentiated and lost tumorigenic properties by treatment with recombinant fibroblast interferon and mezerein. This approach permitted cloning of multiple genes displaying enhanced expression when melanoma cells terminally differentiated, called melanoma differentiation associated (mda) genes. One mda gene, mda-7, has risen to the top of the list based on its relevance to cancer and now inflammation and other pathological states, which based on presence of a secretory sequence, chromosomal location, and an IL-10 signature motif has been named interleukin-24 (MDA-7/IL-24). Discovered in the early 1990s, MDA-7/IL-24 has proven to be a potent, near ubiquitous cancer suppressor gene capable of inducing cancer cell death through apoptosis and toxic autophagy in cancer cells in vitro and in preclinical animal models in vivo. In addition, MDA-7/IL-24 embodied profound anticancer activity in a Phase I/II clinical trial following direct injection with an adenovirus (Ad.mda-7; INGN-241) in tumors in patients with advanced cancers. In multiple independent studies, MDA-7/IL-24 has been implicated in many pathological states involving inflammation and may play a role in inflammatory bowel disease, psoriasis, cardiovascular disease, rheumatoid arthritis, tuberculosis, and viral infection. This review provides an up-to-date review on the multifunctional gene mda-7/IL-24, which may hold potential for the therapy of not only cancer, but also other pathological states.
Collapse
|
13
|
Obesity Enhances the Conversion of Adipose-Derived Stromal/Stem Cells into Carcinoma-Associated Fibroblast Leading to Cancer Cell Proliferation and Progression to an Invasive Phenotype. Stem Cells Int 2017. [PMID: 29527228 PMCID: PMC5748106 DOI: 10.1155/2017/9216502] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Obesity is associated with enhanced tumor growth and progression. Within the adipose tissue are adipose-derived stromal/stem cells (ASCs) that have been shown to convert into carcinoma-associated fibroblast (CAFs) in the presence of tumor-derived factors. However, the impact of obesity on the ASCs and on the conversion of ASCs into CAFs has not been demonstrated. In the current study, ASCs isolated from lean donors (BMI < 25; lnASCs) were compared with ASCs isolated from obese donors (BMI > 30, obASCs). The contribution of tumor-derived factors on the conversion of ASCs to CAFs was investigated. Following exposure to cancer cells, obASCs expressed higher levels of CAF markers, including NG2, alpha-SMA, VEGF, FAP, and FSP, compared to lnASCs. To investigate the crosstalk between ASCs and breast cancer cells, MCF7 cells were serially cocultured with lnASCs or obASCs. After coculture with lnASCs and obASCs, MCF7 cells demonstrated enhanced proliferation and expressed an invasive phenotype morphologically, with more pronounced effects following exposure to obASCs. Long-term exposure to obASCs also enhanced the expression of protumorgenic factors. Together, these results suggest that obesity alters ASCs to favor their rapid conversion into CAFs, which in turn enhances the proliferative rate, the phenotype, and gene expression profile of breast cancer cells.
Collapse
|
14
|
Shabgah AG, Navashenaq JG, Shabgah OG, Mohammadi H, Sahebkar A. Interleukin-22 in human inflammatory diseases and viral infections. Autoimmun Rev 2017; 16:1209-1218. [PMID: 29037907 DOI: 10.1016/j.autrev.2017.10.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 08/08/2017] [Indexed: 12/24/2022]
Abstract
Interleukin-22 (IL22) is one of the members of IL10 family. Elevated levels of this cytokine can be seen in diseases caused by T lymphocytes, such as Psoriasis, Rheumatoid arthritis, interstitial lung diseases. IL22 is produced by different cells in both innate and acquired immunities. Different types of T cells are able to produce IL22, but the major IL22-producing T-cell is the TCD4. TH22 cell is a new line of TCD4 cells, which differentiated from naive T cells in the presence of TNFα and IL6; 50% of peripheral blood IL22 is produced by these cells. IL22 has important functions in host defense at mucosal surfaces as well as in tissue repair. In this review, we assess the current understanding of this cytokine and focus on the possible roles of IL-22 in autoimmune diseases.
Collapse
Affiliation(s)
- Arezoo Gowhari Shabgah
- Immunology Research Center, Avicenna Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Blood Borne Infections Research Center, AcademicCenter for Education, Culture and Research (ACECR), Razavi Khorasan Branch,Mashhad, Iran
| | - Jamshid Gholizadeh Navashenaq
- Immunology Research Center, Avicenna Research Institute, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Omid Gohari Shabgah
- Parasitology Department, Medical sciencesfaculty, Tarbiat Modares University, Tehran, Iran
| | - Hamed Mohammadi
- ImmunologyResearch Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Sahebkar
- BiotechnologyResearch Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Valle Oseguera CA, Spencer JV. Human cytomegalovirus interleukin-10 enhances matrigel invasion of MDA-MB-231 breast cancer cells. Cancer Cell Int 2017; 17:24. [PMID: 28228690 PMCID: PMC5307693 DOI: 10.1186/s12935-017-0399-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 02/08/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND While some risk factors for breast cancer are well-known, the influence of other factors, particularly virus infection, remains unclear. Human cytomegalovirus (HCMV) is widespread in the general population, and both molecular and epidemiological evidence has indicated links between HCMV and breast cancer. The HCMV protein cmvIL-10 is a potent suppressor of immune function that has also been shown to promote proliferation and migration of breast cancer cells. In this study, the impact of cmvIL-10 on tumor cell invasion through a simulated basement membrane was investigated. RESULTS MDA-MB-231 breast cancer cells exhibited invasion through a matrigel layer that was significantly enhanced in the presence of either purified cmvIL-10 or supernatants from HCMV-infected cells containing secreted cmvIL-10. Transcriptional profiling revealed that cmvIL-10 altered expression of several genes implicated in metastasis. Exposure to cmvIL-10 resulted in higher MMP-3 mRNA levels, greater protein expression, and increased enzymatic activity. Treatment with cmvIL-10 also increased expression of both urokinase plasminogen receptor (uPAR) and plasminogen activator inhibitor-1 (PAI-1), which can stimulate MMP-3 activity and have previously been identified as poor prognostic markers in breast cancer patients. Finally, MDA-MB-231 cells treated with cmvIL-10 showed significant downregulation of metastasis suppressor 1 (MTSS1), a scaffolding protein that regulates cytoskeletal rearrangements and is frequently lost in metastatic tumors. CONCLUSIONS HCMV, and in particular the secreted viral cytokine, cmvIL-10, can induce cellular changes that facilitate cell migration and invasion. These findings indicate that HCMV may be associated with promoting the malignant spread of breast cancer cells and suggest that antiviral treatment may be a useful complement to chemotherapy in some patients.
Collapse
Affiliation(s)
- Cendy A Valle Oseguera
- Department of Biology, University of San Francisco, 2130 Fulton Street, San Francisco, CA 94117 USA
| | - Juliet V Spencer
- Department of Biology, University of San Francisco, 2130 Fulton Street, San Francisco, CA 94117 USA
| |
Collapse
|
16
|
Cao H, Xiang T, Zhang C, Yang H, Jiang L, Liu S, Huang X. MDA7 combined with targeted attenuated Salmonella vector SL7207/pBud-VP3 inhibited growth of gastric cancer cells. Biomed Pharmacother 2016; 83:809-815. [PMID: 27497809 DOI: 10.1016/j.biopha.2016.07.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 07/07/2016] [Accepted: 07/13/2016] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND/AIM To investigate the therapeutic effect of MDA7 combined with apoptin targeted attenuated Salmonella typhimurium vector SL7207/pBud-VP3 on gastric cancer cells. MATERIALS AND METHODS MDA7 was inserted into pBud-VP3 using molecular cloning technology to obtain the eukaryotic expression plasmid pBud-VP3-MDA7 and it was transformed into attenuated Salmonella typhimurium SL7207 by high voltage electroporation to obtain SL7207/pBud-VP3-MDA7. Mice bearing a sarcoma of gastric cancer cells were treated with SL7207/pBud-VP3-MDA7 and the growth-suppressing effect was assessed by measurement of tumor volume. Western blot was used to identify the MDA7 expression products. IL-6, INF-γ, TNF-α and caspase-3, VEGF in tumor tissue were detected by RT-PCR and immunohistochemistry. RESULTS SL7207/pBud-VP3-MDA7 was successfully constructed and expression of the protein MDA7 was identified in tumor tissue. SL7207/pBud-VP3-MDA7 significantly caused tumor inhibition and regression (p<0.05). The level of expression of cytokines IL-6, INF-γ, TNF-α in tumor tissue was significantly higher than in the other groups (p<0.05). The expression of caspase-3 was up-regulated and VEGF was down-regulated (p<0.05). CONCLUSION This study shows that SL7207/pBud-VP3-MDA7 has inhibitory effect on the growth of gastric cancer cells. The mechanism involved is related to the promotion of tumor apoptosis, immunity regulation and inhibition of tumor blood vessels.
Collapse
Affiliation(s)
- Hongdan Cao
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Tingxiu Xiang
- Artron BioResearch Inc., 3938 North Fraser Way, Burnaby, BC V5 J 5H6, Canada
| | - Chaohong Zhang
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Hong Yang
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Lingqun Jiang
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Shanli Liu
- Chongqing Medical and Pharmaceutical Higher specialty College, Road 82, Shapingba District University City, Chongqing 401331, China
| | - Xiaolan Huang
- Ph.D Research Center for Medical and Social Development, Chongqing Medical University, Road 1, Yuzhong District School of Medicine, Chongqing 400016, China.
| |
Collapse
|
17
|
MA QUNFENG, JIN BANGMING, ZHANG YAO, SHI YINAN, ZHANG CHI, LUO DAN, WANG PENGKUN, DUAN CUIMI, SONG HEYU, LI XUE, DENG XUEFENG, CHEN ZHINAN, WANG ZILING, JIANG HONG, LIU YAN. Secreted recombinant human IL-24 protein inhibits the proliferation of esophageal squamous cell carcinoma Eca-109 cells in vitro and in vivo. Oncol Rep 2016; 35:2681-90. [DOI: 10.3892/or.2016.4633] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 12/16/2015] [Indexed: 11/05/2022] Open
|
18
|
Menezes ME, Shen XN, Das SK, Emdad L, Guo C, Yuan F, Li YJ, Archer MC, Zacksenhaus E, Windle JJ, Subler MA, Ben-David Y, Sarkar D, Wang XY, Fisher PB. MDA-7/IL-24 functions as a tumor suppressor gene in vivo in transgenic mouse models of breast cancer. Oncotarget 2015; 6:36928-42. [PMID: 26474456 PMCID: PMC4741906 DOI: 10.18632/oncotarget.6047] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/23/2015] [Indexed: 12/31/2022] Open
Abstract
Melanoma differentiation associated gene-7/Interleukin-24 (MDA-7/IL-24) is a novel member of the IL-10 gene family that selectively induces apoptosis and toxic autophagy in a broad spectrum of human cancers, including breast cancer, without harming normal cells or tissues. The ability to investigate the critical events underlying cancer initiation and progression, as well as the capacity to test the efficacy of novel therapeutics, has been significantly advanced by the development of genetically engineered mice (GEMs) that accurately recapitulate specific human cancers. We utilized three transgenic mouse models to better comprehend the in vivo role of MDA-7/IL-24 in breast cancer. Using the MMTV-PyMT spontaneous mammary tumor model, we confirmed that exogenously introducing MDA-7/IL-24 using a Cancer Terminator Virus caused a reduction in tumor burden and also produced an antitumor "bystander" effect. Next we performed xenograft studies in a newly created MMTV-MDA-7 transgenic model that over-expresses MDA-7/IL-24 in the mammary glands during pregnancy and lactation, and found that MDA-7/IL-24 overexpression delayed tumor growth following orthotopic injection of a murine PDX tumor cell line (mPDX) derived from a tumor formed in an MMTV-PyMT mouse. We also crossed the MMTV-MDA-7 line to MMTV-Erbb2 transgenic mice and found that MDA-7/IL-24 overexpression delayed the onset of mammary tumor development in this model of spontaneous mammary tumorigenesis as well. Finally, we assessed the role of MDA-7/IL-24 in immune regulation, which can potentially contribute to tumor suppression in vivo. Our findings provide further direct in vivo evidence for the role of MDA-7/IL-24 in tumor suppression in breast cancer in immune-competent transgenic mice.
Collapse
Affiliation(s)
- Mitchell E. Menezes
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Xue-Ning Shen
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Swadesh K. Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Fang Yuan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - You-Jun Li
- Department of Anatomy, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Michael C. Archer
- Departments of Medical Biophysics, University of Toronto, Ontario, Canada
- Nutritional Sciences, University of Toronto, Ontario, Canada
| | - Eldad Zacksenhaus
- Departments of Medical Biophysics, University of Toronto, Ontario, Canada
- Toronto General Research Institute - University Health Network, Toronto, Ontario, Canada
| | - Jolene J. Windle
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Mark A. Subler
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Yaacov Ben-David
- Departments of Medical Biophysics, University of Toronto, Ontario, Canada
- Division of Biology, the Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, China
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| | - Paul B. Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
- VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
19
|
Iankov ID, Kurokawa CB, D'Assoro AB, Ingle JN, Domingo-Musibay E, Allen C, Crosby CM, Nair AA, Liu MC, Aderca I, Federspiel MJ, Galanis E. Inhibition of the Aurora A kinase augments the anti-tumor efficacy of oncolytic measles virotherapy. Cancer Gene Ther 2015; 22:438-44. [PMID: 26272026 PMCID: PMC4589445 DOI: 10.1038/cgt.2015.36] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 06/03/2015] [Accepted: 06/19/2015] [Indexed: 12/20/2022]
Abstract
Oncolytic measles virus (MV) strains have demonstrated broad spectrum preclinical anti-tumor, including breast cancer. Aurora A kinase controls mitotic spindle formation and plays a critical role in malignant transformation. We hypothesized that, by causing mitotic arrest, the Aurora A kinase inhibitor MLN8237 (alisertib) can increase MV oncolytic effect and efficacy. Alisertib enhanced MV oncolysis in vitro and significantly improved outcome in vivo against breast cancer xenografts. In a disseminated MDA-231-lu-P4 lung metastatic model, the MV/alisertib combination treatment markedly increased median survival to 82.5 days with 20% of the animals being long term survivors vs. 48 days median survival for the control animals. Similarly, in a pleural effusion model of advanced breast cancer, the MV/alisertib combination significantly improved outcome with a 74.5 day median survival versus the single agent groups (57 and 40 days respectively). Increased viral gene expression and IL-24 upregulation were demonstrated, representing possible mechanisms for the observed increase in antitumor effect. Inhibiting Aurora A kinase with alisertib represents a novel approach to enhance measles virus-mediated oncolysis and antitumor effect. Both oncolytic MV strains and alisertib are currently tested in clinical trials, this study therefore provides the basis for translational applications of this combinatorial strategy in the treatment of patients with advanced breast cancer.
Collapse
Affiliation(s)
- I D Iankov
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - C B Kurokawa
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - A B D'Assoro
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - J N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | | | - C Allen
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - C M Crosby
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - A A Nair
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - M C Liu
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - I Aderca
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - M J Federspiel
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - E Galanis
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Oncology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
20
|
Pang H, Zhao H. Stratified pathway analysis to identify gene sets associated with oral contraceptive use and breast cancer. Cancer Inform 2014; 13:73-8. [PMID: 25574128 PMCID: PMC4263464 DOI: 10.4137/cin.s13973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 08/15/2014] [Accepted: 08/19/2014] [Indexed: 01/02/2023] Open
Abstract
Cancer biomarker discovery can facilitate drug development, improve staging of patients, and predict patient prognosis. Because cancer is the result of many interacting genes, analysis based on a set of genes with related biological functions or pathways may be more informative than single gene-based analysis for cancer biomarker discovery. The relevant pathways thus identified may help characterize different aspects of molecular phenotypes related to the tumor. Although it is well known that cancer patients may respond to the same treatment differently because of clinical variables and variation of molecular phenotypes, this patient heterogeneity has not been explicitly considered in pathway analysis in the literature. We hypothesize that combining pathway and patient clinical information can more effectively identify relevant pathways pertinent to specific patient subgroups, leading to better diagnosis and treatment. In this article, we propose to perform stratified pathway analysis based on clinical information from patients. In contrast to analysis using all the patients, this more focused analysis has the potential to reveal subgroup-specific pathways that may lead to more biological insights into disease etiology and treatment response. As an illustration, the power of our approach is demonstrated through its application to a breast cancer dataset in which the patients are stratified according to their oral contraceptive use.
Collapse
Affiliation(s)
- Herbert Pang
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA. ; School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
21
|
Valle Oseguera CA, Spencer JV. cmvIL-10 stimulates the invasive potential of MDA-MB-231 breast cancer cells. PLoS One 2014; 9:e88708. [PMID: 24520416 PMCID: PMC3919807 DOI: 10.1371/journal.pone.0088708] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 01/10/2014] [Indexed: 01/05/2023] Open
Abstract
Cancer is the result of unregulated cell growth that leads to tumor formation, and in many cases, metastases. Although there are several risk factors associated with cancer, one area that remains poorly understood is the impact of infectious disease. Human cytomegalovirus (HCMV) is a member of the herpesvirus family that is highly prevalent in the population. HCMV usually causes clinical disease only in immune compromised individuals, but recent evidence suggests that HCMV may be strongly associated with some forms of cancer, particularly glioblastoma and breast cancer. We investigated the possibility that cmvIL-10, a viral cytokine with homology to human IL-10 that is secreted from infected cells, could act in a paracrine manner to alter the tumor microenvironment, induce cell signaling, and increase the invasive potential of cancer cells. We found that human MDA-MB-231 breast cancer cells express the IL-10 receptor and that exposure to cmvIL-10 results in activation of Stat3, a transcription factor strongly associated with enhanced metastatic potential and chemo-resistance. In addition, cmvIL-10 stimulated an increase in DNA synthesis and cell proliferation, protected MDA-MB-231 cells from etoposide-induced apoptosis, and also greatly enhanced chemotaxis toward epidermal growth factor (EGF). These results suggest a significant and wide-ranging role for cmvIL-10 in the progression of breast cancer and could have broad implications for the diagnosis and treatment of cancer in HCMV-positive patients.
Collapse
Affiliation(s)
- Cendy A. Valle Oseguera
- Department of Biology, University of San Francisco, San Francisco, California, United States of America
| | - Juliet V. Spencer
- Department of Biology, University of San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
22
|
Molecular targets and signaling pathways regulated by interleukin (IL)-24 in mediating its antitumor activities. J Mol Signal 2013; 8:15. [PMID: 24377906 PMCID: PMC3879428 DOI: 10.1186/1750-2187-8-15] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/21/2013] [Indexed: 01/06/2023] Open
Abstract
Cancer remains a major health issue in the world and the effectiveness of current therapies is limited resulting in disease recurrence and resistance to therapy. Therefore to overcome disease recurrence and have improved treatment efficacy there is a continued effort to develop and test new anticancer drugs that are natural or synthetic - (conventional chemotherapeutics, small molecule inhibitors) and biologic (antibody, tumor suppressor genes, oligonucleotide) product. In parallel, efforts for identifying molecular targets and signaling pathways to which cancer cells are "addicted" are underway. By inhibiting critical signaling pathways that is crucial for cancer cell survival, it is expected that the cancer cells will undergo a withdrawal symptom akin to "de-addiction" resulting in cell death. Thus, the key for having an improved and greater control on tumor growth and metastasis is to develop a therapeutic that is able to kill tumor cells efficiently by modulating critical signaling pathways on which cancer cells rely for their survival.Currently several small molecule inhibitors targeted towards unique molecular signaling pathways have been developed and tested in the clinic. Few of these inhibitors have shown efficacy while others have failed. Thus, targeting a single molecule or pathway may be insufficient to completely block cancer cell proliferation and survival. It is therefore important to identify and test an anticancer drug that can inhibit multiple signaling pathways in a cancer cell, control growth of both primary and metastatic tumors and is safe.One biologic agent that has the characteristics of serving as a potent anticancer drug is interleukin (IL)-24. IL-24 suppresses multiple signaling pathways in a broad-spectrum of human cancer cells leading to tumor cell death, inhibition of tumor angiogenesis and metastasis. Additionally, combining IL-24 with other therapies demonstrated additive to synergistic antitumor activity. Clinical testing of IL-24 as a gene-based therapeutic for the treatment of solid tumors demonstrated that IL-24 is efficacious and is safe. The unique features of IL-24 support its further development as an anticancer drug for cancer treatment.In this review we summarize the current understanding on the molecular targets and signaling pathways regulated by IL-24 in mediating its anticancer activity.
Collapse
|
23
|
Li YJ, Liu G, Li Y, Vecchiarelli-Federico LM, Liu JC, Zacksenhaus E, Shan SW, Yang BB, Li Q, Dash R, Fisher PB, Archer MC, Ben-David Y. mda-7/IL-24 expression inhibits breast cancer through upregulation of growth arrest-specific gene 3 (gas3) and disruption of β1 integrin function. Mol Cancer Res 2013; 11:593-603. [PMID: 23468528 DOI: 10.1158/1541-7786.mcr-12-0496] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Melanoma differentiation-associated gene (MDA)-7)/interleukin (IL)-24, a member of the IL-10 family of cytokines, inhibits growth of various human cancer cells, yet the underlying mechanism is largely unknown. Here, we report that mda-7/IL-24 efficiently suppresses the development of rat mammary tumors in vivo. Microarray analysis for genes differentially expressed in rat mammary tumor cells overexpressing MDA-7/IL-24 compared with those that do not express this cytokine identified growth arrest-specific gene-3 (gas3) as a target for mda-7/IL-24. Upregulation of gas3 by mda-7/IL-24 was STAT3 dependent. Induction of gas3 inhibited attachment and proliferation of tumor cells in vitro and in vivo by inhibiting the interaction of β1 integrin with fibronectin. A mutated GAS3, which is unable to bind β1 integrin, was also unable to inhibit fibronectin-mediated attachment and cell growth both in adherent and suspension cultures, suggesting that GAS3 exerts its effects through interaction with and regulation of β1 integrin. Thus, mda-7/IL-24 inhibits breast cancer growth, at least in part, through upregulation of GAS3 and disruption of β1 integrin function. Importantly, the expression of the mda-7/IL-24 receptor, IL-20R1, is highly correlated with GAS3 expression in human breast cancer (P = 1.02 × 10(-9)), and the incidence of metastases is significantly reduced in patients with HER2(+) breast cancer expressing high-levels of IL-20R1. Together, our results identify a novel MDA-7/IL-24-GAS3-β1integrin-fibronectin signaling pathway that suppresses breast cancer growth and can be targeted for therapy.
Collapse
Affiliation(s)
- You-Jun Li
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Interleukin-24 (IL-24), a member of the IL-10 cytokine family whose physiological function remains largely unknown, has been shown to induce apoptosis when expressed in an adenoviral background. It is yet little understood, why IL-24 alone induced apoptosis only in a limited number of tumor cell lines. Analyzing an influenza A virus vector expressing IL-24 for its oncolytic potential revealed enhanced pro-apoptotic activity of the chimeric virus compared with virus or IL-24 alone. Interestingly, IL-24-mediated enhancement of influenza-A-induced apoptosis did not require viral replication but critically depended on toll-like receptor 3 (TLR3) and caspase-8. Immunoprecipitation of TLR3 showed that infection by influenza A virus induced formation of a TLR3-associated signaling complex containing TRIF, RIP1, FADD, cFLIP and pro-caspase-8. Co-administration of IL-24 decreased the presence of cFLIP in the TLR3-associated complex, converting it into an atypical, TLR3-associated death-inducing signaling complex (TLR3 DISC) that induced apoptosis by enabling caspase-8 activation at this complex. The sensitizing effect of IL-24 on TLR3-induced apoptosis, mediated by influenza A virus or the TLR3-specific agonist poly(I:C), was also evident on tumor spheroids. In conclusion, rather than acting as an apoptosis inducer itself, IL-24 sensitizes cancer cells to TLR-mediated apoptosis by enabling the formation of an atypical DISC which, in the case of influenza A virus or poly(I:C), is associated with TLR3.
Collapse
|
25
|
Lee KM, Kang HA, Park M, Lee HY, Song MJ, Ko K, Oh JW, Kang HS. Interleukin-24 Suppresses the Growth of Vascular Smooth Muscle Cells by Inhibiting H 2O 2-Induced Reactive Oxygen Species Production. Pharmacology 2012; 90:332-41. [DOI: 10.1159/000343242] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 09/04/2012] [Indexed: 11/19/2022]
|
26
|
Zhu W, Wei L, Zhang H, Chen J, Qin X. Oncolytic adenovirus armed with IL-24 inhibits the growth of breast cancer in vitro and in vivo. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:51. [PMID: 22640485 PMCID: PMC3511263 DOI: 10.1186/1756-9966-31-51] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 04/06/2012] [Indexed: 11/18/2022]
Abstract
Background Interleukin-24 (IL-24) is a cytokine that belongs to the IL-10 family. It can selectively induce cancer cell apoptosis which has been utilized as a cancer gene therapy strategy. Methods A recombinant type five adenovirus containing IL-24 gene (designated CNHK600-IL24) was constructed, whose replication is activated only in tumor cells. The replication of CNHK600-IL24 in breast tumor cells and fibroblasts were assessed by TCID50 and MTT assay; the secretion of IL-24 was measured by ELISA and western blotting. The in vivo anti-tumor effect of CNHK600-IL24 was investigated in nude mice carrying orthotopic or metastatic breast tumor. Results We observed that CNHK600-IL24 could replicate efficiently and resulted in high level IL-24 expression and massive cell death in human breast cancer cell MDA-MB-231 but not in normal fibroblast cell MRC-5. In addition, orthotopic breast tumor growth in the nude mice model was significantly suppressed when CNHK600-IL24 was administered. In the metastatic model generated by tail vein injection, CNHK600-IL24 virotherapy significantly improved survival compared with the same virus expressing EGFP (median survival CNHK600-IL24, 55 days vs. CNHK600-EGFP, 41 day, p < 0.05 Mantal-Cox test). A similar phenomenon was observed in the metastatic model achieved by left ventricular injection as suggested by in vivo luminescence imaging of tumor growth. Conclusion The oncolytic adenovirus armed with IL-24, which exhibited enhanced anti-tumor activity and improved survival, is a promising candidate for virotherapy of breast cancer.
Collapse
Affiliation(s)
- Wei Zhu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | | | | | | | | |
Collapse
|
27
|
Pan X, Wu L, Cao J, Guo W, Wang Z, Han B, Hu W. Recombinant adenovirus vector-mediated human MDA-7 gene transfection suppresses hepatocellular carcinoma growth in a mouse xenograft model. J Biomed Res 2012; 26:53-8. [PMID: 23554730 PMCID: PMC3596080 DOI: 10.1016/s1674-8301(12)60007-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 11/04/2011] [Accepted: 12/17/2011] [Indexed: 11/28/2022] Open
Abstract
Hepatocellular carcinoma is one of the most common tumors in the world. The purpose of the present study was to investigate the inhibitory effects of adenoviral transduction of human melanoma differentiation-associated gene-7 (MDA-7) gene on hepatocellular carcinoma, so as to provide a theoretical basis for gene therapy of the disease. The human MDA-7 gene was cloned into replication-defective adenovirus specific to HepG2 cells using recombinant virus technology. RT-PCR and Western blotting assays were used to determine the expression of human MDA-7 mRNA and MDA-7 protein in HepG2 cells in vitro. Induction of apoptosis by overexpression of the human MDA-7 gene was determined by flow cytometry. In-vivo efficacy of adenoviral delivery of the human MDA-7 gene was assessed in nude mice bearing HepG2 cell lines in vivo by determining inhibition of tumor growth, VEGF and CD34 expression, and microvascular density (MVD). The results showed that AdGFP/MDA-7 induced apoptosis of HepG2 cells in vitro and significantly inhibited tumor growth in vivo (P < 0.05). The intratumoral MVD decreased significantly in the treated tumors (P < 0.05). We conclude the recombination adenovirus AdGFP/MDA-7 can effectively express biologically active human MDA-7, which leads to inhibition of hepatocellular carcinoma growth.
Collapse
Affiliation(s)
- Xinting Pan
- Department of Hepatobiliary Surgery, the Affiliated Hospital of Medical College of Qingdao University, Qingdao, Shandong 266000, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Buzas K, Oppenheim JJ, Zack Howard OM. Myeloid cells migrate in response to IL-24. Cytokine 2011; 55:429-34. [PMID: 21703864 PMCID: PMC3148305 DOI: 10.1016/j.cyto.2011.05.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 05/08/2011] [Accepted: 05/21/2011] [Indexed: 01/23/2023]
Abstract
IL-24 (melanoma differentiation associated gene 7 product) is a member of the IL-10 cytokine family that has been reported to possess anti-tumor activity. IL-24 is produced by immune tissues and its expression can be induced in human peripheral blood mononuclear cells by pathogen-associated molecules. While immune cells are known to produce IL-24, the response of immune cells to IL-24 is unclear. Using recombinant human IL-24, we demonstrated that IL-24 induces human monocyte and neutrophil migration, in vitro. An in vivo chemotaxis model showed that IL-24 attracted CD11b positive myeloid cells. To further characterize the chemotactic IL-24 response and type(s) of receptor(s) utilized by IL-24, we treated monocytes with signaling pathway inhibitors. IL-24-induced migration was reduced by pertussis toxin treatment, thus implicating G-protein coupled receptors in this process. Additionally, MEK and JAK inhibitors markedly decreased monocyte migration toward IL-24. These results suggest that IL-24 activates several signaling cascades in immune cells eliciting migration of myeloid cells, which may contribute to the known anti-cancer effects of IL-24.
Collapse
Affiliation(s)
- Krisztina Buzas
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, National Cancer Institute-Frederick, Center for Cancer Research, Frederick MD, USA 21702
| | - Joost J. Oppenheim
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, National Cancer Institute-Frederick, Center for Cancer Research, Frederick MD, USA 21702
| | - O. M. Zack Howard
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, National Cancer Institute-Frederick, Center for Cancer Research, Frederick MD, USA 21702
| |
Collapse
|
29
|
Adenovirus-mediated human interleukin 24 (MDA-7/IL-24) selectively suppresses proliferation and induces apoptosis in keloid fibroblasts. Ann Plast Surg 2011; 66:660-6. [PMID: 21042181 DOI: 10.1097/sap.0b013e3181e05039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Keloids are fibroproliferative dermal lesions characterized by the proliferation of fibroblasts and the formation of excess scar tissue, for which no effective treatment exists. We transfected a replication-incompetent adenovirus vector expressing green fluorescent protein and interleukin-24 gene (Ad-GFP/IL-24) into keloid fibroblasts (KF) and normal dermal fibroblasts (NDF) in vitro to investigate the suppression effects by observation on cell lines growth, apoptosis, mitosis cycle, etc. The expression of GFP and IL-24 mRNA confirmed that Ad-GFP/IL-24 was transfected into KF and NDF successfully. The expression level of secreting IL-24 protein detected by enzyme-linked immunosorbent assay in Ad-GFP/IL-24-treated KF and PBS-treated NDF was higher than controls; treatment with Ad-GFP/IL-24 in KF induced growth suppression (71.83% ± 6.67%, P < 0.05 to 9.79% ± 3.34%, P < 0.01), apoptosis (24.2% ± 3.08% to 66.51% ± 5.29%, P < 0.01) and increased the percentage of the G2/M phase (42.26% ± 6.44%, P < 0.01) in KF but not in NDF. The data showed that the exogenous IL-24 gene could selectively inhibit human KF proliferation and induce significant apoptosis.
Collapse
|
30
|
Splice variants of mda-7/IL-24 differentially affect survival and induce apoptosis in U2OS cells. Cytokine 2011; 56:272-81. [PMID: 21843952 DOI: 10.1016/j.cyto.2011.07.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Revised: 07/17/2011] [Accepted: 07/18/2011] [Indexed: 11/20/2022]
Abstract
Interleukin-24 (mda-7/IL-24) is a cytokine in the IL-10 family that has received a great deal of attention for its properties as a tumor suppressor and as a potential treatment for cancer. In this study, we have identified and characterized five alternatively spliced isoforms of this gene. Several, but not all of these isoforms induce apoptosis in the osteosarcoma cell line U2OS, while none affect the survival of the non-cancerous NOK cell line. One of these isoforms, lacking three exons and encoding the N-terminal end of the mda-7/IL-24 protein sequence, caused levels of apoptosis that were higher than those caused by the full-length mda-7/IL-24 variant. Additionally, we found that the ratio of isoform expression can be modified by the splice factor SRp55. This regulation suggests that alternative splicing of mda-7/IL-24 is under tight control in the cell, and can be modified under various cellular conditions, such as DNA damage. In addition to providing new insights into the function of an important tumor suppressor gene, these findings may also point toward new avenues for cancer treatment.
Collapse
|
31
|
Dash R, Bhutia SK, Azab B, Su ZZ, Quinn BA, Kegelmen TP, Das SK, Kim K, Lee SG, Park MA, Yacoub A, Rahmani M, Emdad L, Dmitriev IP, Wang XY, Sarkar D, Grant S, Dent P, Curiel DT, Fisher PB. mda-7/IL-24: a unique member of the IL-10 gene family promoting cancer-targeted toxicity. Cytokine Growth Factor Rev 2011; 21:381-91. [PMID: 20926331 DOI: 10.1016/j.cytogfr.2010.08.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24) is a unique member of the IL-10 gene family that displays nearly ubiquitous cancer-specific toxicity, with no harmful effects toward normal cells or tissues. mda-7/IL-24 was cloned from human melanoma cells by differentiation induction subtraction hybridization (DISH) and promotes endoplasmic reticulum (ER) stress culminating in apoptosis or toxic autophagy in a broad-spectrum of human cancers, when assayed in cell culture, in vivo in human tumor xenograft mouse models and in a Phase I clinical trial in patients with advanced cancers. This therapeutically active cytokine also induces indirect antitumor activity through inhibition of angiogenesis, stimulation of an antitumor immune response, and sensitization of cancer cells to radiation-, chemotherapy- and antibody-induced killing.
Collapse
Affiliation(s)
- Rupesh Dash
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Immunologic messenger molecules: cytokines, interferons, and chemokines. J Allergy Clin Immunol 2009; 125:S53-72. [PMID: 19932918 DOI: 10.1016/j.jaci.2009.07.008] [Citation(s) in RCA: 296] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 07/09/2009] [Accepted: 07/10/2009] [Indexed: 02/07/2023]
Abstract
Cytokines and chemokines are secreted proteins involved in numerous aspects of cell growth, differentiation, and activation. A prominent feature of these molecules is their effect on the immune system with regard to cell trafficking and development of immune tissue and organs. The nature of an immune response determines which cytokines are produced and ultimately whether the response is cytotoxic, humoral, cell mediated, or allergic. For this chapter, cytokines are grouped according to those that are predominantly antigen-presenting cell or T lymphocyte derived; that mediate cytotoxic, humoral, cell mediated, and allergic immunity; or that are immunosuppressive. A discussion of chemokine function and their role in cell trafficking and disease follows.
Collapse
|
33
|
Fuson KL, Zheng M, Craxton M, Pataer A, Ramesh R, Chada S, Sutton RB. Structural mapping of post-translational modifications in human interleukin-24: role of N-linked glycosylation and disulfide bonds in secretion and activity. J Biol Chem 2009; 284:30526-33. [PMID: 19734147 DOI: 10.1074/jbc.m109.036061] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human interleukin-24 (IL-24) is unique among the IL-10 superfamily as there is considerable evidence that it possesses multiple anti-cancer properties, including direct tumor cell cytotoxicity, helper T cell (TH1) immune stimulation, and anti-angiogenic activities. The primary sequence of human IL-24 differs from homologous cytokines, because it possesses three consensus N-linked glycosylation sites and the potential for a single disulfide bond. To address the significance of these modifications in human IL-24, we analyzed the relationship between post-translational modifications and the cytokine activity of the human IL-24 protein. In contrast to related interleukins, we identified a relationship between net glycosylation, protein solubility, and cytokine activity. In addition, abrogation of the two cysteine residues by mutagenesis dramatically altered the ability of IL-24 to secrete from host cells and resulted in the concomitant loss of IL-24 activity. We conclude that, unlike other IL-10 family members, human IL-24 must be glycosylated to maintain solubility and bioavailability. Further, a single, unique disulfide bond is required for secretion and activity. These structure-function relationships show that, although IL-24 is a member of the IL-19 subfamily of IL-10-like cytokines by sequence similarity, its surface properties and its distinctive disulfide arrangement make it unique. These observations could explain the novel biological activities measured of this cytokine. Understanding the structural basis of IL-24 activity will be important in the interpretation of the function of this cytokine and in the development of scale-up strategies for biophysical and clinical applications.
Collapse
Affiliation(s)
- Kerry L Fuson
- Department of Cell Physiology and Molecular Biophysics, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Kreis S, Philippidou D, Margue C, Behrmann I. IL-24: a classic cytokine and/or a potential cure for cancer? J Cell Mol Med 2009; 12:2505-10. [PMID: 18505472 PMCID: PMC4514127 DOI: 10.1111/j.1582-4934.2008.00372.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
IL-24, a member of the IL-10 family of cytokines, is produced by monocytes and Th2 cells. Interestingly, immune cells do not appear to express specific IL-24 receptor chains (IL-20R1/IL-20R2 and IL-22R/IL-20R2), it is therefore unlikely that IL-24 has classical immune-modulating properties. Skin, on the other hand, seems to represent a major target tissue for IL-24 and related cytokines such as IL-19, -20, and -22. However, the initial interest in IL-24 did not arise from its physiological signalling properties through its cognate receptors but rather because of its tentative ability to selectively kill different cancer cells. In an attempt to further investigate the signalling events underlying the IL-24-induced cancer cell death, we found that melanoma cell lines did not react in the expected and previously described way. Using several different forms and delivery modes of IL-24, we were unable to detect any apoptosis-inducing properties of this cytokine in melanoma cells. In the present ‘Point of view’ we will briefly summarizse these findings and put them in context of published reports stating that IL-24 might be a long sought after treatment for several types of cancer.
Collapse
Affiliation(s)
- S Kreis
- Life Sciences Research Unit, University of Luxembourg, Luxembourg.
| | | | | | | |
Collapse
|
35
|
Xuan W, Li YJ, Liu G, Ben-David Y, Archer MC. Interleukin-24 induces expression of beta4 integrin but suppresses anchorage-independent growth of rat mammary tumor cells by a mechanism that is independent of beta4. Mol Cancer Res 2009; 7:433-42. [PMID: 19258414 DOI: 10.1158/1541-7786.mcr-08-0252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Wistar-Furth rats develop multiple mammary adenocarcinomas following initiation with methylnitrosourea, whereas Copenhagen rats are resistant to the development of mammary tumors. We have previously isolated cell lines from tumors induced in resistant Copenhagen x Wistar-Furth F(1) rats by infusion of a retrovirus harboring v-Ha-ras directly into the main mammary ducts. Some of the cell lines were able to grow in soft agar, but a significant number did not display anchorage-independent growth. Here, we compared by microarray analysis genes that are differentially expressed in these cell lines. The expression of interleukin-24 (IL-24) and beta(4) integrin was highly correlated with the inability of cells to grow in soft agar. Ectopic expression of IL-24 in anchorage-independent cells inhibited their growth in monolayer culture, in soft agar, and in nude mice in vivo and inhibited their ability to migrate and invade in in vitro assays. Furthermore, growth suppression by IL-24 was associated with the transcriptional up-regulation of p27(Kip1) via the activation of Stat3. We showed, for the first time, that beta(4) integrin is a downstream target of IL-24. However, beta(4) does not play a direct role in regulating the proliferative capacity of rat mammary tumor cells. Our results show that IL-24 suppresses the growth of rat mammary carcinoma cells and may play a role in the resistance of Copenhagen rats to mammary carcinogenesis.
Collapse
Affiliation(s)
- Wanli Xuan
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
36
|
Zheng M, Bocangel D, Ramesh R, Ekmekcioglu S, Poindexter N, Grimm EA, Chada S. Interleukin-24 overcomes temozolomide resistance and enhances cell death by down-regulation of O6-methylguanine-DNA methyltransferase in human melanoma cells. Mol Cancer Ther 2008; 7:3842-51. [PMID: 19056673 PMCID: PMC2653264 DOI: 10.1158/1535-7163.mct-08-0516] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Melanoma is the most malignant of skin cancers, highly resistant to chemotherapy and radiotherapy. Temozolomide, a promising new derivative of dacarbazine, is currently being tested for treatment of metastatic melanoma. Resistance to alkylating agents such as temozolomide correlates with increased expression of DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT). Interleukin-24 (IL-24; mda-7) is a tumor suppressor cytokine that selectively inhibits tumor cell growth by inducing apoptosis and cell cycle arrest in melanoma cell lines and solid tumors. This tumor-selective activity has been observed in multiple preclinical animal models and in clinical trials. In this study, we analyzed the ability of Ad-IL-24 and its protein product, IL-24, to overcome temozolomide resistance in human melanoma cells. We have shown that Ad-IL-24 via exogenous IL-24 protein induces combinatorial synergy of temozolomide-induced cell killing in temozolomide-resistant melanoma cells by inhibition of MGMT. Neutralizing antibodies against IL-24 or its receptors significantly blocked the apoptotic activity of IL-24 + MGMT treatment. We show that accumulation of functional p53 is essential for IL-24-induced down-regulation of MGMT. Using either MGMT small interfering RNA, p53 small interfering RNA, or a p53 dominant-negative mutant to block MGMT protein expression resulted in increased sensitization to temozolomide. However, MGMT blockade in combination with IL-24 + temozolomide resulted in loss of combinatorial synergy, indicating that MGMT expression is required for the reversal of temozolomide resistance in melanoma cells. This study shows that IL-24 can play a significant role in overcoming temozolomide resistance and that the clinical efficacy of temozolomide may be improved by using a biochemotherapy combination with IL-24.
Collapse
Affiliation(s)
- Mingzhong Zheng
- Introgen Therapeutics, 2250 Holcombe Boulevard, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Pan X, Sheng W, Zhu Q, Xie Y, Ye Z, Xiang J, Li D, Yang J. Inhibition of pancreatic carcinoma growth by adenovirus-mediated human interleukin-24 expression in animal model. Cancer Biother Radiopharm 2008; 23:425-34. [PMID: 18771346 DOI: 10.1089/cbr.2008.0461] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Interleukin-24 (IL-24) has been shown to be a tumor-suppressor gene and the protein product found to be constitutively expressed by melanocytes, nerve cells, and some primary melanomas. The potential effect of adenovirus (AdV)-mediated IL-24 gene therapy was explored on human pancreatic carcinoma by using a pancreatic carcinoma cell line, patu8988. A recombinant adenovirus, AdVGFP/IL-24, expressing the marker, green fluorescent protein (GFP), and the tumor-suppressor gene, IL-24, was constructed. AdVGFP/IL-24 treatment of pancreatic carcinoma cells in vitro significantly induced pancreatic carcinoma cell cytotoxicity and apoptosis, compared with AdVGFP without IL-24 expression. In nude mice bearing patu8988 tumors, intratumoral injections of AdVGFP/IL-24 significantly inhibited pancreatic carcinoma growth. In addition, the molecular mechanism of tumor suppression was elucidated by downregulating the expression of vascular endothelial growth factor, CD34, and Bcl-2, as well as inhibiting tumor angiogenesis. Therefore, AdVGFP/IL-24 has the potential to serve as a novel tool for pancreatic carcinoma gene therapy.
Collapse
Affiliation(s)
- Xinting Pan
- Department of General Surgery, First Hospital Affiliated to Soochow University, Suzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Effect of recombinant adenovirus vector mediated human interleukin-24 gene transfection on pancreatic carcinoma growth. Chin Med J (Engl) 2008. [DOI: 10.1097/00029330-200810020-00016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
39
|
Commins S, Steinke JW, Borish L. The extended IL-10 superfamily: IL-10, IL-19, IL-20, IL-22, IL-24, IL-26, IL-28, and IL-29. J Allergy Clin Immunol 2008; 121:1108-11. [PMID: 18405958 DOI: 10.1016/j.jaci.2008.02.026] [Citation(s) in RCA: 241] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 02/19/2008] [Accepted: 02/20/2008] [Indexed: 12/21/2022]
Abstract
Cytokines are involved in virtually every aspect of immunity and inflammation. A cascade of responses evolves after cytokine activation, although optimal function might ultimately involve several complementary cytokines. Understanding the function of individual cytokines is complicated because their role can vary depending on the cellular source, target, and phase of the immune response. In fact, numerous cytokines have both proinflammatory and anti-inflammatory potential, with the contrasting outcome observed being determined by the immune cells present and their state of responsiveness to the cytokine. These issues make the study of cytokine biology daunting, particularly so for IL-10 and IL-10-related genes. The IL-10 superfamily is highly pleiotropic. These genes are linked together through genetic similarity and intron-exon gene structure. Significant commonality exists not only through shared receptors but also through conserved signaling cascades. However, its members mediate diverse activities, including immune suppression, enhanced antibacterial and antiviral immunity, antitumor activity, and promotion of self-tolerance in autoimmune diseases.
Collapse
Affiliation(s)
- Scott Commins
- Asthma and Allergic Disease Center, Beirne Carter Center for Immunology Research, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
40
|
Tessema M, Willink R, Do K, Yu YY, Yu W, Machida EO, Brock M, Van Neste L, Stidley CA, Baylin SB, Belinsky SA. Promoter Methylation of Genes in and around the Candidate Lung Cancer Susceptibility Locus6q23-25. Cancer Res 2008; 68:1707-14. [DOI: 10.1158/0008-5472.can-07-6325] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
Recombinant interleukin-24 lacks apoptosis-inducing properties in melanoma cells. PLoS One 2007; 2:e1300. [PMID: 18074024 PMCID: PMC2110900 DOI: 10.1371/journal.pone.0001300] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Accepted: 11/13/2007] [Indexed: 11/18/2022] Open
Abstract
IL-24, also known as melanoma differentiation antigen 7 (mda-7), is a member of the IL-10 family of cytokines and is mainly produced by Th(2) cells as well as by activated monocytes. Binding of IL-24 to either of its two possible heterodimeric receptors IL-20R1/IL-20R2 and IL-22R/IL-20R2 activates STAT3 and/or STAT1 in target tissues such as lung, testis, ovary, keratinocytes and skin. To date, the physiological properties of IL-24 are still not well understood but available data suggest that IL-24 affects epidermal functions by increasing proliferation of dermal cells. In stark contrast to its "normal" and physiological behaviour, IL-24 has been reported to selectively and efficiently kill a vast variety of cancer cells, especially melanoma cells, independent of receptor expression and Jak-STAT signalling. These intriguing properties have led to the development of adenovirally-expressed IL-24, which is currently being evaluated in clinical trials. Using three different methods, we have analysed a large panel of melanoma cell lines with respect to IL-24 and IL-24 receptor expression and found that none of the investigated cell lines expressed sufficient amounts of functional receptor pairs and therefore did not react to IL-24 stimulation with Jak/STAT activation. Results for three cell lines contrasted with previous studies, which reported presence of IL-24 receptors and activation of STAT3 following IL-24 stimulation. Furthermore, evaluating four different sources and modes of IL-24 administration (commercial recombinant IL-24, bacterially expressed GST-IL-24 fusion protein, IL-24 produced from transfected Hek cells, transiently over-expressed IL-24) no induction or increase in cell death was detected when compared to appropriate control treatments. Thus, we conclude that the cytokine IL-24 itself has no cancer-specific apoptosis-inducing properties in melanoma cells.
Collapse
|
42
|
Inoue S, Hartman A, Branch CD, Bucana CD, Bekele BN, Stephens LC, Chada S, Ramesh R. mda-7 In combination with bevacizumab treatment produces a synergistic and complete inhibitory effect on lung tumor xenograft. Mol Ther 2007; 15:287-94. [PMID: 17235306 DOI: 10.1038/sj.mt.6300035] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Bevacizumab, a humanized monoclonal antibody against vascular endothelial growth factor (VEGF), has shown antitumor activity by inhibiting tumor angiogenesis in preclinical and clinical studies. However, bevacizumab monotherapy does not induce complete tumor regression. Therefore, additional treatments must be combined with bevacizumab to promote tumor regression. We previously showed that melanoma differentiation associated gene-7 (mda-7) protein exerts potent antitumor and antiangiogenic activity. Thus, in this study, we investigated the therapeutic effects of mda-7 in combination with bevacizumab using lung cancer as a model. In vitro, treatment of human umbilical vein endothelial cells with conditioned medium from Ad-mda7 plus bevacizumab-treated lung tumor cells showed reduced VEGF ligand-receptor binding, and decreased cell survival, resulting in growth arrest and apoptosis. In vivo, treatment of subcutaneous lung tumor xenografts with bevacizumab plus Ad-mda7 resulted in significant tumor growth inhibition and improved survival compared to tumor growth in control mice. Furthermore, tumors in all the Ad-mda7 plus bevacizumab-treated mice completely regressed, and these were tumor free through the study's end. Molecular analysis showed enhanced tumor cell apoptosis and reduced VEGF and CD31 expression in Ad-mda7 plus bevacizumab-treated tumors. Thus, Ad-mda7 and bevacizumab treatment produces a synergistic and complete therapeutic effect against human lung cancer.
Collapse
Affiliation(s)
- Satoshi Inoue
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Tahara I, Miyake K, Hanawa H, Kurai T, Hirai Y, Ishizaki M, Uchida E, Tajiri T, Shimada T. Systemic cancer gene therapy using adeno-associated virus type 1 vector expressing MDA-7/IL24. Mol Ther 2007; 15:1805-11. [PMID: 17551500 DOI: 10.1038/sj.mt.6300225] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Melanoma differentiation-associated gene-7/interleukin-24 (mda-7/IL24), selectively induces apoptosis in cancer cells without harming normal cells. It also exerts immunomodulatory and antiangiogenic effects, as well as potent antitumor bystander effects, making it an ideal candidate for a new anticancer gene therapy. Here, we examined the feasibility of adeno-associated virus type 1 (AAV1) vector-mediated systemic gene therapy using mda-7/IL24. In vitro studies showed that medium conditioned by AAV1-mda7-transducedC2C12 cells induces tumor cell-specific apoptosis and inhibits angiogenesis in a human umbilical vein endothelial cell tube formation assay. To assess the in vivo effects of AAV1-mediated systemic delivery of MDA-7/IL24, we generated a subcutaneous tumor model by injecting Ehrlich ascites tumor cells into the dorsum of DDY mice. A single intravenous injection of AAV1-mda7 (2.0 x 10(11) viral genomes) significantly inhibited tumor growth. In addition, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), and immunohistochemical analyses showed significant induction of tumor-cell-specific apoptosis and reduction of microvessel formation within the tumors, and there was a significant increase in survival among the AAV1-mda7-treated mice. These results clearly demonstrate that continuous systemic delivery of MDA-7/IL24 can serve as an effective treatment for cancer. Thus, AAV1 vector-mediated systemic delivery of MDA-7/IL24 represents a potentially important new approach to anticancer therapy.
Collapse
Affiliation(s)
- Ichiro Tahara
- Department of Biochemistry and Molecular Biology, Division of Gene Therapy Research, Center for Advanced Medical Technology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Shanker M, Gopalan B, Patel S, Bocangel D, Chada S, Ramesh R. Vitamin E succinate in combination with mda-7 results in enhanced human ovarian tumor cell killing through modulation of extrinsic and intrinsic apoptotic pathways. Cancer Lett 2007; 254:217-26. [PMID: 17449172 DOI: 10.1016/j.canlet.2007.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 03/06/2007] [Accepted: 03/07/2007] [Indexed: 10/23/2022]
Abstract
Adenovirus-mediated mda-7 (Ad-mda7) gene transfer has been shown to induce apoptosis in various human cancer cells while sparing normal cells. Vitamin E succinate (VES) is also known to exhibit antitumor activity against a number of human cancer cell lines. We hypothesized that a combination of the two agents would produce an enhanced antitumor effect in MDAH2774 human ovarian cancer cells. Treatment of MDAH2774 cells with Ad-mda7 plus VES resulted in enhanced antitumor activity that involved the activation of two apoptotic pathways. Activation of the extrinsic pathway was demonstrated by increased cell-surface Fas expression and cleavage of Bid and caspase-8. Activation of the intrinsic pathway was demonstrated by disruption of mitochondrial potential; and activation of downstream capase-9 and caspase-3 via cytochrome C release. In contrast, the combination of Ad-mda7 plus VES did not show any antitumor activity against normal fibroblasts, indicating selective tumor cell killing. Our in vitro results provide a basis for further preclinical testing of Ad-mda7 plus VES as a potential cancer treatment strategy.
Collapse
Affiliation(s)
- Manish Shanker
- Department of Thoracic and Cardiovascular Surgery/Unit 445, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|