1
|
Zhang L, He J, Yu X, Zhang D. Prognostic Factors in Pediatric Alveolar Rhabdomyosarcoma: SEER Analysis of 277 Cases. Clin Pediatr (Phila) 2024; 63:1371-1378. [PMID: 38153032 DOI: 10.1177/00099228231220236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Alveolar rhabdomyosarcoma (ARMS) is a rare but highly aggressive cancer predominantly affecting children and adolescents. This study explores prognostic factors for pediatric and adolescent ARMS, using the Surveillance, Epidemiology, and End Results (SEER) database. Leveraging SEER data (2000-2019), we analyzed 277 cases. Employing Kaplan-Meier survival analysis and Cox proportional hazards models, we identified significant prognostic factors. Gender distribution was nearly equal (56.0% boys, 44.0% girls), with the majority (70.8%) from the white ethnic group. Primary tumors were predominantly in extremities (37.2%). Distant metastases significantly increased mortality risk (hazard ratio [HR], 3.13; 95% CI: 2.14-4.58) and regional lymph node involvement raised mortality risk (HR, 1.36; 95% CI: 0.96-1.92). Chemotherapy-only treatment had higher mortality risk than chemoradiotherapy (HR, 1.16; 95% CI: 0.97-2.67). Conclusively, our study identifies distant metastases, regional lymph node involvement, and treatment modality as crucial predictors of overall survival in pediatric ARMS.
Collapse
Affiliation(s)
- Li Zhang
- Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jiali He
- Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xianhai Yu
- Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Deying Zhang
- Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
| |
Collapse
|
2
|
Xu J, Li C, Kang X. The epigenetic regulatory effect of histone acetylation and deacetylation on skeletal muscle metabolism-a review. Front Physiol 2023; 14:1267456. [PMID: 38148899 PMCID: PMC10749939 DOI: 10.3389/fphys.2023.1267456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/24/2023] [Indexed: 12/28/2023] Open
Abstract
Skeletal muscles, the largest organ responsible for energy metabolism in most mammals, play a vital role in maintaining the body's homeostasis. Epigenetic modification, specifically histone acetylation, serves as a crucial regulatory mechanism influencing the physiological processes and metabolic patterns within skeletal muscle metabolism. The intricate process of histone acetylation modification involves coordinated control of histone acetyltransferase and deacetylase levels, dynamically modulating histone acetylation levels, and precisely regulating the expression of genes associated with skeletal muscle metabolism. Consequently, this comprehensive review aims to elucidate the epigenetic regulatory impact of histone acetylation modification on skeletal muscle metabolism, providing invaluable insights into the intricate molecular mechanisms governing epigenetic modifications in skeletal muscle metabolism.
Collapse
Affiliation(s)
| | | | - Xiaolong Kang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| |
Collapse
|
3
|
HDACs and the epigenetic plasticity of cancer cells: Target the complexity. Pharmacol Ther 2022; 238:108190. [PMID: 35430294 DOI: 10.1016/j.pharmthera.2022.108190] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022]
Abstract
Cancer cells must adapt to the hostile conditions of the microenvironment in terms of nutrition, space, and immune system attack. Mutations of DNA are the drivers of the tumorigenic process, but mutations must be able to hijack cellular functions to sustain the spread of mutant genomes. Transcriptional control is a key function in this context and is controlled by the rearrangement of the epigenome. Unlike genomic mutations, the epigenome of cancer cells can in principle be reversed. The discovery of the first epigenetic drugs triggered a contaminating enthusiasm. Unfortunately, the complexity of the epigenetic machinery has frustrated this enthusiasm. To develop efficient patient-oriented epigenetic therapies, we need to better understand the nature of this complexity. In this review, we will discuss recent advances in understanding the contribution of HDACs to the maintenance of the transformed state and the rational for their selective targeting.
Collapse
|
4
|
Mohankumar K, Shrestha R, Safe S. Nuclear receptor 4A1 (NR4A1) antagonists target paraspeckle component 1 (PSPC1) in cancer cells. Mol Carcinog 2022; 61:73-84. [PMID: 34699643 PMCID: PMC8665050 DOI: 10.1002/mc.23362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 01/03/2023]
Abstract
Paraspeckles compound 1 (PSPC1) is a multifunctional protein that plays an important role in cancer cells, where PSPC1 is a master regulator of pro-oncogenic responses that includes activation of TGFβ (TGFβ1), TGFβ-dependent EMT, and metastasis. The pro-oncogenic activities of PSPC1 closely resembled those observed for the orphan nuclear receptor 4A1 (NR4A1, Nur77) and knockdown of NR4A1 decreased expression of PSPC1 in MDA-MB-231 breast, H1299 lung, and SNU449 liver cancer cells. Similar results were observed in these same cell lines after treatment with bisindole-derived (CDIMs) NR4A1 antagonists. Moreover, PSPC1-dependent regulation of TGFβ, genes associated with cancer stem cells and epithelial to mesenchymal transition (EMT) were also downregulated after NR4A1 silencing or treatment of breast, lung, and liver cancer cells with CDIM/NR4A1 antagonists. Results of chromatin immunoprecipitation (ChIP) assays suggest that NR4A1 regulates PSPC1 through interaction with an NBRE sequence in the PSPC1 gene promoter. These results coupled with in vivo studies showing that NR4A1 antagonists inhibit breast tumor growth and downregulate PSPC1 in tumors indicate that the pro-oncogenic nuclear PSPC1 factor can be targeted by CDIM/NR4A1 antagonists.
Collapse
Affiliation(s)
- Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843 USA
| | - Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA, 77843
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843 USA
| |
Collapse
|
5
|
Shrestha R, Mohankumar K, Martin G, Hailemariam A, Lee SO, Jin UH, Burghardt R, Safe S. Flavonoids kaempferol and quercetin are nuclear receptor 4A1 (NR4A1, Nur77) ligands and inhibit rhabdomyosarcoma cell and tumor growth. J Exp Clin Cancer Res 2021; 40:392. [PMID: 34906197 PMCID: PMC8670039 DOI: 10.1186/s13046-021-02199-9] [Citation(s) in RCA: 12] [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: 06/22/2021] [Accepted: 11/26/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Flavonoids exhibit both chemopreventive and chemotherapeutic activity for multiple tumor types, however, their mechanisms of action are not well defined. Based on some of their functional and gene modifying activities as anticancer agents, we hypothesized that kaempferol and quercetin were nuclear receptor 4A1 (NR4A1, Nur77) ligands and confirmed that both compounds directly bound NR4A1 with KD values of 3.1 and 0.93 μM, respectively. METHODS The activities of kaempferol and quercetin were determined in direct binding to NR4A1 protein and in NR4A1-dependent transactivation assays in Rh30 and Rh41 rhabdomyosarcoma (RMS) cells. Flavonoid-dependent effects as inhibitors of cell growth, survival and invasion were determined in XTT and Boyden chamber assays respectively and changes in protein levels were determined by western blots. Tumor growth inhibition studies were carried out in athymic nude mice bearing Rh30 cells as xenografts. RESULTS Kaempferol and quercetin bind NR4A1 protein and inhibit NR4A1-dependent transactivation in RMS cells. NR4A1 also regulates RMS cell growth, survival, mTOR signaling and invasion. The pro-oncogenic PAX3-FOXO1 and G9a genes are also regulated by NR4A1 and, these pathways and genes are all inhibited by kaempferol and quercetin. Moreover, at a dose of 50 mg/kg/d kaempferol and quercetin inhibited tumor growth in an athymic nude mouse xenograft model bearing Rh30 cells. CONCLUSION These results demonstrate the clinical potential for repurposing kaempferol and quercetin for clinical applications as precision medicine for treating RMS patients that express NR4A1 in order to increase the efficacy and decrease dosages of currently used cytotoxic drugs.
Collapse
Affiliation(s)
- Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Greg Martin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Amanuel Hailemariam
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Syng-Ook Lee
- Department of Food Science and Technology, Keimyung University, Daegu, 42601, Republic of Korea
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA
| | - Robert Burghardt
- Department of Veterinary Integrated Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX, 77843-4466, USA.
| |
Collapse
|
6
|
Gossel LDH, Heim C, Pfeffermann LM, Moser LM, Bönig HB, Klingebiel TE, Bader P, Wels WS, Merker M, Rettinger E. Retargeting of NK-92 Cells against High-Risk Rhabdomyosarcomas by Means of an ERBB2 (HER2/Neu)-Specific Chimeric Antigen Receptor. Cancers (Basel) 2021; 13:cancers13061443. [PMID: 33809981 PMCID: PMC8004684 DOI: 10.3390/cancers13061443] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/14/2021] [Accepted: 03/18/2021] [Indexed: 12/11/2022] Open
Abstract
The dismal prognosis of pediatric and young adult patients with high-risk rhabdomyosarcoma (RMS) underscores the need for novel treatment options for this patient group. In previous studies, the tumor-associated surface antigen ERBB2 (HER2/neu) was identified as targetable in high-risk RMS. As a proof of concept, in this study, a novel treatment approach against RMS tumors using a genetically modified natural killer (NK)-92 cell line (NK-92/5.28.z) as an off-the-shelf ERBB2-chimeric antigen receptor (CAR)-engineered cell product was preclinically explored. In cytotoxicity assays, NK-92/5.28.z cells specifically recognized and efficiently eliminated RMS cell suspensions, tumor cell monolayers, and 3D tumor spheroids via the ERBB2-CAR even at effector-to-target ratios as low as 1:1. In contrast to unmodified parental NK-92 cells, which failed to lyse RMS cells, NK-92/5.28.z cells proliferated and became further activated through contact with ERBB2-positive tumor cells. Furthermore, high amounts of effector molecules, such as proinflammatory and antitumoral cytokines, were found in cocultures of NK-92/5.28.z cells with tumor cells. Taken together, our data suggest the enormous potential of this approach for improving the immunotherapy of treatment-resistant tumors, revealing the dual role of NK-92/5.28.z cells as CAR-targeted killers and modulators of endogenous adaptive immunity even in the inhibitory tumor microenvironment of high-risk RMS.
Collapse
Affiliation(s)
- Leonie D. H. Gossel
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (L.D.H.G.); (C.H.); (L.M.M.); (P.B.); (M.M.)
| | - Catrin Heim
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (L.D.H.G.); (C.H.); (L.M.M.); (P.B.); (M.M.)
| | - Lisa-Marie Pfeffermann
- Department of Cellular Therapeutics/Cell Processing, Institute for Transfusion Medicine and Immunohematology Frankfurt am Main, Goethe University Medical School, 60528 Frankfurt am Main, Germany; (L.-M.P.); (H.B.B.)
| | - Laura M. Moser
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (L.D.H.G.); (C.H.); (L.M.M.); (P.B.); (M.M.)
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany; (T.E.K.); (W.S.W.)
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), 60590 Frankfurt am Main, Germany
| | - Halvard B. Bönig
- Department of Cellular Therapeutics/Cell Processing, Institute for Transfusion Medicine and Immunohematology Frankfurt am Main, Goethe University Medical School, 60528 Frankfurt am Main, Germany; (L.-M.P.); (H.B.B.)
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98198-7720, USA
| | - Thomas E. Klingebiel
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany; (T.E.K.); (W.S.W.)
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), 60590 Frankfurt am Main, Germany
- Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany
| | - Peter Bader
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (L.D.H.G.); (C.H.); (L.M.M.); (P.B.); (M.M.)
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany; (T.E.K.); (W.S.W.)
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), 60590 Frankfurt am Main, Germany
| | - Winfried S. Wels
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany; (T.E.K.); (W.S.W.)
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), 60590 Frankfurt am Main, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt am Main, Germany
| | - Michael Merker
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (L.D.H.G.); (C.H.); (L.M.M.); (P.B.); (M.M.)
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany; (T.E.K.); (W.S.W.)
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), 60590 Frankfurt am Main, Germany
| | - Eva Rettinger
- Department for Children and Adolescents, Division for Stem Cell Transplantation, Immunology and Intensive Care Medicine, University Hospital Frankfurt, Goethe University, 60590 Frankfurt am Main, Germany; (L.D.H.G.); (C.H.); (L.M.M.); (P.B.); (M.M.)
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany; (T.E.K.); (W.S.W.)
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), 60590 Frankfurt am Main, Germany
- Correspondence: ; Tel.: +49-(0)69-6301-80631; Fax: +49-(0)69-6301-4202
| |
Collapse
|
7
|
Liu J, Liu Z, Li W, Zhang S. SOCS2 is a potential prognostic marker that suppresses the viability of hepatocellular carcinoma cells. Oncol Lett 2021; 21:399. [PMID: 33777222 PMCID: PMC7988697 DOI: 10.3892/ol.2021.12660] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/19/2021] [Indexed: 01/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-associated mortality worldwide. Thus, there is an urgent requirement to identify novel diagnostic and prognostic biomarkers for this disease. The present study aimed to identify the hub genes associated with the progression and prognosis of patients with HCC. A total of three expression profiles of HCC tissues were extracted from the Gene Expression Omnibus (GEO) database, followed by the identification of differentially expressed genes (DEGs) using the GEO2R method. The identified DEGs were assessed for survival significance using Kaplan-Meier analysis. Among the 15 identified DEGs in HCC tissues [cytochrome P450 family 39 subfamily A member 1, cysteine rich angiogenic inducer 61, Fos proto-oncogene, forkhead transcription factor 1 (FOXO1), growth arrest and DNA damage inducible β, Inhibitor of DNA binding 1, interleukin-1 receptor accessory protein, metallothionein-1M, pleckstrin homology-like domain family A member 1, Rho family GTPase 3, serine dehydratase, suppressor of cytokine signaling 2 (SOCS2), tyrosine aminotransferase (TAT), S100 calcium-binding protein P and serine protease inhibitor Kazal-type 1 (SPINK1)]. Low expression levels of FOXO1, SOCS2 and TAT and high SPINK1 expression indicated poor survival outcomes for patients with HCC. In addition, SOCS2 was associated with distinct stages of HCC progression in patients and presented optimal diagnostic value. In vitro functional experiments indicated that overexpression of SOCS2 inhibited HCC cell proliferation and migration. Taken together, the results of the present study suggest that SOCS2 may act as a valuable prognostic marker that is closely associated with HCC progression.
Collapse
Affiliation(s)
- Jiankun Liu
- Department of Gastroenterology, 920th Hospital of The PLA Joint Logistics Support Force, Kunming, Yunnan 650032, P.R. China
| | - Zhiyong Liu
- Department of Gastroenterology, 920th Hospital of The PLA Joint Logistics Support Force, Kunming, Yunnan 650032, P.R. China.,Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Wei Li
- Department of General Surgery, Changzheng Hospital, Second Military Medical University, Shanghai 200003, P.R. China
| | - Shurong Zhang
- Department of Gastroenterology, 920th Hospital of The PLA Joint Logistics Support Force, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
8
|
Shrestha R, Mohankumar K, Jin UH, Martin G, Safe S. The Histone Methyltransferase Gene G9A Is Regulated by Nuclear Receptor 4A1 in Alveolar Rhabdomyosarcoma Cells. Mol Cancer Ther 2020; 20:612-622. [PMID: 33277444 DOI: 10.1158/1535-7163.mct-20-0474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/09/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
The histone methyltransferase G9A (EHMT2) gene catalyzes methylation of histone 3 lysine 9 (H3K9), and this gene silencing activity contributes to the tumor promoter-like activity of G9A in several tumor types including alveolar rhabdomyosarcoma (ARMS). Previous studies show the orphan nuclear receptor 4A1 (NR4A1, Nur77) is overexpressed in rhabdomyosarcoma and exhibits pro-oncogenic activity. In this study, we show that knockdown of NR4A1 in ARMS cells decreased expression of G9A mRNA and protein. Moreover, treatment of ARMS cells with several bis-indole-derived NR4A1 ligands (antagonists) including 1,1-bis(3'-indolyl)-1-(4-hydroxyphenyl)methane (CDIM8), 3,5-dimethyl (3,5-(CH3)2), and 3-bromo-5-methoxy (3-Br-5-OCH3) analogs also decreased G9A expression. Furthermore, NR4A1 antagonists also decreased G9A expression in breast, lung, liver, and endometrial cancer cells confirming that G9A is an NR4A1-regulated gene in ARMS and other cancer cell lines. Mechanistic studies showed that the NR4A1/Sp1 complex interacted with the GC-rich 511 region of the G9A promoter to regulate G9A gene expression. Moreover, knockdown of NR4A1 or treatment with NR4A1 receptor antagonists decreased overall H3K9me2, H3K9me2 associated with the PTEN promoter, and PTEN-regulated phospho-Akt. In vivo studies showed that the NR4A1 antagonist (3-Br-5-OCH3) inhibited tumor growth in athymic nude mice bearing Rh30 ARMS cells and confirmed that G9A was an NR4A1-regulated gene that can be targeted by NR4A1 receptor antagonists.
Collapse
Affiliation(s)
- Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Un-Ho Jin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Gregory Martin
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Stephen Safe
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas. .,Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| |
Collapse
|
9
|
Shrestha R, Mohankumar K, Safe S. Bis-indole derived nuclear receptor 4A1 (NR4A1) antagonists inhibit TGFβ-induced invasion of embryonal rhabdomyosarcoma cells. Am J Cancer Res 2020; 10:2495-2509. [PMID: 32905449 PMCID: PMC7471359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/13/2020] [Indexed: 06/11/2023] Open
Abstract
Transforming growth factor β (TGFβ) enhances invasion of breast and lung cancer cells through phosphorylation-dependent nuclear export of the nuclear receptor 4A1 (NR4A1, Nur77). This response is inhibited by the NR4A1 antagonist 1,1-bis(3'-indoly)-1-(p-hydroxyphenyl) methane (CDIM8) and we hypothesized that similar effects would be observed in Rhabdomyosarcoma (RMS) cells. Although some kinase inhibitors block TGFβ-induced invasion of embryonal RMS (ERMS) cells, the mechanism differs from breast and lung cancer cells since NR4A1 is extranuclear in ERMS cells. However, CDIM8 blocks basal and TGFβ-induced invasion of RD and SMS-CTR ERMS cell lines but not Rh30 alveolar RMS (ARMS) cells. Moreover, this response in ERMS cells was independent of SMAD7 degradation or activation of SMAD2/SMAD3. β-Catenin silencing decreased ERMS cell invasion and CDIM8 induced proteasome-independent downregulation of β-catenin. The novel mechanism of CDIM8-mediated inhibition of basal and TGFβ-induced ERMS cell invasion was due to activation of the Bcl-2-NR4A1 complex, mitochondrial disruption, induction of the tumor suppressor-like cytokine interleukin-24 (IL-24) which in turn downregulates β-catenin expression. Thus, the NR4A1 antagonist inhibits TGFβ-induced invasion of ERMS cells through initial targeting of cytosolic NR4A1.
Collapse
Affiliation(s)
- Rupesh Shrestha
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege Station, TX 77843, USA
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M UniversityCollege Station, TX 77843, USA
| | - Stephen Safe
- Department of Biochemistry and Biophysics, Texas A&M UniversityCollege Station, TX 77843, USA
- Department of Veterinary Physiology and Pharmacology, Texas A&M UniversityCollege Station, TX 77843, USA
| |
Collapse
|
10
|
Mohankumar K, Li X, Sung N, Cho YJ, Han SJ, Safe S. Bis-Indole-Derived Nuclear Receptor 4A1 (NR4A1, Nur77) Ligands as Inhibitors of Endometriosis. Endocrinology 2020; 161:5758064. [PMID: 32099996 PMCID: PMC7105386 DOI: 10.1210/endocr/bqaa027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023]
Abstract
Endometriosis is an inflammatory disease that primarily affects women during their reproductive years, and since current hormonal therapies are of concern, new hormone-independent treatment regimens are needed. The orphan nuclear receptor 4A1 (NR4A1, Nur77) is expressed in patient-derived (stromal) endometriotic cells and also epithelial cell lines, and we observed that knockdown of NR4A1 in patient-derived ectopic endometrium-isolated ovarian endometrioma (ESECT)-7 and ESECT-40 cells decreased cell proliferation and induced apoptosis. Moreover, the treatment of these cells with bis-indole derived NR4A1 ligands 1,1-bis(3'-indolyl)-1-(p-hydroxyphenyl)methane (DIM-C-pPhOH) and its buttressed 3-chloro-5-methoxy analog (DIM-C-pPhOH-3-Cl-5-OCH3) inhibited cell growth and induced apoptosis and related genes. The compounds exhibit NR4A1 antagonist activities in both functional and transactivation assays whereas these effects were not observed in normal endometrial cells. We also observed that NR4A1 knockdown and treatment with NR4A1 antagonists decreased fibrosis, α-smooth muscle actin, and related pro-fibrotic genes in ESECT-7 and ESECT-40 cells, and similar results were observed in epithelial-derived endometriotic cell lines. Moreover, in an endometriosis mouse model with auto-transplantation and also in severe combined immune deficiency mice transplanted with human endometriotic cells treatment with 25 mg/kg/day DIM-C-pPhOH-3-Cl-5-OCH3 significantly inhibited growth and expansion of endometriotic lesions. Thus, bis-indole-derived NR4A1 ligands represent a novel class of drugs as nonhormonal therapy for endometriosis.
Collapse
Affiliation(s)
- Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX US
| | - Xi Li
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX US
| | - Nuri Sung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX US
| | - Yeon Jean Cho
- Department of Obstetrics and Gynecology, Dong-A University, College of Medicine, Busan, Republic of Korea
| | - Sang Jun Han
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX US
- Correspondence: Stephen Safe, Department of Veterinary Physiology & Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466. E-mail: ; or Sang Jun Han, Department of molecular and Cell Biology, Baylor college of Medicine, Houston, TX 77030. E-mail:
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX US
- Correspondence: Stephen Safe, Department of Veterinary Physiology & Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466. E-mail: ; or Sang Jun Han, Department of molecular and Cell Biology, Baylor college of Medicine, Houston, TX 77030. E-mail:
| |
Collapse
|
11
|
Liao S, Yang Y, Chen S, Bi Y, Huang Q, Wei Z, Qin A, Liu B. IL-24 inhibits endometrial cancer cell proliferation by promoting apoptosis through the mitochondrial intrinsic signaling pathway. Biomed Pharmacother 2020; 124:109831. [PMID: 31972354 DOI: 10.1016/j.biopha.2020.109831] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/02/2020] [Accepted: 01/02/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Endometrial cancer is a type of malignant tumor of the female reproductive system. Preserving fertility in endometrial cancer patients is currently a formidable challenge. Interleukin-24 (IL-24) is a unique cytokine tumor suppressor gene belonging to the IL-10 cytokine family. IL-24 has broad-spectrum antitumor activity through different signaling pathways but does not affect normal cells. IL-24 gene therapy may provide a new method for the treatment of endometrial cancer. METHODS Transfection was used for gene transfer. The expression of IL-24 and related pathway proteins in endometrial cancer tissue and the Ishikawa cell line was detected by immunohistochemistry and Western blotting, respectively. The antitumor function of IL-24 was examined in vitro and in vivo. Cell proliferation was determined by CCK-8 assay, cell migration was shown by wound-healing assay, and cell invasion was detected by Transwell assay. Apoptosis was analyzed by TUNEL assay, and HE staining was performed to observe the morphology of the samples. RESULTS Immunohistochemical analysis showed different expression levels of IL-24 in human endometrial cancer tissues and normal endometrial tissues. IL-24 increased protein expression of BAX and Cytochrome C, while BCL-2, MMP-3, VEGF, Caspase-9 and Caspase-3 expression was decreased. Overexpression of IL-24 inhibited cell proliferation, migration and invasion, but increased cell apoptosis in endometrial cancer. Mechanistically, we demonstrated that IL-24 inhibited endometrial cancer cell growth by inducing cell apoptosis through the mitochondrial intrinsic signaling pathway. In addition, IL-24 inhibited tumor development by inducing cell apoptosis and inhibiting angiogenesis, as shown in xenograft tumor experiments. CONCLUSIONS Our study demonstrates the antitumor effect of IL-24 on endometrial cancer and shows that IL-24 may be a promising therapeutic gene for endometrial cancer gene therapy.
Collapse
Affiliation(s)
- Shengbin Liao
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Yihua Yang
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Saiqiong Chen
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Yin Bi
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Qiuyan Huang
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Zhiyao Wei
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China
| | - Aiping Qin
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China.
| | - Bo Liu
- Center of Reproductive Medicine, Guangxi Medical University First Affiliated Hospital, Nanning, Guangxi, China.
| |
Collapse
|
12
|
Hedrick E, Mohankumar K, Lacey A, Safe S. Inhibition of NR4A1 Promotes ROS Accumulation and IL24-Dependent Growth Arrest in Rhabdomyosarcoma. Mol Cancer Res 2019; 17:2221-2232. [PMID: 31462501 DOI: 10.1158/1541-7786.mcr-19-0408] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/16/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022]
Abstract
Nuclear receptor 4A1 (NR4A1, Nur77) is overexpressed in rhabdomyosarcoma (RMS), and inactivation of NR4A1 (siNR4A1) or treatment with the NR4A1 antagonist 1,1-bis(3'-indoly)-1-(p-hydroxy-phenyl)methane (DIM-C-pPhOH) has antiproliferative and proapoptotic effects on RMS cells. However, the mechanism by which NR4A1 inhibition exerts these effects is poorly defined. Here, we report that NR4A1 silencing or inhibition resulted in accumulation of reactive oxygen species (ROS) and ROS-dependent induction of the tumor suppressor-like cytokine IL24 in RMS cells. Mechanistically, NR4A1 was found to regulate the expression of the proreductant genes thioredoxin domain-containing 5 (TXNDC5) and isocitrate dehydrogenase 1 (IDH1), which are downregulated in RMS cells following NR4A1 knockdown or inhibition. Silencing TXNDC5 and IDH1 also induced ROS accumulation and IL24 expression in RMS cells, suggesting that NR4A1 antagonists mediate their antiproliferative and apoptotic effects through modulation of proreductant gene expression. Finally, cotreatment with the antioxidant glutathione or IL24-blocking antibody reversed the effects of NR4A1 inhibition, demonstrating the importance of both ROS and IL24 in mediating the cellular responses. IMPLICATIONS: Overall, these data elucidate the mechanism by which NR4A1 inhibition functions to inhibit the proliferation, survival, and migration of RMS cells.
Collapse
Affiliation(s)
- Erik Hedrick
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Kumaravel Mohankumar
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Alexandra Lacey
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas.
| |
Collapse
|
13
|
Nakano K, Takahashi S. Translocation-Related Sarcomas. Int J Mol Sci 2018; 19:ijms19123784. [PMID: 30487384 PMCID: PMC6320865 DOI: 10.3390/ijms19123784] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
Chromosomal translocations are observed in approximately 20% of soft tissue sarcomas (STS). With the advances in pathological examination technology, the identification of translocations has enabled precise diagnoses and classifications of STS, and it has been suggested that the presence of and differences in translocations could be prognostic factors in some translocation-related sarcomas. Most of the translocations in STS were not regarded as targets of molecular therapies until recently. However, trabectedin, an alkylating agent, has shown clinical benefits against translocation-related sarcoma based on a modulation of the transcription of the tumor's oncogenic fusion proteins. Many molecular-targeted drugs that are specific to translocations (e.g., anaplastic lymphoma kinase and tropomyosin kinase related fusion proteins) have emerged. The progress in gene technologies has allowed researchers to identify and even induce new translocations and fusion proteins, which might become targets of molecular-targeted therapies. In this review, we discuss the clinical significance of translocation-related sarcomas, including their diagnoses and targeted therapies.
Collapse
Affiliation(s)
- Kenji Nakano
- Department of Medical Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo 135-0063, Japan.
| | - Shunji Takahashi
- Department of Medical Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo 135-0063, Japan.
| |
Collapse
|