1
|
Jansson C, Mengelbier LH. Retinoic acid promotes differentiation of WiT49- but not of CCG99-11 Wilms tumour cells. Cancer Rep (Hoboken) 2023:e1819. [PMID: 37186071 DOI: 10.1002/cnr2.1819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/14/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Most children with Wilms tumour are successfully treated with multidrug chemotherapy and surgery. These treatments cause severe side effects for the patients, an issue that needs to be addressed by exploring other treatment options with less or no side effects. One option is to complement current therapies with agents that could potentially induce tumour cell differentiation, for example retinoic acid (RA). AIMS To facilitate quick assessment of an agent's effect on Wilms tumour differentiation by a rapid in vitro model system. METHODS AND RESULTS Here WiT49 and CCG99-11 Wilms tumour cells were treated with 10 μM RA for 72 h or 9 days. Cultured cells were scraped off from Petri dishes, pelleted and embedded in paraffin in the same way as clinical tumour specimens are preserved. Cell morphology and differentiation were evaluated by analyses of haematoxylin eosin (H&E) and immunohistochemical stainings. Based on H&E, WT1 and CKAE1/3 stainings, RA treatment induced further epithelial differentiation of WiT49 cells, whereas there was no sign of induced maturation in CCG99-11 cells. Ki67 staining showed that RA inhibited cell proliferation in both cell lines. CONCLUSIONS Our study shows that in vitro culturing of WiT49 and CCG99-11 cells, followed by pelleting and paraffin embedding of cell pellets, could aid in a quick evaluation of potential differentiating agents against Wilms tumour. In addition, our results strengthen previous results that retinoic acid could be a potential complement to regular Wilms tumour treatment.
Collapse
Affiliation(s)
- Caroline Jansson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Sweden
| | | |
Collapse
|
2
|
Zhao JY, Yuan XK, Luo RZ, Wang LX, Gu W, Yamane D, Feng H. Phospholipase A and acyltransferase 4/retinoic acid receptor responder 3 at the intersection of tumor suppression and pathogen restriction. Front Immunol 2023; 14:1107239. [PMID: 37063830 PMCID: PMC10102619 DOI: 10.3389/fimmu.2023.1107239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
Phospholipase A and acyltransferase (PLAAT) 4 is a class II tumor suppressor with phospholipid metabolizing abilities. It was characterized in late 2000s, and has since been referred to as 'tazarotene-induced gene 3' (TIG3) or 'retinoic acid receptor responder 3' (RARRES3) as a key downstream effector of retinoic acid signaling. Two decades of research have revealed the complexity of its function and regulatory roles in suppressing tumorigenesis. However, more recent findings have also identified PLAAT4 as a key anti-microbial effector enzyme acting downstream of interferon regulatory factor 1 (IRF1) and interferons (IFNs), favoring protection from virus and parasite infections. Unveiling the molecular mechanisms underlying its action may thus open new therapeutic avenues for the treatment of both cancer and infectious diseases. Herein, we aim to summarize a brief history of PLAAT4 discovery, its transcriptional regulation, and the potential mechanisms in tumor prevention and anti-pathogen defense, and discuss potential future directions of PLAAT4 research toward the development of therapeutic approaches targeting this enzyme with pleiotropic functions.
Collapse
Affiliation(s)
- Jian-Yong Zhao
- Hospital of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Cangzhou, Hebei, China
| | - Xiang-Kun Yuan
- Hospital of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Cangzhou, Hebei, China
| | - Rui-Zhen Luo
- Hospital of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Cangzhou, Hebei, China
| | - Li-Xin Wang
- Hospital of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Cangzhou, Hebei, China
| | - Wei Gu
- School of Medicine, Chongqing University, Chongqing, China
| | - Daisuke Yamane
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hui Feng
- School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
3
|
Wojcik HM, Lovvorn HN, Hollingshead M, Pierce J, Stotler H, Murphy AJ, Borgel S, Phelps HM, Correa H, Perantoni AO. Exploiting embryonic niche conditions to grow Wilms tumor blastema in culture. Front Oncol 2023; 13:1091274. [PMID: 37007076 PMCID: PMC10061139 DOI: 10.3389/fonc.2023.1091274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
IntroductionWilms Tumor (WT), or nephroblastoma, is the most common pediatric kidney cancer. Most WTs display a “favorable” triphasic histology, in which the tumor is comprised of blastemal, stromal, and epithelial cell types. Blastemal predominance after neoadjuvant chemotherapy or diffuse anaplasia (“unfavorable” histology; 5-8%) portend a worse prognosis. Blastema likely provide the putative cancer stem cells (CSCs), which retain molecular and histologic features characteristic of nephron progenitor cells (NPCs), within WTs. NPCs arise in the metanephric mesenchyme (MM) and populate the cap mesenchyme (CM) in the developing kidney. WT blastemal cells, like NPCs, similarly express markers, SIX2 and CITED1. Tumor xenotransplantation is currently the only dependable method to propagate tumor tissue for research or therapeutic screening, since efforts to culture tumors in vitro as monolayers have invariably failed. Therefore, a critical need exists to propagate WT stem cells rapidly and efficiently for high-throughput, real-time drug screening.MethodsPreviously, our lab developed niche conditions that support the propagation of murine NPCs in culture. Applying similar conditions to WTs, we assessed our ability to maintain key NPC "stemness" markers, SIX2, NCAM, and YAP1, and CSC marker ALDHI in cells from five distinct untreated patient tumors.ResultsAccordingly, our culture conditions maintained the expression of these markers in cultured WT cells through multiple passages of rapidly dividing cells.DiscussionThese findings suggest that our culture conditions sustain the WT blastemal population, as previously shown for normal NPCs. As a result, we have developed new WT cell lines and a multi-passage in vitro model for studying the blastemal lineage/CSCs in WTs. Furthermore, this system supports growth of heterogeneous WT cells, upon which potential drug therapies could be tested for efficacy and resistance.
Collapse
Affiliation(s)
- Heather M. Wojcik
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, United States
| | - Harold N. Lovvorn
- Department of Pediatric Surgery, Monroe Carell Jr. Children’s Hospital at Vanderbilt University, Nashville, TN, United States
| | - Melinda Hollingshead
- Biological Testing Branch/Developmental Therapeutics Program, National Cancer Institute, Frederick, MD, United States
| | - Janene Pierce
- Department of Pediatric Surgery, Monroe Carell Jr. Children’s Hospital at Vanderbilt University, Nashville, TN, United States
| | - Howard Stotler
- Leidos Biomedical Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Andrew J. Murphy
- Department of Pediatric Surgery, Monroe Carell Jr. Children’s Hospital at Vanderbilt University, Nashville, TN, United States
| | - Suzanne Borgel
- Leidos Biomedical Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Hannah M. Phelps
- Department of Pediatric Surgery, Monroe Carell Jr. Children’s Hospital at Vanderbilt University, Nashville, TN, United States
| | - Hernan Correa
- Division of Pediatric Pathology, Monroe Carell Jr. Children’s Hospital at Vanderbilt University, Nashville, TN, United States
| | - Alan O. Perantoni
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, United States
- *Correspondence: Alan O. Perantoni,
| |
Collapse
|
4
|
Histone H3K36me2 demethylase KDM2A promotes bladder cancer progression through epigenetically silencing RARRES3. Cell Death Dis 2022; 13:547. [PMID: 35697678 PMCID: PMC9192503 DOI: 10.1038/s41419-022-04983-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/22/2022] [Accepted: 05/26/2022] [Indexed: 01/21/2023]
Abstract
Epigenetic dysregulation contributes to bladder cancer tumorigenesis. H3K36me2 demethylase KDM2A functions as an important epigenetic regulator of cell fate in many types of tumors. However, its role in bladder cancer remains unknown. Here, we revealed a positive correlation between KDM2A gene copy number gain and upregulation of KDM2A mRNA expression in bladder cancer. Moreover, a super-enhancer (SE) driving KDM2A transcription was found in high-grade bladder cancer, resulting in a significantly higher expression of KDM2A mRNA compared to that in low-grade bladder tumors. KDM2A knockdown (KD) decreased the proliferation, invasion, and spheroid formation of high-grade bladder cancer cells and inhibited tumor growth in mouse xenograft models. Furthermore, we identified RARRES3 as a key KDM2A target gene. KDM2A suppresses RARRES3 expression via demethylation of H3K36me2 in the RARRES3 promoter. Intriguingly, RARRES3 KD attenuated the inhibitory effects of KDM2A depletion on the malignant phenotypes of high-grade bladder cancer cells. The combination of the KDM2A inhibitor IOX1 and the RARRES3 agonist all-trans retinoic acid (ATRA) synergistically inhibited the proliferation of high-grade bladder cancer cells, suggesting that the KDM2A/RARRES3 axis may be a promising therapeutic target for the treatment of high-grade bladder cancer.
Collapse
|
5
|
Yu K, Ji Y, Liu M, Shen F, Xiong X, Gu L, Lu T, Ye Y, Feng S, He J. High Expression of CKS2 Predicts Adverse Outcomes: A Potential Therapeutic Target for Glioma. Front Immunol 2022; 13:881453. [PMID: 35663965 PMCID: PMC9160311 DOI: 10.3389/fimmu.2022.881453] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclin-dependent kinase regulatory subunit 2 (CKS2) is a potential prognostic marker and is overexpressed in various cancers. This study analyzed sequencing and clinical data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus, with external validation using the Chinese Glioma Genome Atlas (CGGA) data. CKS2 expression in the normal brain and tumor tissue was compared. cBioPortal and MethSurv were utilized to scrutinize the prognostic value of CKS2 methylation. Gene set enrichment examination and single-sample gene set enrichment analysis were employed to explore the potential biological functions of CKS2. Cell viability, colony formation, and transwell assays were conducted to evaluate the influence of CKS2 on glioma cell proliferation and invasion. Compared with normal brain tissue, the expression of CKS2 was upregulated in glioma samples (p < 0.001). Multivariate data analysis from TCGA and CGGA indicated that increased expression of CKS2 was an independent risk factor for the prognosis of overall survival in glioma patients. CKS2 methylation was negatively associated with CKS2 expression. Patients with CKS2 hypomethylation had worse overall survival compared with patients with CKS2 methylation, as suggested by the analysis of both TCGA and CGGA datasets. The expression level of CKS2 is closely related to tumor immunity, including the correlation of tumor immune cell infiltration, immune score, and co-expression of multiple immune-related genes. In addition, CKS2 is associated with several immune checkpoints and responses to the chemotherapy drug cisplatin. CKS2 knockdown impeded the expansion and aggression of glioma cell lines. The changes in CKS2 expression may provide a novel prognostic biomarker that can be used to improve patient overall survival rates.
Collapse
Affiliation(s)
- Kai Yu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yulong Ji
- Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nangchang University, Nanchang, China
| | - Min Liu
- Department of Neurosurgery, Poyang County People’s Hospital, Shangrao, China
| | - Fugeng Shen
- Bone Traumatology Department, Shangli County Traditional Chinese Medicine Hospital, Pingxiang, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tianzhu Lu
- Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital of Nangchang University, Nanchang, China
| | - Yingze Ye
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shi Feng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jianying He
- Department of Orthopedic, JiangXi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
- *Correspondence: Jianying He,
| |
Collapse
|
6
|
Ehrlich PF, Tornwall B, Chintagumpala MM, Chi YY, Hoffer FA, Perlman EJ, Kalapurakal JA, Warwick A, Shamberger RC, Khanna G, Hamilton TE, Gow KW, Paulino AC, Gratias EJ, Mullen EA, Geller JI, Fernandez CV, Dome JS. Kidney Preservation and Wilms Tumor Development in Children with Diffuse Hyperplastic Perilobar Nephroblastomatosis: A Report from the Children's Oncology Group Study AREN0534. Ann Surg Oncol 2022; 29:3252-3261. [PMID: 35072864 PMCID: PMC9254258 DOI: 10.1245/s10434-021-11266-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/03/2021] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Diffuse hyperplastic perilobar nephroblastomatosis (DHPLN) represents a unique category of nephroblastomatosis. Treatment has ranged from observation to multiple regimens of chemotherapy. Wilms tumors (WTs) develop in 100% of untreated patients and between 32 and 52% of treated patients. Renal preservation rates have not been previously reported. An aim of the Children's Oncology Group (COG) study AREN0534 was to prospectively evaluate the efficacy of chemotherapy in preserving renal units and preventing WT development in children with DHPLN. METHODS Patients were enrolled through the COG protocol AREN03B2 with central radiological review. DHPLN was defined as the cortical surface of the kidney being composed of hyperplastic rests, with the entire nephrogenic zone involved, and with a thick rind capping all of one or both kidneys. Treatment was with vincristine and dactinomycin (regimen EE4A), with cross-sectional imaging at weeks 6 and 12. If the patient's disease was stable or decreasing, treatment was continued for 19 weeks. Renal preservation, WT development rates at 1 year, and overall survival (OS) are reported. RESULTS Nine patients were enrolled (five females and four males), with a median age at enrollment of 10.22 months (range 2.92-29.11). One patient who was enrolled was deemed unevaluable because they did not meet the radiological criteria for DHPLN, resulting in eight evaluable patients. These eight patients had DHPLN confirmed via radiological criteria (all bilateral). Initial chemotherapy was EE4A for all eight patients, with seven of eight patients starting chemotherapy without tissue diagnosis.One patient who had an upfront partial nephrectomy was found to have DHPLN in the specimen and was subsequently treated with EE4A. All patients remained alive, with a median follow-up of 6.6 years (range 4.5-9.1). No patients were anephric; 14 of 16 kidneys were functioning (87.5%). Six of eight patients (75%) did not have WT on therapy, but two of these patients relapsed within 6 months of stopping therapy; both had favorable histology WT. One patient who was diagnosed with WT on therapy relapsed at 12 months (one of eight [12.5%]) and developed anaplastic histology. CONCLUSIONS Chemotherapy for patients with DHPLN was effective in preserving kidney function. Five-year OS is excellent, however the ideal type and duration of chemotherapy to prevent WT development remains elusive.
Collapse
Affiliation(s)
- Peter F Ehrlich
- Section of Pediatric Surgery, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, MI, USA.
| | - Brett Tornwall
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | | | - Yueh-Yun Chi
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - Fredric A Hoffer
- Fred Hutchison Cancer Center, University of Washington, Seattle, WA, USA
| | | | | | - Anne Warwick
- Walter Reed National Military Medical Center, Washington, DC, USA
| | - Robert C Shamberger
- Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | | | - Thomas E Hamilton
- Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | | | | | | | - Elizabeth A Mullen
- Boston Children's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | | | | | - Jeffrey S Dome
- IWK Children's Hospital, Halifax, NS, Canada
- Children National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
7
|
Fu M, Peng D, Lan T, Wei Y, Wei X. Multifunctional regulatory protein connective tissue growth factor (CTGF): A potential therapeutic target for diverse diseases. Acta Pharm Sin B 2022; 12:1740-1760. [PMID: 35847511 PMCID: PMC9279711 DOI: 10.1016/j.apsb.2022.01.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Connective tissue growth factor (CTGF), a multifunctional protein of the CCN family, regulates cell proliferation, differentiation, adhesion, and a variety of other biological processes. It is involved in the disease-related pathways such as the Hippo pathway, p53 and nuclear factor kappa-B (NF-κB) pathways and thus contributes to the developments of inflammation, fibrosis, cancer and other diseases as a downstream effector. Therefore, CTGF might be a potential therapeutic target for treating various diseases. In recent years, the research on the potential of CTGF in the treatment of diseases has also been paid more attention. Several drugs targeting CTGF (monoclonal antibodies FG3149 and FG3019) are being assessed by clinical or preclinical trials and have shown promising outcomes. In this review, the cellular events regulated by CTGF, and the relationships between CTGF and pathogenesis of diseases are systematically summarized. In addition, we highlight the current researches, focusing on the preclinical and clinical trials concerned with CTGF as the therapeutic target.
Collapse
|
8
|
Zalesak-Kravec S, Huang W, Jones JW, Yu J, Alloush J, Defnet AE, Moise AR, Kane MA. Role of cellular retinol-binding protein, type 1 and retinoid homeostasis in the adult mouse heart: A multi-omic approach. FASEB J 2022; 36:e22242. [PMID: 35253263 DOI: 10.1096/fj.202100901rrr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022]
Abstract
The main active metabolite of Vitamin A, all-trans retinoic acid (RA), is required for proper cellular function and tissue organization. Heart development has a well-defined requirement for RA, but there is limited research on the role of RA in the adult heart. Homeostasis of RA includes regulation of membrane receptors, chaperones, enzymes, and nuclear receptors. Cellular retinol-binding protein, type 1 (CRBP1), encoded by retinol-binding protein, type 1 (Rbp1), regulates RA homeostasis by delivering vitamin A to enzymes for RA synthesis and protecting it from non-specific oxidation. In this work, a multi-omics approach was used to characterize the effect of CRBP1 loss using the Rbp1-/- mouse. Retinoid homeostasis was disrupted in Rbp1-/- mouse heart tissue, as seen by a 33% and 24% decrease in RA levels in the left and right ventricles, respectively, compared to wild-type mice (WT). To further inform on the effect of disrupted RA homeostasis, we conducted high-throughput targeted metabolomics. A total of 222 metabolite and metabolite combinations were analyzed, with 33 having differential abundance between Rbp1-/- and WT hearts. Additionally, we performed global proteome profiling to further characterize the impact of CRBP1 loss in adult mouse hearts. More than 2606 unique proteins were identified, with 340 proteins having differential expression between Rbp1-/- and WT hearts. Pathway analysis performed on metabolomic and proteomic data revealed pathways related to cellular metabolism and cardiac metabolism were the most disrupted in Rbp1-/- mice. Together, these studies characterize the effect of CRBP1 loss and reduced RA in the adult heart.
Collapse
Affiliation(s)
- Stephanie Zalesak-Kravec
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Jace W Jones
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Jenna Alloush
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Amy E Defnet
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Alexander R Moise
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Antitumor Potency of an Anti-CD19 Chimeric Antigen Receptor T-Cell Therapy, Lisocabtagene Maraleucel in Combination With Ibrutinib or Acalabrutinib. J Immunother 2021; 43:107-120. [PMID: 31899702 PMCID: PMC7144752 DOI: 10.1097/cji.0000000000000307] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Supplemental Digital Content is available in the text. Chimeric antigen receptor (CAR) T-cell therapy is a promising treatment for patients with CD19+ B-cell malignancies. Combination strategies that improve CAR T-cell potency, limit tumor environment–mediated immune dysfunction, and directly reduce tumor burden may increase the potential for durable clinical benefit of CAR T-cell therapy. Lisocabtagene maraleucel (liso-cel) is a product therapy candidate being tested in patients with relapsed/refractory non-Hodgkin lymphoma or chronic lymphocytic leukemia. This study assessed the in vitro and in vivo functionality of CAR T cells transduced to express the anti-CD19 CAR of liso-cel in combination with ibrutinib or acalabrutinib. In prolonged stimulation assays, the presence of ibrutinib or acalabrutinib improved the CAR T-cell effector function. RNA-Seq analysis and surface marker profiling of these CAR T cells treated with ibrutinib but not acalabrutinib revealed gene expression changes consistent with skewing toward a memory-like, type 1 T-helper, Bruton tyrosine kinase phenotype. Ibrutinib or acalabrutinib improved CD19+ tumor clearance and prolonged survival of tumor-bearing mice when used in combination with CAR T cells. A combination of the defined cell product therapy candidate, liso-cel, with ibrutinib or acalabrutinib is an attractive approach that may potentiate the promising clinical responses already achieved in CD19+ B-cell malignancies with each of these single agents.
Collapse
|
10
|
Luo JY, Yan SB, Chen G, Chen P, Liang SW, Xu QQ, Gu JH, Huang ZG, Qin LT, Lu HP, Mo WJ, Luo YG, Chen JB. RNA-Sequencing, Connectivity Mapping, and Molecular Docking to Investigate Ligand-Protein Binding for Potential Drug Candidates for the Treatment of Wilms Tumor. Med Sci Monit 2020; 26:e920725. [PMID: 32214060 PMCID: PMC7119447 DOI: 10.12659/msm.920725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Wilms tumor, or nephroblastoma, is a malignant pediatric embryonal renal tumor that has a poor prognosis. This study aimed to use bioinformatics data, RNA-sequencing, connectivity mapping, molecular docking, and ligand-protein binding to identify potential targets for drug therapy in Wilms tumor. Material/Methods Wilms tumor and non-tumor samples were obtained from high throughput gene expression databases, and differentially expressed genes (DEGs) were analyzed using the voom method in the limma package. The overlapping DEGs were obtained from the intersecting drug target genes using the Connectivity Map (CMap) database, and systemsDock was used for molecular docking. Gene databases were searched for gene expression profiles for complementary analysis, analysis of clinical significance, and prognosis analysis to refine the study. Results From 177 cases of Wilms tumor, there were 648 upregulated genes and 342 down-regulated genes. Gene Ontology (GO) enrichment analysis showed that the identified DEGs that affected the cell cycle. After obtaining 21 candidate drugs, there were seven overlapping genes with 75 drug target genes and DEGs. Molecular docking results showed that relatively high scores were obtained when retinoic acid and the cyclin-dependent kinase inhibitor, alsterpaullone, were docked to the overlapping genes. There were significant standardized mean differences for three overlapping genes, CDK2, MAP4K4, and CRABP2. However, four upregulated overlapping genes, CDK2, MAP4K4, CRABP2, and SIRT1 had no prognostic significance. Conclusions RNA-sequencing, connectivity mapping, and molecular docking to investigate ligand-protein binding identified retinoic acid and alsterpaullone as potential drug candidates for the treatment of Wilms tumor.
Collapse
Affiliation(s)
- Jia-Yuan Luo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Shi-Bai Yan
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Peng Chen
- Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Song-Wu Liang
- Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Qiong-Qian Xu
- Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jin-Han Gu
- Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Zhi-Guang Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Li-Ting Qin
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Hui-Ping Lu
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Wei-Jia Mo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yi-Ge Luo
- Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jia-Bo Chen
- Department of Pediatric Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
11
|
Friesenbichler W, Krizmanich W, Lakatos K, Attarbaschi A, Dworzak M, Amann G, Furtwängler R, Graf N, Kager L. Outcome of two patients with bilateral nephroblastomatosis/Wilms tumour treated with an add-on 13-cis retinoic acid therapy - Case report. Pediatr Hematol Oncol 2018; 35:218-224. [PMID: 30260265 DOI: 10.1080/08880018.2018.1515284] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Although the fate of nephrogenic rests varies, they are known to be precursors of Wilms tumour. Thus, nephrogenic rests require adequate treatment to prevent malignant transformation. We added 13-cis retinoic acid to the standard chemotherapy with vincristine and actinomycin-D in two patients with bilateral nephrogenic rests/nephroblastomatosis. Patient 1 also had a history of Wilms tumour. 46 (patient 1) and 81 (patient 2) months after end of treatment, both patients show stable conditions with no signs of relapse or progressive disease. Our observation supports further investigation of retinoic acid in patients with nephrogenic rests and nephroblastomatosis.
Collapse
Affiliation(s)
- Waltraud Friesenbichler
- a Department of Pediatric Hematology and Oncology , St. Anna Children's Hospital, Medical University of Vienna , Vienna , Austria
| | - Wolfgang Krizmanich
- b Department of Radiology , St. Anna Children's Hospital, Medical University of Vienna , Vienna , Austria
| | - Karoly Lakatos
- b Department of Radiology , St. Anna Children's Hospital, Medical University of Vienna , Vienna , Austria
| | - Andishe Attarbaschi
- a Department of Pediatric Hematology and Oncology , St. Anna Children's Hospital, Medical University of Vienna , Vienna , Austria
| | - Michael Dworzak
- a Department of Pediatric Hematology and Oncology , St. Anna Children's Hospital, Medical University of Vienna , Vienna , Austria.,c Children's Cancer Research Institute (CCRI) , Vienna , Austria
| | - Gabriele Amann
- d Institute of Pathology , Medical University of Vienna , Vienna , Austria
| | - Rhoikos Furtwängler
- e Department for Pediatric Hematology and Oncology , Saarland University , Homburg , Germany
| | - Norbert Graf
- e Department for Pediatric Hematology and Oncology , Saarland University , Homburg , Germany
| | - Leo Kager
- a Department of Pediatric Hematology and Oncology , St. Anna Children's Hospital, Medical University of Vienna , Vienna , Austria
| |
Collapse
|
12
|
Percicote AP, Mardegan GL, Gugelmim ES, Ioshii SO, Kuczynski AP, Nagashima S, de Noronha L. Tissue expression of retinoic acid receptor alpha and CRABP2 in metastatic nephroblastomas. Diagn Pathol 2018; 13:9. [PMID: 29378601 PMCID: PMC6389245 DOI: 10.1186/s13000-018-0686-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 01/15/2018] [Indexed: 01/09/2023] Open
Abstract
Background Nephroblastoma or Wilms tumor is the most frequent kidney cancer in children and accounts for 98% of kidney tumors in this age group. Despite favorable prognosis, a subgroup of these patients progresses to recurrence and death. The retinoic acid (RA) pathway plays a role in the chemoprevention and treatment of tumors due to its effects on cell differentiation and its antiproliferative, anti-oxidant, and pro-apoptotic activities. Reports describe abnormal cellular retinoic acid-binding protein 2 (CRABP2) expression in neoplasms and its correlation with prognostic factors and clinical and pathological characteristics. The aim of this study was to evaluate the immunohistochemical expression of retinoic acid receptor alpha (RARA) and CRABP2 in paraffin-embedded samples of nephroblastomas via semiquantitative and quantitative analyses and to correlate this expression with prognostic factors. Methods Seventy-seven cases of nephroblastomas were selected from pediatric oncology services. The respective medical records and surgical specimens were reviewed. Three representative tumor samples and one non-tumor renal tissue sample were selected for the preparation of tissue microarrays (TMA). The Allred scoring system was used for semiquantitative immunohistochemical analyses, whereas a morphometric analysis of the stained area was employed for quantitative evaluation. The nonparametric Mann-Whitney test was used for comparisons between two groups, while the nonparametric Kruskal-Wallis test was used to compare three or more groups. Results Immunopositivity for RARA and CRABP2 was observed in both the nucleus and cytoplasm. All histological components of the nephroblastoma (blastema, epithelium, and stroma) were positive for both markers. RARA, based on semiquantitative analyses, and CRABP2, bases on quantitative analyses, exhibited increased immunohistochemical expression in patients with metastasis, with p values of 0.0247 and 0.0128, respectively. These findings were similar to the results of the quantitative analysis of RARA expression, showing greater immunopositivity in tumor samples of patients subjected to pre-surgical chemotherapy. No significant correlation was found with the other variables studied, such as disease stage, anaplasia, risk group, histological type, nodal involvement, and clinical evolution. Conclusions Semiquantitative and quantitative analyses of the markers RARA and CRABP2 indicate their potential as biomarkers for tumor progression and their participation in nephroblastoma tumorigenesis. Electronic supplementary material The online version of this article (10.1186/s13000-018-0686-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | - Sergio Ossamu Ioshii
- Department of Medical Pathology, Federal University of Paraná and School of Health of the Pontifical Catholic University of Paraná, Curitiba, Brazil
| | | | - Seigo Nagashima
- School of Health of the Pontifical Catholic University of Paraná, Curitiba, Brazil
| | - Lúcia de Noronha
- Department of Medical Pathology, Federal University of Paraná and School of Health of the Pontifical Catholic University of Paraná, Curitiba, Brazil
| |
Collapse
|
13
|
Amarante MK, de Oliveira CEC, Ariza CB, Sakaguchi AY, Ishibashi CM, Watanabe MAE. The predictive value of transforming growth factor-β in Wilms tumor immunopathogenesis. Int Rev Immunol 2017; 36:233-239. [PMID: 28481647 DOI: 10.1080/08830185.2017.1291639] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Wilms tumor is the most common kidney malignancy in children, especially in children aged less than 6 years. Although therapeutic approach has reached successful rates, there is still room for improvement. Considering the tumor microenvironment, cytokines represent important elements of interaction and communication between tumor cells, stroma, and immune cells. In this regard, the transforming growth factor beta (TGF-β) family members play significant functions in physiological and pathological conditions, particularly in cancer. By regulating cell growth, death, and immortalization, TGF-β signaling pathways exert tumor suppressor effects in normal and early tumor cells. Thus, it is not surprising that a high number of human tumors arise due to alterations in genes coding for various TGF-β signaling components. Understanding the ambiguous role of TGF-β in human cancer is of paramount importance for the development of new therapeutic strategies to specifically block the metastatic signaling pathway of TGF-β without affecting its tumor suppressive effect. In this context, this review attempt to summarize the involvement of TGF-β in Wilms tumor.
Collapse
Affiliation(s)
- Marla Karine Amarante
- a Laboratory of Study and Application of DNA Polymorphisms, Department of Pathological Sciences , Biological Sciences Center, State University of Londrina , Londrina-Paraná , Brazil
| | - Carlos Eduardo Coral de Oliveira
- a Laboratory of Study and Application of DNA Polymorphisms, Department of Pathological Sciences , Biological Sciences Center, State University of Londrina , Londrina-Paraná , Brazil
| | - Carolina Batista Ariza
- a Laboratory of Study and Application of DNA Polymorphisms, Department of Pathological Sciences , Biological Sciences Center, State University of Londrina , Londrina-Paraná , Brazil
| | - Alberto Yoichi Sakaguchi
- a Laboratory of Study and Application of DNA Polymorphisms, Department of Pathological Sciences , Biological Sciences Center, State University of Londrina , Londrina-Paraná , Brazil
| | - Cintya Mayumi Ishibashi
- a Laboratory of Study and Application of DNA Polymorphisms, Department of Pathological Sciences , Biological Sciences Center, State University of Londrina , Londrina-Paraná , Brazil
| | - Maria Angelica Ehara Watanabe
- a Laboratory of Study and Application of DNA Polymorphisms, Department of Pathological Sciences , Biological Sciences Center, State University of Londrina , Londrina-Paraná , Brazil
| |
Collapse
|
14
|
The Retinoid Agonist Tazarotene Promotes Angiogenesis and Wound Healing. Mol Ther 2016; 24:1745-1759. [PMID: 27480772 DOI: 10.1038/mt.2016.153] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/26/2016] [Indexed: 12/16/2022] Open
Abstract
Therapeutic angiogenesis is a major goal of regenerative medicine, but no clinically approved small molecule exists that enhances new blood vessel formation. Here we show, using a phenotype-driven high-content imaging screen of an annotated chemical library of 1,280 bioactive small molecules, that the retinoid agonist Tazarotene, enhances in vitro angiogenesis, promoting branching morphogenesis, and tubule remodeling. The proangiogenic phenotype is mediated by retinoic acid receptor but not retinoic X receptor activation, and is characterized by secretion of the proangiogenic factors hepatocyte growth factor, vascular endothelial growth factor, plasminogen activator, urokinase and placental growth factor, and reduced secretion of the antiangiogenic factor pentraxin-3 from adjacent fibroblasts. In vivo, Tazarotene enhanced the growth of mature and functional microvessels in Matrigel implants and wound healing models, and increased blood flow. Notably, in ear punch wound healing model, Tazarotene promoted tissue repair characterized by rapid ear punch closure with normal-appearing skin containing new hair follicles, and maturing collagen fibers. Our study suggests that Tazarotene, an FDA-approved small molecule, could be potentially exploited for therapeutic applications in neovascularization and wound healing.
Collapse
|
15
|
Schneider L, Stöckel D, Kehl T, Gerasch A, Ludwig N, Leidinger P, Huwer H, Tenzer S, Kohlbacher O, Hildebrandt A, Kaufmann M, Gessler M, Keller A, Meese E, Graf N, Lenhof HP. DrugTargetInspector: An assistance tool for patient treatment stratification. Int J Cancer 2016; 138:1765-76. [PMID: 26501925 DOI: 10.1002/ijc.29897] [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: 06/18/2015] [Revised: 09/16/2015] [Accepted: 10/08/2015] [Indexed: 12/21/2022]
Abstract
Cancer is a large class of diseases that are characterized by a common set of features, known as the Hallmarks of cancer. One of these hallmarks is the acquisition of genome instability and mutations. This, combined with high proliferation rates and failure of repair mechanisms, leads to clonal evolution as well as a high genotypic and phenotypic diversity within the tumor. As a consequence, treatment and therapy of malignant tumors is still a grand challenge. Moreover, under selective pressure, e.g., caused by chemotherapy, resistant subpopulations can emerge that then may lead to relapse. In order to minimize the risk of developing multidrug-resistant tumor cell populations, optimal (combination) therapies have to be determined on the basis of an in-depth characterization of the tumor's genetic and phenotypic makeup, a process that is an important aspect of stratified medicine and precision medicine. We present DrugTargetInspector (DTI), an interactive assistance tool for treatment stratification. DTI analyzes genomic, transcriptomic, and proteomic datasets and provides information on deregulated drug targets, enriched biological pathways, and deregulated subnetworks, as well as mutations and their potential effects on putative drug targets and genes of interest. To demonstrate DTI's broad scope of applicability, we present case studies on several cancer types and different types of input -omics data. DTI's integrative approach allows users to characterize the tumor under investigation based on various -omics datasets and to elucidate putative treatment options based on clinical decision guidelines, but also proposing additional points of intervention that might be neglected otherwise. DTI can be freely accessed at http://dti.bioinf.uni-sb.de.
Collapse
Affiliation(s)
- Lara Schneider
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany.,Saarbrücken Graduate School of Computer Science, Saarland University, Saarbrücken, Germany
| | - Daniel Stöckel
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany.,Saarbrücken Graduate School of Computer Science, Saarland University, Saarbrücken, Germany
| | - Tim Kehl
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany.,Saarbrücken Graduate School of Computer Science, Saarland University, Saarbrücken, Germany
| | - Andreas Gerasch
- Center for Bioinformatics, Eberhard-Karls-University, Tübingen, Germany
| | - Nicole Ludwig
- Department of Human Genetics, Medical School, Saarland University, Homburg, Germany
| | - Petra Leidinger
- Department of Human Genetics, Medical School, Saarland University, Homburg, Germany
| | - Hanno Huwer
- Department of Cardiothoracic Surgery, Völklingen Heart Center, Völklingen, Germany
| | - Stefan Tenzer
- Institute for Immunology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Oliver Kohlbacher
- Center for Bioinformatics, Eberhard-Karls-University, Tübingen, Germany.,Biomolecular Interactions, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Andreas Hildebrandt
- Institute of Computer Science, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael Kaufmann
- Center for Bioinformatics, Eberhard-Karls-University, Tübingen, Germany
| | - Manfred Gessler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, and Comprehensive Cancer Center Mainfranken, Würzburg University, Würzburg, Germany
| | - Andreas Keller
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Eckart Meese
- Department of Human Genetics, Medical School, Saarland University, Homburg, Germany
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Medical School, Saarland University, Homburg, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
| |
Collapse
|
16
|
Sambyal V, Guleria K, Kapahi R, Manjari M, Sudan M, Uppal MS, Singh NR. Association of the -2518 A/G Polymorphism of MCP-1 with Breast Cancer in Punjab, North-West India. Asian Pac J Cancer Prev 2015; 16:7243-8. [DOI: 10.7314/apjcp.2015.16.16.7243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
17
|
Aine M, Sjödahl G, Eriksson P, Veerla S, Lindgren D, Ringnér M, Höglund M. Integrative epigenomic analysis of differential DNA methylation in urothelial carcinoma. Genome Med 2015; 7:23. [PMID: 25810763 PMCID: PMC4373102 DOI: 10.1186/s13073-015-0144-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/17/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Urothelial carcinoma of the bladder (UC) is a common malignancy. Although extensive transcriptome analysis has provided insights into the gene expression patterns of this tumor type, the mechanistic underpinnings of differential methylation remain poorly understood. Multi-level genomic data may be used to profile the regulatory potential and landscape of differential methylation in cancer and gain understanding of the processes underlying epigenetic and phenotypic characteristics of tumors. METHODS We perform genome-wide DNA methylation profiling of 98 gene-expression subtyped tumors to identify between-tumor differentially methylated regions (DMRs). We integrate multi-level publically available genomic data generated by the ENCODE consortium to characterize the regulatory potential of UC DMRs. RESULTS We identify 5,453 between-tumor DMRs and derive four DNA methylation subgroups of UC with distinct associations to clinicopathological features and gene expression subtypes. We characterize three distinct patterns of differential methylation and use ENCODE data to show that tumor subgroup-defining DMRs display differential chromatin state, and regulatory factor binding preferences. Finally, we characterize an epigenetic switch involving the HOXA-genes with associations to tumor differentiation states and patient prognosis. CONCLUSIONS Genome-wide DMR methylation patterns are reflected in the gene expression subtypes of UC. UC DMRs display three distinct methylation patterns, each associated with intrinsic features of the genome and differential regulatory factor binding preferences. Epigenetic inactivation of HOX-genes correlates with tumor differentiation states and may present an actionable epigenetic alteration in UC.
Collapse
Affiliation(s)
- Mattias Aine
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Gottfrid Sjödahl
- Division of Urologic Research, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Pontus Eriksson
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Srinivas Veerla
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - David Lindgren
- Division of Translational Cancer Research, Department of Laboratory Medicine Lund, Lund University, Lund, Sweden
| | - Markus Ringnér
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Mattias Höglund
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
18
|
Li L, Zhang J, Weng X, Wen G. Genetic variations in monocyte chemoattractant protein-1 and susceptibility to ovarian cancer. Tumour Biol 2015; 36:233-238. [PMID: 25234717 DOI: 10.1007/s13277-014-2619-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 09/09/2014] [Indexed: 11/27/2022] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1) is a chemokine which plays critical roles in regulating host immune responses. Researches have shown that MCP-1 may greatly participate in the development of different cancers. In the current study, we investigated the effect of MCP-1 on ovarian cancer by examining the association between MCP-1 genetic polymorphisms and the susceptibility to ovarian cancer. MCP-1 -2158A/G and MCP-1 -362C/G polymorphisms were examined in ovarian cancer patients and healthy controls by using polymerase chain reaction-restriction fragment length polymorphism analysis. Results showed that percentages of MCP-1 -2158GG genotype and G allele were significantly higher in ovarian cancer patients than in controls (odd ratio (OR) = 1.87; 95 % confidence interval (CI), 1.19-2.76; P = 0.012 and OR = 1.47; 95 % CI, 1.11-1.79; P = 0.003; data were adjusted for age and smoking status). The MCP-1 -362GG genotype also revealed increased number in patients. Stratification analyses presented that ovarian cancer cases with serous-papillary type had significantly increased percentage of -362GG genotype than those with other types (13.1 versus 5.0 %, P = 0.032; data were adjusted for age and smoking status). Also, we evaluated the relation between these two polymorphisms and serum level of MCP-1. We identified that the subjects with MCP-1 -2158AG and GG genotypes had clearly increased serum level of MCP-1 than those with AA genotype. These data suggest that MCP-1 may be involved in the pathogenesis of ovarian cancer.
Collapse
Affiliation(s)
- Li Li
- Department of Gynaecology, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | | | | | | |
Collapse
|
19
|
Wells JE, Howlett M, Cole CH, Kees UR. Deregulated expression of connective tissue growth factor (CTGF/CCN2) is linked to poor outcome in human cancer. Int J Cancer 2014; 137:504-11. [PMID: 24832082 DOI: 10.1002/ijc.28972] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 04/13/2014] [Accepted: 05/02/2014] [Indexed: 12/14/2022]
Abstract
Connective tissue growth factor (CTGF/CCN2) has long been associated with human cancers. The role it plays in these neoplasms is diverse and tumour specific. Recurring patterns in clinical outcome, histological desmoplasia and mechanisms of action have been found. When CTGF is overexpressed compared to low-expressing normal tissue or is underexpressed compared to high-expressing normal tissue, the functional outcome favours tumour survival and disease progression. CTGF acts by altering proliferation, drug resistance, angiogenesis, adhesion and migration contributing to metastasis. The pattern of CTGF expression and tumour response helps to clarify the role of this matricellular protein across a multitude of human cancers.
Collapse
Affiliation(s)
- Julia E Wells
- Leukaemia and Cancer Division, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Perth, WA, Australia
| | - Meegan Howlett
- Leukaemia and Cancer Division, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Catherine H Cole
- School of Paediatrics and Child Health, The University of Western Australia, Perth, WA, Australia
| | - Ursula R Kees
- Leukaemia and Cancer Division, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
20
|
Levy G, Malik M, Britten J, Gilden M, Segars J, Catherino WH. Liarozole inhibits transforming growth factor-β3--mediated extracellular matrix formation in human three-dimensional leiomyoma cultures. Fertil Steril 2014; 102:272-281.e2. [PMID: 24825427 DOI: 10.1016/j.fertnstert.2014.03.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/20/2014] [Accepted: 03/20/2014] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To investigate the impact of liarozole on transforming growth factor-β3 (TGF-β3) expression, TGF-β3 controlled profibrotic cytokines, and extracellular matrix formation in a three-dimensional (3D) leiomyoma model system. DESIGN Molecular and immunohistochemical analysis in a cell line evaluated in a three-dimensional culture. SETTING Laboratory study. PATIENT(S) None. INTERVENTION(S) Treatment of leiomyoma and myometrial cells with liarozole and TGF-β3 in a three-dimensional culture system. MAIN OUTCOME MEASURE(S) Quantitative real-time reverse-transcriptase polymerase chain reaction and Western blotting to assess fold gene and protein expression of TGF-β3 and TGF-β3 regulated fibrotic cytokines: collagen 1A1 (COL1A1), fibronectin, and versican before and after treatment with liarozole, and confirmatory immunohistochemical stains of treated three-dimensional cultures. RESULT(S) Both TGF-β3 gene and protein expression were elevated in leiomyoma cells compared with myometrium in two-dimensional and 3D cultures. Treatment with liarozole decreased TGF-β3 gene and protein expression. Extracellular matrix components versican, COL1A1, and fibronectin were also decreased by liarozole treatment in 3D cultures. Treatment of 3D cultures with TGF-β3 increased gene expression and protein production of COL1A1, fibronectin, and versican. CONCLUSION(S) Liarozole decreased TGF-β3 and TGF-β3-mediated extracellular matrix expression in a 3D uterine leiomyoma culture system.
Collapse
Affiliation(s)
- Gary Levy
- Program in Reproductive and Adult Endocrinology Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Minnie Malik
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Joy Britten
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Melissa Gilden
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - James Segars
- Program in Reproductive and Adult Endocrinology Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - William H Catherino
- Program in Reproductive and Adult Endocrinology Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, Maryland.
| |
Collapse
|
21
|
Pritchard-Jones K, Maschietto M, Grundy P. Biological Prognostic Factors in Wilms Tumors. RENAL TUMORS OF CHILDHOOD 2014. [DOI: 10.1007/978-3-662-44003-2_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
22
|
Xu Q, Kopp JB. Retinoid and TGF-β families: crosstalk in development, neoplasia, immunity, and tissue repair. Semin Nephrol 2012; 32:287-94. [PMID: 22835460 DOI: 10.1016/j.semnephrol.2012.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transforming growth factor-β (TGF-β) isoforms are profibrotic cytokines, par excellence, and have complex multifunctional effects on many systems, depending on the biologic setting. Retinoids are vitamin A derivatives that also have diverse effects in development, physiology, and disease. The interactions between these classes of molecules are, not surprisingly, highly complex and are dependent on the tissue, cellular, and molecular settings.
Collapse
Affiliation(s)
- Qihe Xu
- Department of Renal Medicine, King's College London, London, UK
| | | |
Collapse
|
23
|
Abstract
Lysophosphatidic acid (LPA; monoacyl-glycerol-3-phosphate) is a lipid mediator that functions as a mitogen and motility factor for many cell types. LPA signals through six specific G protein-coupled receptors, named LPA(1-6), which trigger both overlapping and distinct signaling pathways. LPA is produced from extracellular lysophosphatidylcholine by a secreted lysophospholipase D, named autotaxin (ATX), originally identified as an "autocrine motility factor" for tumor cells. ATX-LPA signaling is vital for embryonic development and promotes tumor formation, angiogenesis, and experimental metastasis in mice. Elevated expression of ATX and/or aberrant expression of LPA receptors are found in several human malignancies, while loss of LPA(6) function has been implicated in bladder cancer. In this review, we summarize our present understanding of ATX and LPA receptor signaling in cancer.
Collapse
Affiliation(s)
- Anna J S Houben
- Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
24
|
Hao Z, Zhang H, Cowell J. Ubiquitin-conjugating enzyme UBE2C: molecular biology, role in tumorigenesis, and potential as a biomarker. Tumour Biol 2011; 33:723-30. [PMID: 22170434 DOI: 10.1007/s13277-011-0291-1] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 12/01/2011] [Indexed: 01/28/2023] Open
Abstract
Ubiquitin-conjugating enzyme 2C (UBE2C) participates in cell cycle progression and checkpoint control by targeted degradation of short-lived proteins. As a conjugating enzyme, it directs polyubiquitination to preferred lysine in the substrates. In addition to its well-known role in cyclin B destruction that is essential for exit from mitosis, UBE2C also plays an important role in mitotic spindle checkpoint control. Cells overexpressing UBE2C ignore the mitotic spindle checkpoint signals and lose genomic stability, which is a hallmark of cancer. UBE2C expression is upregulated upon malignant transformation, and amplification of UBE2C is often seen at the chromosome level in cancers in a manner similar to c-Myc, which is directly upstream of UBE2C. UBE2C levels are upregulated in a wide range of solid tumors and hematological malignancies. The level of expression correlates with the aggressiveness of the tumor. High UBE2C expression is predictive of poor survival and perhaps high risk for relapse. UBE2C immunochemistry may be integrated into the diagnosis of thyroid malignancy and gliomas. This minireview summarizes what is known about the function of UBE2C focusing on its role in the regulation of spindle assembly checkpoint, its part in tumorigenesis, and its potential as a tumor marker for various cancers.
Collapse
Affiliation(s)
- Zhonglin Hao
- Georgia Health Sciences University Cancer Center, Georgia Health Sciences University, 1120 15th street, Augusta, GA 30912, USA.
| | | | | |
Collapse
|
25
|
Wegert J, Bausenwein S, Kneitz S, Roth S, Graf N, Geissinger E, Gessler M. Retinoic acid pathway activity in Wilms tumors and characterization of biological responses in vitro. Mol Cancer 2011; 10:136. [PMID: 22067876 PMCID: PMC3239322 DOI: 10.1186/1476-4598-10-136] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 11/08/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Wilms tumor (WT) is one of the most common malignancies in childhood. With current therapy protocols up to 90% of patients can be cured, but there is still a need to improve therapy for patients with aggressive WT and to reduce treatment intensity where possible. Prior data suggested a deregulation of the retinoic acid (RA) signaling pathway in high-risk WT, but its mode of action remained unclear. RESULTS The association of retinoid signaling and clinical parameters could be validated in a large independent tumor set, but its relevance in primary nephrectomy tumors from very young children may be different. Reduced RA pathway activity and MYCN overexpression were found in high risk tumors as opposed to tumors with low/intermediate risk, suggesting a beneficial impact of RA especially on advanced WT. To search for possible modes of action of retinoids as novel therapeutic options, primary tumor cell cultures were treated in vitro with all-trans-RA (ATRA), 9cis-RA, fenretinide and combinations of retinoids and a histone deacetylase (HDAC) inhibitor. Genes deregulated in high risk tumors showed opposite changes upon treatment suggesting a positive effect of retinoids. 6/7 primary cultures tested reduced proliferation, irrespective of prior RA signaling levels. The only variant culture was derived from mesoblastic nephroma, a distinct childhood kidney neoplasm. Retinoid/HDAC inhibitor combinations provided no synergistic effect. ATRA and 9cis-RA induced morphological changes suggestive of differentiation, while fenretinide induced apoptosis in several cultures tested. Microarray analysis of ATRA treated WT cells revealed differential expression of many genes involved in extracellular matrix formation and osteogenic, neuronal or muscle differentiation. The effects documented appear to be reversible upon drug withdrawal, however. CONCLUSIONS Altered retinoic acid signaling has been validated especially in high risk Wilms tumors. In vitro testing of primary tumor cultures provided clear evidence of a potential utility of retinoids in Wilms tumor treatment based on the analysis of gene expression, proliferation, differentiation and apoptosis.
Collapse
Affiliation(s)
- Jenny Wegert
- Developmental Biochemistry, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
26
|
Wegert J, Bausenwein S, Roth S, Graf N, Geissinger E, Gessler M. Characterization of primary Wilms tumor cultures as an in vitro model. Genes Chromosomes Cancer 2011; 51:92-104. [PMID: 22034155 DOI: 10.1002/gcc.20936] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 09/07/2011] [Indexed: 01/31/2023] Open
Abstract
Functional analysis of gene candidates and testing of novel therapeutics in Wilms tumors (WT) has been hampered by the lack of in vitro model systems. WT are characterized by a spectrum of histological appearances, but published cell lines are mostly derived from rare anaplastic variants or even non-WT. There has been some success in establishing primary cultures, but these are often poorly characterized or only derived from less frequent WT1 mutant tumors. We report the generation of a set of primary WT-cell cultures using a simple cultivation protocol. Our cultures could be established after preoperative chemotherapy and irrespective of histological subtypes or genetic alterations. The presence of tumor-specific genetic alterations validates these cultures as being tumor-derived. Genetic characterization is of utmost importance as some cultures with similar morphological appearance lacked such alterations and either represent clonal variants or normal cells. By immunohistochemistry, the cells are either epithelial or more mesenchymal, and the latter exhibiting a longer life span with 30 or more passages before undergoing senescence. This may be related to WT being embryonal tumors with a strong differentiation potential that may prevail in vitro. Telomeres progressively shorten with cultivation, but their length does not predict lifespan. hTERT transfection may partly allow establishment of immortalized lines, because 2/7 cultures avoid senescence even in later passages. Importantly, these cells can be efficiently manipulated by transfection, making them a useful model system for in vitro testing.
Collapse
Affiliation(s)
- Jenny Wegert
- Developmental Biochemistry, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | | | | | | | | | | |
Collapse
|
27
|
Spreafico F, Pritchard Jones K, Malogolowkin MH, Bergeron C, Hale J, de Kraker J, Dallorso S, Acha T, de Camargo B, Dome JS, Graf N. Treatment of relapsed Wilms tumors: lessons learned. Expert Rev Anticancer Ther 2010; 9:1807-15. [PMID: 19954292 DOI: 10.1586/era.09.159] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Treatment regimens for recurrent Wilms tumor (WT) are currently designed to include drugs that are not used during primary chemotherapy, using a risk-stratified approach. Therapy of recurrent disease depends on the nature of initial treatment, and of recognized prognostic indicators inherent in the primary tumor. Several highly effective chemotherapy regimens, including ifosfamide-carboplatin-etoposide, cyclophosphamide-etoposide and carboplatin-etoposide, are considered first treatment choice for recurrent disease. While intense-dose chemotherapy is uniformly accepted to treat high-risk recurrent WTs, the optimal therapy for standard-risk children has yet to be defined, owing to the small number of such patients and their relatively better prognosis compared with high-risk recurrences. Recurrent tumors among those defined as very-high risk are likely to develop chemoresistant disease, and novel therapeutic strategies will be necessary to cure these patients. Evidence on how to properly administer surgery and radiotherapy at relapse is more fragmentary. The authors have reviewed the available experiences concerning the treatment of recurrent WT, and have attempted to provide the most up-to-date recommendations regarding the optimal risk-based treatment for these patients.
Collapse
Affiliation(s)
- Filippo Spreafico
- Pediatric Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133 Milano, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Delacroix L, Moutier E, Altobelli G, Legras S, Poch O, Choukrallah MA, Bertin I, Jost B, Davidson I. Cell-specific interaction of retinoic acid receptors with target genes in mouse embryonic fibroblasts and embryonic stem cells. Mol Cell Biol 2010; 30:231-44. [PMID: 19884340 PMCID: PMC2798310 DOI: 10.1128/mcb.00756-09] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Revised: 07/15/2009] [Accepted: 10/22/2009] [Indexed: 11/20/2022] Open
Abstract
All-trans retinoic acid (RA) induces transforming growth factor beta (TGF-beta)-dependent autocrine growth of mouse embryonic fibroblasts (MEFs). We have used chromatin immunoprecipitation to map 354 RA receptor (RAR) binding loci in MEFs, most of which were similarly occupied by the RAR alpha and RAR gamma receptors. Only a subset of the genes associated with these loci are regulated by RA, among which are several critical components of the TGF-beta pathway. We also show RAR binding to a novel series of target genes involved in cell cycle regulation, transformation, and metastasis, suggesting new pathways by which RA may regulate proliferation and cancer. Few of the RAR binding loci contained consensus direct-repeat (DR)-type elements. The majority comprised either degenerate DRs or no identifiable DRs but anomalously spaced half sites. Furthermore, we identify 462 RAR target loci in embryonic stem (ES) cells and show that their occupancy is cell type specific. Our results also show that differences in the chromatin landscape regulate the accessibility of a subset of more than 700 identified loci to RARs, thus modulating the repertoire of target genes that can be regulated and the biological effects of RA.
Collapse
Affiliation(s)
- Laurence Delacroix
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, 1 Rue Laurent Fries, 67404 Illkirch Cédex, France, Immunologie et Maladies Infectieuses, GIGA +2, Bat B34, 1 ave. de l'Hôpital, 4000 Liège, Belgium, Ecole Supérieur de Biotechnologie de Strasbourg, Pole API, 67400 Illkirch, France
| | - Emmanuel Moutier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, 1 Rue Laurent Fries, 67404 Illkirch Cédex, France, Immunologie et Maladies Infectieuses, GIGA +2, Bat B34, 1 ave. de l'Hôpital, 4000 Liège, Belgium, Ecole Supérieur de Biotechnologie de Strasbourg, Pole API, 67400 Illkirch, France
| | - Gioia Altobelli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, 1 Rue Laurent Fries, 67404 Illkirch Cédex, France, Immunologie et Maladies Infectieuses, GIGA +2, Bat B34, 1 ave. de l'Hôpital, 4000 Liège, Belgium, Ecole Supérieur de Biotechnologie de Strasbourg, Pole API, 67400 Illkirch, France
| | - Stephanie Legras
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, 1 Rue Laurent Fries, 67404 Illkirch Cédex, France, Immunologie et Maladies Infectieuses, GIGA +2, Bat B34, 1 ave. de l'Hôpital, 4000 Liège, Belgium, Ecole Supérieur de Biotechnologie de Strasbourg, Pole API, 67400 Illkirch, France
| | - Olivier Poch
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, 1 Rue Laurent Fries, 67404 Illkirch Cédex, France, Immunologie et Maladies Infectieuses, GIGA +2, Bat B34, 1 ave. de l'Hôpital, 4000 Liège, Belgium, Ecole Supérieur de Biotechnologie de Strasbourg, Pole API, 67400 Illkirch, France
| | - Mohamed-Amin Choukrallah
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, 1 Rue Laurent Fries, 67404 Illkirch Cédex, France, Immunologie et Maladies Infectieuses, GIGA +2, Bat B34, 1 ave. de l'Hôpital, 4000 Liège, Belgium, Ecole Supérieur de Biotechnologie de Strasbourg, Pole API, 67400 Illkirch, France
| | - Isabelle Bertin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, 1 Rue Laurent Fries, 67404 Illkirch Cédex, France, Immunologie et Maladies Infectieuses, GIGA +2, Bat B34, 1 ave. de l'Hôpital, 4000 Liège, Belgium, Ecole Supérieur de Biotechnologie de Strasbourg, Pole API, 67400 Illkirch, France
| | - Bernard Jost
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, 1 Rue Laurent Fries, 67404 Illkirch Cédex, France, Immunologie et Maladies Infectieuses, GIGA +2, Bat B34, 1 ave. de l'Hôpital, 4000 Liège, Belgium, Ecole Supérieur de Biotechnologie de Strasbourg, Pole API, 67400 Illkirch, France
| | - Irwin Davidson
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, 1 Rue Laurent Fries, 67404 Illkirch Cédex, France, Immunologie et Maladies Infectieuses, GIGA +2, Bat B34, 1 ave. de l'Hôpital, 4000 Liège, Belgium, Ecole Supérieur de Biotechnologie de Strasbourg, Pole API, 67400 Illkirch, France
| |
Collapse
|
29
|
Abstract
A 9-month-old girl presented with massive bilateral diffuse nephroblastomatosis. After response to actinomycin D and vincristine over a period of 1 year, the nephroblastomatosis continuously progressed under this treatment. As retinoic acid signaling is critical for normal renal development and nephroblastomatosis seems histologically as undifferentiated embryonal tissue, we added 13-cis retinoic acid to the chemotherapy regimen. Three months thereafter, kidney volumes declined significantly over a period of 1 year. Interestingly, nephroblastomatosis-associated acquired von Willebrand disease also resolved. Retinoic acid maybe a novel nontoxic treatment option for nephroblastomatosis requiring further systematic evaluation.
Collapse
|
30
|
Rambow F, Malek O, Geffrotin C, Leplat JJ, Bouet S, Piton G, Hugot K, Bevilacqua C, Horak V, Vincent-Naulleau S. Identification of differentially expressed genes in spontaneously regressing melanoma using the MeLiM swine model. Pigment Cell Melanoma Res 2008; 21:147-61. [PMID: 18426408 DOI: 10.1111/j.1755-148x.2008.00442.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Partial and some few cases of complete spontaneous regression have been observed in cutaneous melanoma patients but little is known about the molecular mechanisms involved. The Melanoblastoma-bearing Libechov Minipig (MeLiM) is a suitable animal model to study the phenomenon of spontaneous regression because MeLiM pigs exhibit naturally occurring melanomas which regress completely 6 months after birth. In this study, we used suppression subtractive hybridization (SSH) to identify molecular determinants of melanoma regression within swine melanoma tissues and melanoma cell cultures. Several markers involved in cell-adhesion, -communication, -motility, signal transduction, negative regulation of cell proliferation, transport and immune response were identified that correlated with melanoma regression whereas the main genes involved in melanin synthesis showed a strong downregulation. For the most differentially expressed genes, we validated the results obtained by SSH with qRT-PCR and with immunohistochemistry for some of them (CD9, MITF, RARRES1). Most notable, for the first time in melanoma, we identified the retinoic acid responder 1 gene (RARRES1) as a main actor of the regression process in melanoma. This first gene expression study in swine melanoma regression, may contribute to the finding of new therapeutic targets for human melanoma treatment.
Collapse
Affiliation(s)
- F Rambow
- CEA, DSV, IRCM, SREIT, Laboratoire de Radiobiologie et d'Etude du Génome, Jouy-en-Josas, F-78352 France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Li MH, Sanchez T, Pappalardo A, Lynch KR, Hla T, Ferrer F. Induction of antiproliferative connective tissue growth factor expression in Wilms' tumor cells by sphingosine-1-phosphate receptor 2. Mol Cancer Res 2008; 6:1649-56. [PMID: 18922980 DOI: 10.1158/1541-7786.mcr-07-2048] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Connective tissue growth factor (CTGF), a member of the CCN family of secreted matricellular proteins, regulates fibrosis, angiogenesis, cell proliferation, apoptosis, tumor growth, and metastasis. However, the role of CTGF and its regulation mechanism in Wilms' tumor remains largely unknown. We found that the bioactive lipid sphingosine-1-phosphate (S1P) induced CTGF expression in a concentration- and time-dependent manner in a Wilms' tumor cell line (WiT49), whereas FTY720-phosphate, an S1P analogue that binds all S1P receptors except S1P2, did not. Further, the specific S1P2 antagonist JTE-013 completely inhibited S1P-induced CTGF expression, whereas the S1P1 antagonist VPC44116 did not, indicating that this effect was mediated by S1P2. This was confirmed by adenoviral transduction of S1P2 in WiT49 cells, which showed that overexpression of S1P2 increased the expression of CTGF. Induction of CTGF by S1P was sensitive to ROCK inhibitor Y-27632 and c-Jun NH2-terminal kinase inhibitor SP600125, suggesting the requirement of RhoA/ROCK and c-Jun NH2-terminal kinase pathways for S1P-induced CTGF expression. Interestingly, the expression levels of CTGF were decreased in 8 of 10 Wilms' tumor tissues compared with matched normal tissues by quantitative real-time PCR and Western blot analysis. In vitro, human recombinant CTGF significantly inhibited the proliferation of WiT49 cells. In addition, overexpression of CTGF resulted in significant inhibition of WiT49 cell growth. Taken together, these data suggest that CTGF protein induced by S1P2 might act as a growth inhibitor in Wilms' tumor.
Collapse
Affiliation(s)
- Mei-Hong Li
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
32
|
Malik M, Webb J, Catherino WH. Retinoic acid treatment of human leiomyoma cells transformed the cell phenotype to one strongly resembling myometrial cells. Clin Endocrinol (Oxf) 2008; 69:462-70. [PMID: 18248652 PMCID: PMC2610401 DOI: 10.1111/j.1365-2265.2008.03207.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Uterine leiomyomas are clinically significant tumours that may develop due to an altered differentiation pathway. We have previously identified a dysregulated retinoic acid (RA) pathway that reduced retinoic exposure in human leiomyoma surgical specimens, and have shown that the leiomyoma phenotype was characterized by excessive and disorganized extracellular matrix (ECM). OBJECTIVE The goal of this study was to determine the impact of RA exposure on the disrupted ECM phenotype of leiomyomas. DESIGN AND METHODS Study of immortalized and molecularly confirmed cells generated from surgical specimens of spontaneous uterine leiomyoma and matched myometrium. RESULTS Immortalized leiomyoma and myometrial cells retained the molecular characteristics of their progenitor tissue. Proliferation of leiomyoma cells was inhibited by all-trans retinoic acid (ATRA). Furthermore, there was a dose-dependent decrease in soluble extracellular collagen protein in ATRA-treated leiomyoma cells. Exposure of leiomyoma cells to ATRA resulted in a dose-dependent inhibition of templates for specific ECM protein production including collagen 1, collagen 4, fibronectin and versican. Notably, expression levels in treated leiomyoma cells approached those found in myometrial cells. These mRNA alterations translated into altered protein. Down-regulation was also observed among the RA pathway genes such as CYP26A1 with exposure to ATRA. Finally, ATRA down-regulated TGF-beta3 mRNA expression and the TGF-beta regulated genes in leiomyoma cells. CONCLUSION Exposure of leiomyomas to ATRA down-regulated cell proliferation, ECM formation, RA metabolism and TGF-beta regulation, suggesting that RA exposure can alter the leiomyoma phenotype to one that more closely approximates normal myometrium.
Collapse
Affiliation(s)
- Minnie Malik
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4799, USA
| | | | | |
Collapse
|
33
|
Connective tissue growth factor (CTGF) and cancer progression. J Biomed Sci 2008; 15:675-85. [PMID: 18622762 DOI: 10.1007/s11373-008-9264-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 06/06/2008] [Indexed: 12/14/2022] Open
Abstract
Connective tissue growth factor (CTGF) is a member of the CCN family of secreted, matrix-associated proteins encoded by immediate early genes that play various roles in angiogenesis and tumor growth. CCN family proteins share uniform modular structure which mediates various cellular functions such as regulation of cell division, chemotaxis, apoptosis, adhesion, motility, angiogenesis, neoplastic transformation, and ion transport. Recently, CTGF expression has been shown to be associated with tumor development and progression. There is growing body of evidence that CTGF may regulate cancer cell migration, invasion, angiogenesis, and anoikis. In this review, we will highlight the influence of CTGF expression on the biological behavior and progression of various cancer cells, as well as its regulation on various types of protein signals and their mechanisms.
Collapse
|
34
|
Lan Y, Zhang Y, Wang J, Lin C, Ittmann MM, Wang F. Aberrant expression of Cks1 and Cks2 contributes to prostate tumorigenesis by promoting proliferation and inhibiting programmed cell death. Int J Cancer 2008; 123:543-51. [PMID: 18498131 DOI: 10.1002/ijc.23548] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The mammalian Cks family consists of 2 well-conserved small proteins, Cks1 and Cks2. Cks1 has been shown to promote cell-cycle progression by triggering degradation of p27(kip1). The function of Cks2 in somatic mammalian cells is not well understood although it is required for the first metaphase/anaphase transition during the meiosis. Emerging evidence shows that elevated expression of Cks1 and Cks2 is often found in a variety of tumors, and is correlated with poor survival rate of the patients. Here we demonstrated that expression of Cks1 and Cks2 were elevated in prostate tumors of human and animal models, as well as prostatic cancer cell lines. Forced expression of Cks1 and Cks2 in benign prostate tumor epithelial cells promoted cell population growth. Knockdown of Cks1 expression in malignant prostate tumor cells inhibited proliferation, anchorage-independent growth, and migration activities, whereas knockdown of Cks2 expression induced programmed cell death and inhibited the tumorigenicity. Collectively, the data suggest that elevated expression of Cks1 contributes to the tumorigenicity of prostate tumor cells by promoting cell growth and elevated expression of Cks2 protects the cells from apoptosis. Thus, the finding suggests a novel therapeutic strategy for prostatic cancer based on inhibiting Cks1 and Cks2 activity.
Collapse
Affiliation(s)
- Yongsheng Lan
- Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030-3303, USA
| | | | | | | | | | | |
Collapse
|
35
|
Durkin ME, Yuan BZ, Zhou X, Zimonjic DB, Lowy DR, Thorgeirsson SS, Popescu NC. DLC-1:a Rho GTPase-activating protein and tumour suppressor. J Cell Mol Med 2008; 11:1185-207. [PMID: 17979893 PMCID: PMC4401278 DOI: 10.1111/j.1582-4934.2007.00098.x] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The deleted in liver cancer 1 (DLC-1) gene encodes a GTPase activating protein that acts as a negative regulator of the Rho family of small GTPases. Rho proteins transduce signals that influence cell morphology and physiology, and their aberrant up-regulation is a key factor in the neoplastic process, including metastasis. Since its discovery, compelling evidence has accumulated that demonstrates a role for DLC-1 as a bona fide tumour suppressor gene in different types of human cancer. Loss of DLC-1 expression mediated by genetic and epigenetic mechanisms has been associated with the development of many human cancers, and restoration of DLC-1 expression inhibited the growth of tumour cells in vivo and in vitro. Two closely related genes, DLC-2 and DLC-3, may also be tumour suppressors. This review presents the current status of progress in understanding the biological functions of DLC-1 and its relatives and their roles in neoplasia.
Collapse
Affiliation(s)
- Marian E Durkin
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Adamson PC, Matthay KK, O'Brien M, Reaman GH, Sato JK, Balis FM. A phase 2 trial of all-trans-retinoic acid in combination with interferon-alpha2a in children with recurrent neuroblastoma or Wilms tumor: A Pediatric Oncology Branch, NCI and Children's Oncology Group Study. Pediatr Blood Cancer 2007; 49:661-5. [PMID: 16900483 DOI: 10.1002/pbc.21011] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND The combination of the antiproliferative and differentiation-inducing effects of retinoids together with the antiproliferative, immunostimulatory, and differentiation-potentiating effects of interferon-alpha (IFN-alpha) were the basis for the development of this combination in pediatric patients with refractory neuroblastoma or Wilms tumor. PROCEDURE A phase 2 trial of all-trans-retinoic acid (ATRA), administered orally at a dose of 90 mg/m(2)/day in three divided doses for 3 consecutive days per week, and IFN-alpha2a, administered subcutaneously daily at a dose of 3 x 10(6) U/m(2)/day for 5 consecutive days per week, in 4 week cycles was performed. A two-stage design was used for each disease stratum. RESULTS Seventeen patients (16 evaluable) with neuroblastoma, median age 9 years, and 15 patients (14 evaluable) with Wilms tumor, median age 6 years, were enrolled. Overall, the combination was well tolerated, with headache being the most common toxicity observed. There were no complete or partial responses. The median number of cycles administered was 1 (range 1-9). Four patients with neuroblastoma had stable disease for 12 or more weeks. CONCLUSIONS The combination of ATRA and IFN-alpha2a was inactive in children with relapsed or refractory neuroblastoma and Wilms tumor. The lack of activity with this combination in children with refractory neuroblastoma is similar to the disappointing phase 2 results of single agent 13-cis-retinoic-acid (13cRA) and does not support further development of ATRA for children with relapsed neuroblastoma.
Collapse
Affiliation(s)
- Peter C Adamson
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW We will review the 2005 and 2006 literature on pediatric genitourinary tumors. RECENT FINDINGS Survival continues to improve for primary renal, bladder/prostate and testicular tumors in childhood. The addition of more intensive chemotherapy for anaplastic histology disease, recognition of loss of heterozygosity for chromosomes 1p and 16q as an adverse prognostic factor in favorable histology Wilms' tumor, and the utilization of molecular markers to better characterize all renal tumors will better enable individualized therapy. Recognition and treatment of anaplastic histology and bilateral Wilms' tumor remains a challenge. In rhabdomyosarcoma, genitourinary site and embryonal histology confer a relatively favorable prognosis. SUMMARY Advances in molecular oncology, diagnostic imaging, surgical approaches and long-term follow-up of childhood cancer survivors drive risk-stratified therapy in pediatric genitourinary tumors.
Collapse
Affiliation(s)
- Sharon M Castellino
- Department of Pediatrics, Hematology/Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| | | |
Collapse
|
38
|
Ullmannova V, Popescu NC. Inhibition of cell proliferation, induction of apoptosis, reactivation of DLC1, and modulation of other gene expression by dietary flavone in breast cancer cell lines. ACTA ACUST UNITED AC 2007; 31:110-8. [PMID: 17418982 PMCID: PMC1950447 DOI: 10.1016/j.cdp.2007.02.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2007] [Indexed: 12/19/2022]
Abstract
BACKGROUND Dietary flavone was previously shown to increase the expression of deleted in liver cancer-1 gene (DLC-1) in HT-29 colon carcinoma cell line [Herzog A, Kindermann B, Doring F, Daniel H, Wenzel U. Pleiotropic molecular effects of the pro-apoptotic dietary constituent flavone in human colon cancer cells identified by protein and mRNA expression profiling. Proteomics 2004;4:2455-64]. DLC-1 that encodes a Rho GTPase-activating protein, functions as a tumor suppressor gene and is frequently inactivated or down-regulated in several common cancers. Restoration of DLC-1 expression suppresses in vitro tumor cells proliferation and tumorigenicity in vivo. METHODS Here, the effect of flavone was examined in several DLC-1-deficient cell lines derived from different types human cancer using assays for cell proliferation, gene expression and transfer. RESULTS We show that exposure to 150 microM flavone increased DLC1 expression in breast but not in liver or prostate carcinoma cells or a nonmalignant breast epithelial cell line. Flavone restored the expression of DLC1 in the breast carcinoma cell lines MDA-MB-468, MDA-MB-361, and BT20 as well as in the colon carcinoma cell line HT-29 all of which are DLC-1-negative due to promoter hypermethylation. We further show that flavone inhibited cell proliferation, induced cell cycle arrest at G(2)-M, increased p21(Waf1) gene expression, and caused apoptosis. Microarray analysis of these aggressive and metastatic breast carcinoma cells revealed 29 flavone-responsive genes, among which the DNA damage-inducible GADD genes were up-regulated and the proto-oncogene STMN1 and IGFBP3 were down-regulated. CONCLUSIONS Flavone-mediated alterations of genes that regulate tumor cell proliferation, cell cycle, and apoptosis contribute to chemopreventive and antitumoral effects of flavone. Alone or in combination with demethylating agents, flavone may be an effective adjunct to chemotherapy in preventing breast cancer metastasis.
Collapse
Affiliation(s)
- Veronika Ullmannova
- Laboratory of Experimental Carcinogenesis, Center for Cancer Research, National Cancer Institute, 37 Convent Drive, MSC 4264, Bethesda, MD 20892-4255, USA
| | | |
Collapse
|
39
|
Zirn B, Samans B, Wittmann S, Pietsch T, Leuschner I, Graf N, Gessler M. Target genes of the WNT/β-catenin pathway in Wilms tumors. Genes Chromosomes Cancer 2006; 45:565-74. [PMID: 16575872 DOI: 10.1002/gcc.20319] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The WNT/beta-catenin pathway is involved in numerous human cancers. Mutations of the CTNNB1 (beta-catenin) gene have also been detected in a subset of pediatric Wilms tumors, but the target genes of the deregulated WNT/beta-catenin pathway in these tumors have yet to be identified. To compare gene expression profiles of Wilms tumors with and without mutations of CTNNB1, we used 11.5-k cDNA microarrays. Most of the tumors (86%) had received preoperative chemotherapy as mandated by the European SIOP protocol. The comparison between Wilms tumors with and without CTNNB1 mutations revealed several target genes specifically deregulated in CTNNB1-mutated Wilms tumors. Among these, PITX2, APCDD1, and two members of the endothelin axis (EDN3 and EDNRA) are directly activated downstream targets of the WNT/beta-catenin pathway that may enhance proliferation of these tumor cells. In addition, several upstream inhibitors of WNT/beta-catenin signaling like WIF1 and PRDC were also strongly up-regulated in the CTNNB1-mutated Wilms tumors. This overexpression may be a negative feedback mechanism in tumors with uncontrolled WNT signaling. Moreover, we identified deregulated genes in both the retinoic acid and the RAS pathways, such as ATX/ENPP2 and RIS1, suggesting an association between these two pathways with that of WNT. In addition, the strong representation of muscle-related genes in the expression profile of CTNNB1-mutated Wilms tumors corresponded to histologically detectable areas of myomatous cells in these tumors that displayed intense and preferential nuclear beta-catenin antibody staining. This article contains Supplementary Material available at http://www.interscience.wiley.com/jpages/1045-2257/suppmat.
Collapse
Affiliation(s)
- Birgit Zirn
- Physiological Chemistry I, Biozentrum, University of Wuerzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Zirn B, Hartmann O, Samans B, Krause M, Wittmann S, Mertens F, Graf N, Eilers M, Gessler M. Expression profiling of Wilms tumors reveals new candidate genes for different clinical parameters. Int J Cancer 2006; 118:1954-62. [PMID: 16287080 DOI: 10.1002/ijc.21564] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Wilms tumor is the most frequent renal neoplasm in children, but our understanding of its genetic basis is still limited. We performed cDNA microarray experiments using 63 primary Wilms tumors with the aim of detecting new candidate genes associated with malignancy grade and tumor progression. All tumors had received preoperative chemotherapy as mandated by the SIOP protocol, which sets this study apart from related approaches in the Unites States that are based on untreated samples. The stratification of expression data according to clinical criteria allowed a rather clear distinction between different subsets of Wilms tumors. Clear-cut differences in expression patterns were discovered between relapse-free as opposed to relapsed tumors and tumors with intermediate risk as opposed to high risk histology. Several differentially expressed genes, e.g.TRIM22, CENPF, MYCN, CTGF, RARRES3 and EZH2, were associated with Wilms tumor progression. For a subset of differentially expressed genes, microarray data were confirmed by real-time RT-PCR on the original set of tumors. Interestingly, we found the retinoic acid pathway to be deregulated at different levels in advanced tumors suggesting that treatment of these tumors with retinoic acid may represent a promising novel therapeutic approach.
Collapse
Affiliation(s)
- B Zirn
- Physiological Chemistry I, Biozentrum, University of Wuerzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|