1
|
Hiramoto K, Oikawa H. Momordica charantia Extract Ameliorates Melanoma Cell Proliferation and Invasion into Mouse Lungs by Suppressing PAX3 Expression. Int J Mol Sci 2024; 25:12800. [PMID: 39684511 DOI: 10.3390/ijms252312800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Melanomas, which develop on malignant transformations of melanocytes, are highly malignant and prone to metastasis; therefore, effective drugs are required. The Momordica charantia (MC) extract has been shown to suppress cancer cell proliferation and invasion; however, the effect of the MC extract on melanoma in living organisms remains unclear. In this study, we investigated the mechanism underlying the amelioration of melanoma cell extravasation into mouse lungs by the MC extract. Male C57BL/6j mice (aged 8 weeks) were injected with B16 melanoma cells (1 × 105 cells/mouse). Subsequently, they were orally administered the MC extract daily for 2 weeks; mouse lung samples were obtained on the final day and analyzed. The MC extract ameliorated melanoma proliferation and infiltration into the lungs caused by melanoma cell treatment. It also increased phosphatase and tensin homolog deletion from chromosome 10 and suppressed paired box gene 3 (PAX3) and the phosphatidylinositol trisphosphate/RAC-alpha serine/threonine-protein kinase/mammalian target of rapamycin complex 1 signaling. Furthermore, it decreased microphthalmia-associated transcription factors and induced the suppression of cyclin-dependent kinase 2, hepatocyte growth factor receptor, B-cell/CLL lymphoma 2, and Ras-related proteins. Our findings suggest that the MC extract suppresses tumor survival genes by regulating PAX3, thereby ameliorating melanoma proliferation and invasion.
Collapse
Affiliation(s)
- Keiichi Hiramoto
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka 513-8607, Japan
| | - Hirotaka Oikawa
- Department of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki, Suzuka 513-8607, Japan
| |
Collapse
|
2
|
Shaw T, Barr FG, Üren A. The PAX Genes: Roles in Development, Cancer, and Other Diseases. Cancers (Basel) 2024; 16:1022. [PMID: 38473380 PMCID: PMC10931086 DOI: 10.3390/cancers16051022] [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: 02/12/2024] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Since their 1986 discovery in Drosophila, Paired box (PAX) genes have been shown to play major roles in the early development of the eye, muscle, skeleton, kidney, and other organs. Consistent with their roles as master regulators of tissue formation, the PAX family members are evolutionarily conserved, regulate large transcriptional networks, and in turn can be regulated by a variety of mechanisms. Losses or mutations in these genes can result in developmental disorders or cancers. The precise mechanisms by which PAX genes control disease pathogenesis are well understood in some cases, but much remains to be explored. A deeper understanding of the biology of these genes, therefore, has the potential to aid in the improvement of disease diagnosis and the development of new treatments.
Collapse
Affiliation(s)
- Taryn Shaw
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20001, USA
| | - Frederic G Barr
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD 20892, USA
| | - Aykut Üren
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20001, USA
| |
Collapse
|
3
|
Morcos PN, Schlender J, Burghaus R, Moss J, Lloyd A, Childs BH, Macy ME, Reid JM, Chung J, Garmann D. Model-informed approach to support pediatric dosing for the pan-PI3K inhibitor copanlisib in children and adolescents with relapsed/refractory solid tumors. Clin Transl Sci 2023; 16:1197-1209. [PMID: 37042099 PMCID: PMC10339701 DOI: 10.1111/cts.13523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/21/2023] [Accepted: 03/17/2023] [Indexed: 04/13/2023] Open
Abstract
Copanlisib is an intravenously administered phosphatidylinositol 3-kinase (PI3K) inhibitor which was investigated in pediatric patients with relapsed/refractory solid tumors. A model-informed approach was undertaken to support and confirm an empirically selected starting dose of 28 mg/m2 for pediatric patients ≥1 year old, corresponding to 80% of the adult recommended dose adjusted for body surface area. An adult physiologically based pharmacokinetic (PBPK) model was initially established using copanlisib physicochemical and disposition properties and clinical pharmacokinetics (PK) data and was shown to adequately capture clinical PK across a range of copanlisib doses in adult cancer patients. The adult PBPK model was then extended to the pediatric population through incorporation of age-dependent anatomical and physiological changes and used to simulate copanlisib exposures in pediatric cancer patient age groups. The pediatric PBPK model predicted that the copanlisib 28 mg/m2 dose would achieve similar copanlisib exposures across pediatric ages when compared with historical adult exposures following the approved copanlisib 60 mg dose administered on Days 1, 8, and 15 of a 28-day cycle. Clinical PK were collected from a phase I study in pediatric patients with relapsed/refractory solid tumors (aged ≥4 years). An established adult population PK model was extended to incorporate an allometrically-scaled effect of body surface area and confirmed that the copanlisib maximum tolerated dose of 28 mg/m2 was appropriate to achieve uniform copanlisib exposures across the investigated pediatric age range and consistent exposures to historical data in adult cancer patients. The model-informed approach successfully supported and confirmed the copanlisib pediatric dose recommendation.
Collapse
Affiliation(s)
| | - Jan Schlender
- Pharmacometrics/Modeling & Simulation, Pharmaceuticals DivisionBayer AGWuppertalGermany
| | - Rolf Burghaus
- Pharmacometrics/Modeling & Simulation, Pharmaceuticals DivisionBayer AGWuppertalGermany
| | | | | | | | - Margaret E. Macy
- Department of Pediatrics, University of Colorado and Center for Cancer and Blood DisordersChildren's Hospital ColoradoAuroraColoradoUSA
| | - Joel M. Reid
- Department of PharmacologyMayo Clinic Graduate School of Biomedical SciencesRochesterMinnesotaUSA
| | - John Chung
- Bayer HealthCare Pharmaceuticals, Inc.WhippanyNew JerseyUSA
| | - Dirk Garmann
- Pharmacometrics/Modeling & Simulation, Pharmaceuticals DivisionBayer AGWuppertalGermany
| |
Collapse
|
4
|
Kahsay A, Rodriguez-Marquez E, López-Pérez A, Hörnblad A, von Hofsten J. Pax3 loss of function delays tumour progression in kRAS-induced zebrafish rhabdomyosarcoma models. Sci Rep 2022; 12:17149. [PMID: 36229514 PMCID: PMC9561152 DOI: 10.1038/s41598-022-21525-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/28/2022] [Indexed: 01/04/2023] Open
Abstract
Rhabdomyosarcoma is a soft tissue cancer that arises in skeletal muscle due to mutations in myogenic progenitors that lead to ineffective differentiation and malignant transformation. The transcription factors Pax3 and Pax7 and their downstream target genes are tightly linked with the fusion positive alveolar subtype, whereas the RAS pathway is usually involved in the embryonal, fusion negative variant. Here, we analyse the role of Pax3 in a fusion negative context, by linking alterations in gene expression in pax3a/pax3b double mutant zebrafish with tumour progression in kRAS-induced rhabdomyosarcoma tumours. Several genes in the RAS/MAPK signalling pathway were significantly down-regulated in pax3a/pax3b double mutant zebrafish. Progression of rhabdomyosarcoma tumours was also delayed in the pax3a/pax3b double mutant zebrafish indicating that Pax3 transcription factors have an unappreciated role in mediating malignancy in fusion negative rhabdomyosarcoma.
Collapse
Affiliation(s)
- A. Kahsay
- grid.12650.300000 0001 1034 3451Integrative Medical Biology (IMB), Umeå University, Johan Bures Väg 12, 90187 Umeå, Sweden
| | - E. Rodriguez-Marquez
- grid.12650.300000 0001 1034 3451Integrative Medical Biology (IMB), Umeå University, Johan Bures Väg 12, 90187 Umeå, Sweden
| | - A. López-Pérez
- grid.12650.300000 0001 1034 3451Umeå Centre for Molecular Medicine (UCMM), Umeå University, Johan Bures Väg 12, 90187 Umeå, Sweden
| | - A. Hörnblad
- grid.12650.300000 0001 1034 3451Umeå Centre for Molecular Medicine (UCMM), Umeå University, Johan Bures Väg 12, 90187 Umeå, Sweden
| | - J. von Hofsten
- grid.12650.300000 0001 1034 3451Integrative Medical Biology (IMB), Umeå University, Johan Bures Väg 12, 90187 Umeå, Sweden
| |
Collapse
|
5
|
Camero S, Camicia L, Marampon F, Ceccarelli S, Shukla R, Mannarino O, Pizer B, Schiavetti A, Pizzuti A, Tombolini V, Marchese C, Dominici C, Megiorni F. BET inhibition therapy counteracts cancer cell survival, clonogenic potential and radioresistance mechanisms in rhabdomyosarcoma cells. Cancer Lett 2020; 479:71-88. [PMID: 32200036 DOI: 10.1016/j.canlet.2020.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
The antitumour effects of OTX015, a first-in-class BET inhibitor (BETi), were investigated as a single agent or in combination with ionizing radiation (IR) in preclinical in vitro models of rhabdomyosarcoma (RMS), the most common childhood soft tissue sarcoma. Herein, we demonstrated the upregulation of BET Bromodomain gene expression in RMS tumour biopsies and cell lines compared to normal skeletal muscle. In vitro experiments showed that OTX015 significantly reduced RMS cell proliferation by altering cell cycle modulators and apoptotic related proteins due to the accumulation of DNA breaks that cells are unable to repair. Interestingly, OTX015 also impaired migration capacity and tumour-sphere architecture by downregulating pro-stemness genes and was able to potentiate ionizing radiation effects by reducing the expression of different drivers of tumour dissemination and resistance mechanisms, including the GNL3 gene, that we correlated for the first time with the RMS phenotype. In conclusion, our research sheds further light on the molecular events of OTX015 action against RMS cells and indicates this novel BETi as an effective option to improve therapeutic strategies and overcome the development of resistant cancer cells in patients with RMS.
Collapse
Affiliation(s)
- Simona Camero
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Lucrezia Camicia
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Simona Ceccarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Rajeev Shukla
- Department of Perinatal and Paediatric Pathology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| | - Olga Mannarino
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Barry Pizer
- Department of Oncology, Alder Hey Children's NHS Foundation Trust, Eaton Road, Liverpool, L12 2AP, UK.
| | - Amalia Schiavetti
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Antonio Pizzuti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Vincenzo Tombolini
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Cinzia Marchese
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Carlo Dominici
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesca Megiorni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| |
Collapse
|
6
|
Integrative Bayesian Analysis Identifies Rhabdomyosarcoma Disease Genes. Cell Rep 2019; 24:238-251. [PMID: 29972784 DOI: 10.1016/j.celrep.2018.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/29/2018] [Accepted: 06/01/2018] [Indexed: 12/15/2022] Open
Abstract
Identifying oncogenic drivers and tumor suppressors remains a challenge in many forms of cancer, including rhabdomyosarcoma. Anticipating gene expression alterations resulting from DNA copy-number variants to be particularly important, we developed a computational and experimental strategy incorporating a Bayesian algorithm and CRISPR/Cas9 "mini-pool" screen that enables both genome-scale assessment of disease genes and functional validation. The algorithm, called iExCN, identified 29 rhabdomyosarcoma drivers and suppressors enriched for cell-cycle and nucleic-acid-binding activities. Functional studies showed that many iExCN genes represent rhabdomyosarcoma line-specific or shared vulnerabilities. Complementary experiments addressed modes of action and demonstrated coordinated repression of multiple iExCN genes during skeletal muscle differentiation. Analysis of two separate cohorts revealed that the number of iExCN genes harboring copy-number alterations correlates with survival. Our findings highlight rhabdomyosarcoma as a cancer in which multiple drivers influence disease biology and demonstrate a generalizable capacity for iExCN to unmask disease genes in cancer.
Collapse
|
7
|
The PAX3-FOXO1 oncogene alters exosome miRNA content and leads to paracrine effects mediated by exosomal miR-486. Sci Rep 2019; 9:14242. [PMID: 31578374 PMCID: PMC6775163 DOI: 10.1038/s41598-019-50592-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/16/2019] [Indexed: 12/11/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children. The alveolar subtype (ARMS) is clinically more aggressive, and characterized by an oncogenic fusion protein PAX3-FOXO1 that drives oncogenic cellular properties. Exosomes are small, secreted vesicles that affect paracrine signaling. We show that PAX3-FOXO1 transcript alters exosome content of C2C12 myoblasts, leading to pro-tumorigenic paracrine effects in recipient cells. Microarray analysis revealed alteration in miRNA content of exosomes, affecting cellular networks involved in cell metabolism, growth signaling, and cellular invasion. Overexpression and knockdown studies showed that miR-486-5p is an effector of PAX3-FOXO1, and mediates its paracrine effects in exosomes, including promoting recipient cell migration, invasion, and colony formation. Analysis of human RMS cells showed miR-486-5p is enriched in both cells and exosomes, and to a higher extent in ARMS subtypes. Analysis of human serum samples showed that miR-486-5p is enriched in exosomes of patients with RMS, and follow-up after chemotherapy showed decrease to control values. Our findings identify a novel role of both PAX3-FOXO1 and its downstream effector miR-486-5p in exosome-mediated oncogenic paracrine effects of RMS, and suggest its possible use as a biomarker.
Collapse
|
8
|
An Examination of the Role of Transcriptional and Posttranscriptional Regulation in Rhabdomyosarcoma. Stem Cells Int 2017. [PMID: 28638414 PMCID: PMC5468592 DOI: 10.1155/2017/2480375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is an aggressive family of soft tissue tumors that most commonly manifests in children. RMS variants express several skeletal muscle markers, suggesting myogenic stem or progenitor cell origin of RMS. In this review, the roles of both recently identified and well-established microRNAs in RMS are discussed and summarized in a succinct, tabulated format. Additionally, the subtypes of RMS are reviewed along with the involvement of basic helix-loop-helix (bHLH) proteins, Pax proteins, and microRNAs in normal and pathologic myogenesis. Finally, the current and potential future treatment options for RMS are outlined.
Collapse
|
9
|
Ampofo E, Schmitt BM, Menger MD, Laschke MW. The regulatory mechanisms of NG2/CSPG4 expression. Cell Mol Biol Lett 2017; 22:4. [PMID: 28536635 PMCID: PMC5415841 DOI: 10.1186/s11658-017-0035-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/22/2017] [Indexed: 12/24/2022] Open
Abstract
Neuron-glial antigen 2 (NG2), also known as chondroitin sulphate proteoglycan 4 (CSPG4), is a surface type I transmembrane core proteoglycan that is crucially involved in cell survival, migration and angiogenesis. NG2 is frequently used as a marker for the identification and characterization of certain cell types, but little is known about the mechanisms regulating its expression. In this review, we provide evidence that the regulation of NG2 expression underlies inflammation and hypoxia and is mediated by methyltransferases, transcription factors, including Sp1, paired box (Pax) 3 and Egr-1, and the microRNA miR129-2. These regulatory factors crucially determine NG2-mediated cellular processes such as glial scar formation in the central nervous system (CNS) or tumor growth and metastasis. Therefore, they are potential targets for the establishment of novel NG2-based therapeutic strategies in the treatment of CNS injuries, cancer and other conditions of these types.
Collapse
Affiliation(s)
- Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Beate M Schmitt
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
10
|
Abstract
Rhabdomyosarcoma (RMS) is a myogenic tumor classified as the most frequent soft tissue sarcoma affecting children and adolescents. The histopathological classification includes 5 different histotypes, with 2 most predominant referred as to embryonal and alveolar, the latter being characterized by adverse outcome. The current molecular classification identifies 2 major subsets, those harboring the fused Pax3-Foxo1 transcription factor generating from a recurrent specific translocation (fusion-positive RMS), and those lacking this signature but harboring mutations in the RAS/PI3K/AKT signaling axis (fusion-negative RMS). Since little attention has been devoted to RMS metabolism until now, in this review we summarize the "state of art" of metabolism and discuss how some of the molecular signatures found in this cancer, as observed in other more common tumors, can predict important metabolic challenges underlying continuous cell growth, oxidative stress resistance and metastasis, which could be the subject of future targeted therapies.
Collapse
Affiliation(s)
- Eugenio Monti
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy
| | - Alessandro Fanzani
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy.,b Interuniversity Institute of Myology , Rome , Italy
| |
Collapse
|
11
|
TBX2 represses PTEN in rhabdomyosarcoma and skeletal muscle. Oncogene 2015; 35:4212-24. [PMID: 26686089 PMCID: PMC4916052 DOI: 10.1038/onc.2015.486] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 10/08/2015] [Accepted: 11/07/2015] [Indexed: 12/22/2022]
Abstract
Rhabdomyosarcoma (RMS) is the most frequent soft tissue sarcoma in children that shares many features of developing skeletal muscle. TBX2, a T-box family member, is highly up regulated in tumor cells of both major RMS subtypes where it functions as an oncogene. TBX2 is a repressor that is often over expressed in cancer cells and functions in bypassing cell growth control, including the repression of the cell cycle regulators p14 and p21. We have found that TBX2 directly represses the tumor suppressor PTEN in both RMS and normal muscle. Exogenous expression of TBX2 in normal muscle cells down regulates PTEN, and depletion or interference with TBX2 in RMS cells up regulates PTEN. Human RMS tumors show high levels of TBX2 and correspondingly low levels of PTEN. The expression of PTEN in clinical RMS samples is relatively uncharacterized and we establish that suppression of PTEN is a frequent event in both subtypes of RMS. TBX2 represses PTEN by directly binding to the promoter and recruiting the histone deacetylase, HDAC1. RMS cells have high levels of activated AKT due to the deregulation of PI3K signaling, and depletion or interference with TBX2, which up regulates PTEN, results in a reduction of phospho-AKT. We have also found that the highly related T-box family member TBX3 does not repress PTEN in the muscle lineage. This work suggests that TBX2 is a central component of the PTEN/PI3K/AKT signaling pathway deregulation in RMS cells and that targeting TBX2 in RMS tumors may offer a novel therapeutic approach for RMS.
Collapse
|
12
|
Dye DE, Medic S, Ziman M, Coombe DR. Melanoma biomolecules: independently identified but functionally intertwined. Front Oncol 2013; 3:252. [PMID: 24069584 PMCID: PMC3781348 DOI: 10.3389/fonc.2013.00252] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 09/09/2013] [Indexed: 01/31/2023] Open
Abstract
The majority of patients diagnosed with melanoma present with thin lesions and generally these patients have a good prognosis. However, 5% of patients with early melanoma (<1 mm thick) will have recurrence and die within 10 years, despite no evidence of local or metastatic spread at the time of diagnosis. Thus, there is a need for additional prognostic markers to help identify those patients that may be at risk of recurrent disease. Many studies and several meta-analyses have compared gene and protein expression in melanocytes, naevi, primary, and metastatic melanoma in an attempt to find informative prognostic markers for these patients. However, although a large number of putative biomarkers have been described, few of these molecules are informative when used in isolation. The best approach is likely to involve a combination of molecules. We believe one approach could be to analyze the expression of a group of interacting proteins that regulate different aspects of the metastatic pathway. This is because a primary lesion expressing proteins involved in multiple stages of metastasis may be more likely to lead to secondary disease than one that does not. This review focuses on five putative biomarkers – melanoma cell adhesion molecule (MCAM), galectin-3 (gal-3), matrix metalloproteinase 2 (MMP-2), chondroitin sulfate proteoglycan 4 (CSPG4), and paired box 3 (PAX3). The goal is to provide context around what is known about the contribution of these biomarkers to melanoma biology and metastasis. Although each of these molecules have been independently identified as likely biomarkers, it is clear from our analyses that each are closely linked with each other, with intertwined roles in melanoma biology.
Collapse
Affiliation(s)
- Danielle E Dye
- School of Biomedical Science & Curtin Health Innovation Research Institute, Faculty of Health, Curtin University , Perth, WA , Australia
| | | | | | | |
Collapse
|
13
|
Storer NY, White RM, Uong A, Price E, Nielsen GP, Langenau DM, Zon LI. Zebrafish rhabdomyosarcoma reflects the developmental stage of oncogene expression during myogenesis. Development 2013; 140:3040-50. [PMID: 23821038 DOI: 10.1242/dev.087858] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Rhabdomyosarcoma is a pediatric malignancy thought to arise from the uncontrolled proliferation of myogenic cells. Here, we have generated models of rhabdomyosarcoma in the zebrafish by inducing oncogenic KRAS(G12D) expression at different stages during muscle development. Several zebrafish promoters were used, including the cdh15 and rag2 promoters, which drive gene expression in early muscle progenitors, and the mylz2 promoter, which is expressed in differentiating myoblasts. The tumors that developed differed in their ability to recapitulate normal myogenesis. cdh15:KRAS(G12D) and rag2:KRAS(G12D) fish developed tumors that displayed an inability to complete muscle differentiation as determined by histological appearance and gene expression analyses. By contrast, mylz2:KRAS(G12D) tumors more closely resembled mature skeletal muscle and were most similar to well-differentiated human rhabdomyosarcoma in terms of gene expression. mylz2:KRAS(G12D) fish showed significantly improved survival compared with cdh15:KRAS(G12D) and rag2:KRAS(G12D) fish. Tumor-propagating activity was enriched in myf5-expressing cell populations within all of the tumor types. Our results demonstrate that oncogenic KRAS(G12D) expression at different stages during muscle development has profound effects on the ability of tumor cells to recapitulate normal myogenesis, altering the tumorigenic capability of these cells.
Collapse
Affiliation(s)
- Narie Y Storer
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Yin L, Cai WJ, Liu CX, Chen YZ, Hu JM, Jiang JF, Li HA, Cui XB, Chang XY, Zhang WJ, Sun K, Li F. Analysis of PTEN methylation patterns in soft tissue sarcomas by MassARRAY spectrometry. PLoS One 2013; 8:e62971. [PMID: 23690972 PMCID: PMC3656904 DOI: 10.1371/journal.pone.0062971] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 03/27/2013] [Indexed: 12/29/2022] Open
Abstract
Soft tissue sarcomas (STSs) are a rare and fascinating group of diseases that can be subdivided into specific reciprocal translocations in STSs (SRTSs) and nonspecific reciprocal translocations in STSs (NRTSs). PTEN mutations are rare in STSs, suggesting that PTEN expression may be lost by alternative mechanisms such as methylation. In order to reveal whether aberrant PTEN methylation occurs in STSs, MassARRAY Spectrometry was carried to detect methylation patterns of PTEN in STSs. We evaluated methylation levels in 41 CpG sites from −2,515 to −2,186 bp (amplicon A) and −1,786 to −1,416 bp (amplicon B) relative to the translation initiation site in 110 different cases (46 cases of SRTSs, 40 cases of NRTSs, and 24 cases of normal controls). In addition, immunohistochemistry (IHC) was used to detect the loss of PTEN to determine whether PTEN alterations were responsible for decreased PTEN expression. Our data showed that expression of PTEN was diminished in 49 (57%) STSs, whereas the remaining cases (43%) were classified as high expression. Our previous results found that only 2 of 86 cases (2.3%) had a PTEN mutation suggesting that PTEN may be mainly downregulated in STSs by methylation, but not by mutation of PTEN itself. We observed that amplicon A was hypermethylated in STSs with low PTEN expression, whereas normal controls had low methylation levels (P<0.0001), which was not present in amplicon B (P>0.05), nor were there significant differences in the methylation levels in PTEN between SRTS and NRTS cases. The majority of individual CpG units within two amplicons was demonstrated to be hypermethylated. These findings indicate that PTEN hypermethylation is a common event in STSs suggesting that the inactivation of PTEN may be due to hypermethylation in the promoter of PTEN. The aberrant methylation of the CpG sites within PTEN promoter may serve as a potential candidate biomarker for STSs.
Collapse
Affiliation(s)
- Liang Yin
- Department of Pathology and Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Wei-Juan Cai
- Department of Clinical Laboratory, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Chun-Xia Liu
- Department of Pathology and Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Yun-Zhao Chen
- Department of Pathology and Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Jian-Ming Hu
- Department of Pathology and Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Jin-Fang Jiang
- Department of Pathology and Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Hong-An Li
- Department of Pathology and Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Xiao-Bin Cui
- Department of Pathology and Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Xiang-Yun Chang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Wen Jie Zhang
- Department of Pathology and Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
| | - Kan Sun
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
- * E-mail: (KS); (FL)
| | - Feng Li
- Department of Pathology and Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, P.R. China
- * E-mail: (KS); (FL)
| |
Collapse
|
15
|
Sokolowski E, Turina CB, Kikuchi K, Langenau DM, Keller C. Proof-of-concept rare cancers in drug development: the case for rhabdomyosarcoma. Oncogene 2013; 33:1877-89. [PMID: 23665679 DOI: 10.1038/onc.2013.129] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 02/22/2013] [Accepted: 02/27/2013] [Indexed: 12/14/2022]
Abstract
Rare diseases typically affect fewer than 200,000 patients annually, yet because thousands of rare diseases exist, the cumulative impact is millions of patients worldwide. Every form of childhood cancer qualifies as a rare disease-including the childhood muscle cancer, rhabdomyosarcoma (RMS). The next few years promise to be an exceptionally good era of opportunity for public-private collaboration for rare and childhood cancers. Not only do certain governmental regulation advantages exist, but these advantages are being made permanent with special incentives for pediatric orphan drug-product development. Coupled with a growing understanding of sarcoma tumor biology, synergy with pharmaceutical muscle disease drug-development programs, and emerging publically available preclinical and clinical tools, the outlook for academic-community-industry partnerships in RMS drug development looks promising.
Collapse
Affiliation(s)
- E Sokolowski
- Department of Student Affairs, Oregon State University, Corvallis, OR, USA
| | - C B Turina
- 1] Department of Student Affairs, Oregon State University, Corvallis, OR, USA [2] Pediatric Cancer Biology Program, Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR, USA
| | - K Kikuchi
- Pediatric Cancer Biology Program, Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR, USA
| | - D M Langenau
- 1] Division of Molecular Pathology and Cancer Center, Massachusetts General Hospital, Boston, MA, USA [2] Harvard Medical School and Harvard Stem Cell Institute, Boston, MA, USA
| | - C Keller
- Pediatric Cancer Biology Program, Department of Pediatrics, Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
16
|
Loh AHP, Brennan RC, Lang WH, Hickey RJ, Malkas LH, Sandoval JA. Dissecting the PI3K Signaling Axis in Pediatric Solid Tumors: Novel Targets for Clinical Integration. Front Oncol 2013; 3:93. [PMID: 23638435 PMCID: PMC3636761 DOI: 10.3389/fonc.2013.00093] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 04/05/2013] [Indexed: 12/19/2022] Open
Abstract
Children with solid tumors represent a unique population. Recent improvements in pediatric solid tumor survival rates have been confined to low- and moderate-risk cancers, whereas minimal to no notable improvement in survival have been observed in high-risk and advanced-stage childhood tumors. Treatments for patients with advanced disease are rarely curative, and responses to therapy are often followed by relapse, which highlights the large unmet need for novel therapies. Recent advances in cancer treatment have focused on personalized therapy, whereby patients are treated with agents that best target the molecular drivers of their disease. Thus, a better understanding of the pathways that drive cancer or drug resistance is of critical importance. One such example is the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway, which is activated in many solid cancer patients and represents a target for therapy. PI3K/Akt/mTOR pathway activation has also been observed in tumors resistant to agents targeting upstream receptor tyrosine kinases (RTKs). Agents that target this pathway have the potential to shut down survival pathways, and are being explored both in the setting of pathway-activating mutations and for their ability to restore sensitivity to upstream signaling targeted agents. Here, we examine the role of the PI3K/Akt/mTOR pathway in pediatric solid tumors, review the novel agents being explored to target this pathway, and explore the potential role of the inhibition of this pathway in the clinical development of these agents in children.
Collapse
Affiliation(s)
- Amos H P Loh
- Department of Surgery, St. Jude Children's Research Hospital Memphis, TN, USA
| | | | | | | | | | | |
Collapse
|
17
|
Rengaswamy V, Kontny U, Rössler J. New approaches for pediatric rhabdomyosarcoma drug discovery: targeting combinatorial signaling. Expert Opin Drug Discov 2011; 6:1103-25. [PMID: 22646865 DOI: 10.1517/17460441.2011.611498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Rhabdomyosarcomas (RMS) are rare heterogeneous pediatric tumors that are treated by surgery, chemotherapy and irradiation. New therapeutic approaches are needed, especially in the advanced stages to target the pro-oncogenic signals. Exploring the molecular interactions of the regulatory signals and their roles in the developmental aspects of different subtypes of RMS is essential to identify potential targets and develop new therapeutic drugs. AREAS COVERED Insights into different drug discovery approaches are discussed with specific emphasis on gene expression profiling, fusion protein, role of small interfering RNA (siRNA)- and microRNA (miRNA)-based discovery approaches, targeting cancer stem cells, and in vitro and in vivo model systems. Targeting some overexpressed signals along with the possibilities of combination therapy of validated drug targets is discussed. Additionally, methods to overcome the limitations of discovery-based research are briefly discussed. EXPERT OPINION Due to drug resistance, ineffective therapy in advanced stages and relapse, there is a demand to explore new drug targets and discovery approaches. Implementing miRNA-based profiling would reveal the extent of miR-based regulation, various biomarkers and potential targets in RMS. A suitable combination of innovative techniques and the use of model systems might assist the identification and validation of novel targets and drug discovery methods. Combining specific drugs along with type-specific target inhibition of overexpressed mRNAs through siRNA approaches would enable the development of personalized therapy.
Collapse
Affiliation(s)
- Venkatesh Rengaswamy
- University Hospital Freiburg, Center for Pediatrics and Adolescent Medicine, Clinic IV: Pediatric Hematology and Oncology, Mathildenstr. 1, 79106 Freiburg , Germany +49 761 270 43000 ; +49 761 270 45180 ;
| | | | | |
Collapse
|
18
|
Abstract
Rhabdomyosarcomas (RSCs) are skeletal muscle neoplasms found in humans and domestic mammals. The A/J inbred strain developed a high frequency (between 70–80%) of adult pleomorphic type (APT) RSC at >20 months of age while BALB/cByJ also develop RSC but less frequently. These neoplasms invaded skeletal muscle surrounding either the axial or proximal appendicular skeleton and were characterized by pleomorphic cells with abundant eosinophilic cytoplasm, multiple nuclei, and cross striations. The diagnosis was confirmed by detection of alpha-sarcomeric actin and myogenin in the neoplastic cells using immunocytochemistry. The A/J strain, but not the related BALB/c substrains, is also characterised by a progressive muscular dystrophy homologous to limb-girdle muscular dystrophy type 2B. The association between the development of RSC in similar muscle groups to those most severely affected by the progressive muscular dystrophy suggested that these neoplasms developed from abnormal regeneration of the skeletal muscle exacerbated by the dysferlin mutation. Transcriptome analyses of RSCs revealed marked downregulation of genes in muscular development and function signaling networks. Non-synonymous coding SNPs were found in Myl1, Abra, Sgca, Ttn, and Kcnj12 suggesting these may be important in the pathogenesis of RSC. These studies suggest that A strains of mice can be useful models for dissecting the molecular genetic basis for development, progression, and ultimately for testing novel anticancer therapeutic agents dealing with rhabdomyosarcoma.
Collapse
|
19
|
Medic S, Rizos H, Ziman M. Differential PAX3 functions in normal skin melanocytes and melanoma cells. Biochem Biophys Res Commun 2011; 411:832-7. [PMID: 21802410 DOI: 10.1016/j.bbrc.2011.07.053] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 07/14/2011] [Indexed: 12/17/2022]
Abstract
The PAX3 transcription factor is the key regulator of melanocyte development during embryogenesis and is also frequently found in melanoma cells. While PAX3 is known to regulate melanocyte differentiation, survival, proliferation and migration during development, it is not clear if its function is maintained in adult melanocytes and melanoma cells. To clarify this we have assessed which genes are targeted by PAX3 in these cells. We show here that similar to its roles in development, PAX3 regulates complex differentiation networks in both melanoma cells and melanocytes, in order to maintain cells as "stem" cell-like (via NES and SOX9). We show also that mediators of migration (MCAM and CSPG4) are common to both cell types but more so in melanoma cells. By contrast, PAX3-mediated regulation of melanoma cell proliferation (through TPD52) and survival (via BCL2L1 and PTEN) differs from that in melanocytes. These results suggest that by controlling cell proliferation, survival and migration as well as maintaining a less differentiated "stem" cell like phenotype, PAX3 may contribute to melanoma development and progression.
Collapse
Affiliation(s)
- Sandra Medic
- School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | | | | |
Collapse
|
20
|
Hathaway JD, Haque A. Insights into the Role of PAX-3 in the Development of Melanocytes and Melanoma. ACTA ACUST UNITED AC 2011; 4:1-6. [PMID: 24790680 DOI: 10.2174/1874079001104010001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Melanoma is the deadliest form of skin cancer in the United States with an increasing prevalence. However, the development of melanoma from a melanocyte precursor is still poorly defined. Understanding the molecules responsible for melanoma progression may lead to improved targeted therapy. One potential molecule is the paired box-3 (PAX-3) protein, which has been implicated in the development of melanocytes and malignant melanoma. In melanoma, the expression of PAX-3 is believed to be differentially regulated, and has been linked with malignancies and staging of the disease. The loss of PAX-3 regulation has also been associated with the loss of transforming growth factor-beta (TGF-β) activity, but its effect on PAX-3 in differentiated melanocytes as well as metastatic melanoma remains unclear. Understanding PAX-3 regulation could potentially shift melanoma to a less aggressive and less metastatic disease. This review summarizes our current knowledge on PAX-3 during melanocyte development, its regulation, and its implications in the development of novel chemo-immunotherapeutics against metastatic melanoma.
Collapse
Affiliation(s)
- Jessica Diann Hathaway
- Department of Microbiology and Immunology, Charles Darby Children's Research Institute, and Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425
| | - Azizul Haque
- Department of Microbiology and Immunology, Charles Darby Children's Research Institute, and Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425
| |
Collapse
|
21
|
Abstract
The PAX3 transcription factor is critical for the proper development of neural crest lineages including melanocytes. These cells show continued PAX3 expression from formation to differentiation. While many expression, misexpression and mutation studies clarify the importance of PAX3 in melanocyte development, less well understood, and more perplexing, is the continued PAX3 expression in the adult skin. In this article we explore the multiple roles of PAX3 in melanocyte genesis, and draw on evidence from expression in developing melanoblasts, adult melanocytes and melanocyte stem cells. From this, we present a more encompassing theory that PAX3 is a key regulator of the myriad steps in melanocytic cell determination. These roles may be accomplished by differential association with cofactors, via alternate transcripts or posttranslational protein modification(s). In light of the plethora of information gleaned from development we then consider its roles in melanoma and provide here a comprehensive consideration of the significance of PAX3 expression in melanoma. PAX3 and Pax3 indicate human and mouse transcription factors respectively.
Collapse
Affiliation(s)
- Sandra Medic
- School of Exercise, Biomedical and Health Sciences, Edith Cowan University, 100 Joondalup Drive, Joondalup, Perth, Western Australia
| | | |
Collapse
|
22
|
Fernandez K, Serinagaoglu Y, Hammond S, Martin LT, Martin PT. Mice lacking dystrophin or alpha sarcoglycan spontaneously develop embryonal rhabdomyosarcoma with cancer-associated p53 mutations and alternatively spliced or mutant Mdm2 transcripts. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:416-34. [PMID: 20019182 DOI: 10.2353/ajpath.2010.090405] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Altered expression of proteins in the dystrophin-associated glycoprotein complex results in muscular dystrophy and has more recently been implicated in a number of forms of cancer. Here we show that loss of either of two members of this complex, dystrophin in mdx mice or alpha sarcoglycan in Sgca(-/-) mice, results in the spontaneous development of muscle-derived embryonal rhabdomyosarcoma (RMS) after 1 year of age. Many mdx and Sgca(-/-) tumors showed increased expression of insulin-like growth factor 2, retinoblastoma protein, and phosphorylated Akt and decreased expression of phosphatase and tensin homolog gene, much as is found in a human RMS. Further, all mdx and Sgca(-/-) RMS analyzed had increased expression of p53 and murine double minute (mdm)2 protein and contained missense p53 mutations previously identified in human cancers. The mdx RMS also contained missense mutations in Mdm2 or alternatively spliced Mdm2 transcripts that lacked an exon encoding a portion of the p53-binding domain. No Pax3:Fkhr or Pax7:Fkhr translocation mRNA products were evident in any tumor. Expression of natively glycosylated alpha dystroglycan and alpha sarcoglycan was reduced in mdx RMS, whereas dystrophin expression was absent in almost all human RMS, both for embryonal and alveolar RMS subtypes. These studies show that absence of members of the dystrophin-associated glycoprotein complex constitutes a permissive environment for spontaneous development of embryonal RMS associated with mutation of p53 and mutation or altered splicing of Mdm2.
Collapse
Affiliation(s)
- Karen Fernandez
- Division of Hematology/Oncology, Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | | | | | | | | |
Collapse
|
23
|
De Giovanni C, Landuzzi L, Nicoletti G, Lollini PL, Nanni P. Molecular and cellular biology of rhabdomyosarcoma. Future Oncol 2009; 5:1449-75. [DOI: 10.2217/fon.09.97] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Rhabdomyosarcoma is a group of soft-tissue sarcomas that share features of skeletal myogenesis, but show extensive heterogeneity in histology, age and site of onset, and prognosis. This review matches recent molecular data with biological features of rhabdomyosarcoma. Alterations in molecular pathways, animal models, cell of origin and potential new therapeutic targets are discussed.
Collapse
Affiliation(s)
- Carla De Giovanni
- Department of Experimental Pathology, Cancer Research Section, University of Bologna, Bologna, Italy
| | - Lorena Landuzzi
- Laboratory of Experimental Oncology, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Giordano Nicoletti
- Laboratory of Experimental Oncology, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Pier-Luigi Lollini
- Department of Hematology and Oncological Sciences ‘L. e A. Seragnoli’, Viale Filopanti 22, Bologna 40126, Italy
| | - Patrizia Nanni
- Department of Experimental Pathology, Cancer Research Section, University of Bologna, Bologna, Italy
| |
Collapse
|
24
|
Charytonowicz E, Cordon-Cardo C, Matushansky I, Ziman M. Alveolar rhabdomyosarcoma: Is the cell of origin a mesenchymal stem cell? Cancer Lett 2009; 279:126-36. [DOI: 10.1016/j.canlet.2008.09.039] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 09/26/2008] [Accepted: 09/30/2008] [Indexed: 12/26/2022]
|
25
|
Makawita S, Ho M, Durbin AD, Thorner PS, Malkin D, Somers GR. Expression of insulin-like growth factor pathway proteins in rhabdomyosarcoma: IGF-2 expression is associated with translocation-negative tumors. Pediatr Dev Pathol 2009; 12:127-35. [PMID: 18788888 DOI: 10.2350/08-05-0477.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Accepted: 07/29/2008] [Indexed: 01/08/2023]
Abstract
Recent studies have shown a significant involvement of insulin-like growth factor (IGF) signaling components in the pathogenesis of rhabdomyosarcoma (RMS). Furthermore, there has been some evidence to indicate that differential expression of IGF pathway genes can distinguish RMS subtypes. The present study utilized immunohistochemistry to determine the expression patterns of IGF1, IGF2, IGF binding protein 2 (IGFBP2), IGF receptor 1 (IGF1R), and IGF receptor 2 (IGF2R) in 24 embryonal RMS (ERMS) and 8 alveolar RMS (ARMS). A majority of tumors were positive for IGF2, IGFBP2, IGF1R, and IGF2R and negative for IGF1 expression. However, only IGF2 showed a significant difference in expression between the ERMS and ARMS subtypes, with higher levels of expression in ERMS (P = 0.0003). Within the ARMS subtype, IGF2 positivity was limited to PAX/FKHR translocation-negative tumors. The staining pattern for all 5 proteins was diffuse cytoplasmic in the majority of tumors. Analysis of RMS cell lines by real-time reverse transcriptase-polymerase chain reaction for IGF2 expression revealed significantly higher mean expression levels in ERMS and translocation-negative ARMS cell lines when compared to translocation-positive ARMS cell lines (P = 0.0027). Stable introduction of PAX3/FKHR into an ERMS cell line also demonstrated a significant reduction in IGF2 expression. The results of this study show that expression of the IGF2 ligand is associated with translocation-negative tumors and may serve as a diagnostic aid in distinguishing RMS subtypes. Furthermore, the in vitro results are supportive of a role for the PAX3/FKHR fusion gene in the inhibition of IGF2 expression.
Collapse
Affiliation(s)
- Shalini Makawita
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
26
|
Kubic JD, Young KP, Plummer RS, Ludvik AE, Lang D. Pigmentation PAX-ways: the role of Pax3 in melanogenesis, melanocyte stem cell maintenance, and disease. Pigment Cell Melanoma Res 2009; 21:627-45. [PMID: 18983540 DOI: 10.1111/j.1755-148x.2008.00514.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Transcription factors initiate programs of gene expression and are catalysts in downstream molecular cascades that modulate a variety of cellular processes. Pax3 is a transcription factor that is important in the melanocyte and influences melanocytic proliferation, resistance to apoptosis, migration, lineage specificity and differentiation. In this review, we focus on Pax3 and the molecular pathways that Pax3 is a part of during melanogenesis and in the melanocyte stem cell. These roles of Pax3 are emphasized during the development of diseases and syndromes resulting from either too much or too little Pax3 function. Due to its key task in melanocyte stem cells and tumors, the Pax3 pathway may provide an ideal target for either stem cell or cancer therapies.
Collapse
Affiliation(s)
- Jennifer D Kubic
- Section of Dermatology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | | | | | | |
Collapse
|
27
|
Abstract
The paired box genes are a family of nine developmental control genes, which in human beings (PAX) and mice (Pax) encode nuclear transcription factors. The temporal and spatial expressions of these highly conserved genes are tightly regulated during foetal development including organogenesis. PAY/Paxgenes are switched off during the terminal differentiation of most structures. Specific mutations within a number of PAX/Pax genes lead to developmental abnormalities in both human beings and mice. Mutation in PAX3 causes Waardenburg syndrome, and craniofacial-deafness-hand syndrome. The Splotch phenotype in mouse exhibits defects in neural crest derivatives such as, pigment cells, sympathetic ganglia and cardiac neural crest-derived structures. The PAX family also plays key roles in several human malignancies. In particular, PAX3 is involved in rhabdomyosarcoma and tumours of neural crest origin, including melanoma and neuroblastoma. This review critically evaluates the roles of PAX/Pax in oncogenesis. It especially highlights recent advances in knowledge of how their genetic alterations directly interfere in the transcriptional networks that regulate cell differentiation, proliferation, migration and survival and may contribute to oncogenesis.
Collapse
Affiliation(s)
- Qiuyu Wang
- School of Biology, Chemistry and Health Science, Manchester Metropolitan University, and Department of Pathology Sciences, Christie Hospital, Manchester, United Kingdom
| | | | | | | | | | | |
Collapse
|