1
|
Zou Z, Zhong L. Anaplastic thyroid cancer: Genetic roles, targeted therapy, and immunotherapy. Genes Dis 2025; 12:101403. [PMID: 40271195 PMCID: PMC12018003 DOI: 10.1016/j.gendis.2024.101403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/02/2024] [Accepted: 08/02/2024] [Indexed: 04/25/2025] Open
Abstract
Anaplastic thyroid cancer (ATC) stands as the most formidable form of thyroid malignancy, presenting a persistent challenge in clinical management. Recent years have witnessed a gradual unveiling of the intricate genetic underpinnings governing ATC through next-generation sequencing. The emergence of this genetic landscape has paved the way for the exploration of targeted therapies and immunotherapies in clinical trials. Despite these strides, the precise mechanisms governing ATC pathogenesis and the identification of efficacious treatments demand further investigation. Our comprehensive review stems from an extensive literature search focusing on the genetic implications, notably the pivotal MAPK and PI3K-AKT-mTOR signaling pathways, along with targeted therapies and immunotherapies in ATC. Moreover, we screen and summarize the advances and challenges in the current diagnostic approaches for ATC, including the invasive tissue sampling represented by fine needle aspiration and core needle biopsy, immunohistochemistry, and 18F-fluorodeoxyglucose positron emission tomography/computed tomography. We also investigate enormous studies on the prognosis of ATC and outline independent prognostic factors for future clinical assessment and therapy for ATC. By synthesizing this literature, we aim to encapsulate the evolving landscape of ATC oncology, potentially shedding light on novel pathogenic mechanisms and avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zhao Zou
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Linhong Zhong
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging and Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
2
|
Pita JM, Raspé E, Coulonval K, Decaussin-Petrucci M, Tarabichi M, Dom G, Libert F, Craciun L, Andry G, Wicquart L, Leteurtre E, Trésallet C, Marlow LA, Copland JA, Durante C, Maenhaut C, Cavaco BM, Dumont JE, Costante G, Roger PP. CDK4 phosphorylation status and rational use for combining CDK4/6 and BRAF/MEK inhibition in advanced thyroid carcinomas. Front Endocrinol (Lausanne) 2023; 14:1247542. [PMID: 37964967 PMCID: PMC10641312 DOI: 10.3389/fendo.2023.1247542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/26/2023] [Indexed: 11/16/2023] Open
Abstract
Background CDK4/6 inhibitors (CDK4/6i) have been established as standard treatment against advanced Estrogen Receptor-positive breast cancers. These drugs are being tested against several cancers, including in combinations with other therapies. We identified the T172-phosphorylation of CDK4 as the step determining its activity, retinoblastoma protein (RB) inactivation, cell cycle commitment and sensitivity to CDK4/6i. Poorly differentiated (PDTC) and anaplastic (ATC) thyroid carcinomas, the latter considered one of the most lethal human malignancies, represent major clinical challenges. Several molecular evidence suggest that CDK4/6i could be considered for treating these advanced thyroid cancers. Methods We analyzed by two-dimensional gel electrophoresis the CDK4 modification profile and the presence of T172-phosphorylated CDK4 in a collection of 98 fresh-frozen tissues and in 21 cell lines. A sub-cohort of samples was characterized by RNA sequencing and immunohistochemistry. Sensitivity to CDK4/6i (palbociclib and abemaciclib) was assessed by BrdU incorporation/viability assays. Treatment of cell lines with CDK4/6i and combination with BRAF/MEK inhibitors (dabrafenib/trametinib) was comprehensively evaluated by western blot, characterization of immunoprecipitated CDK4 and CDK2 complexes and clonogenic assays. Results CDK4 phosphorylation was detected in all well-differentiated thyroid carcinomas (n=29), 19/20 PDTC, 16/23 ATC and 18/21 thyroid cancer cell lines, including 11 ATC-derived ones. Tumors and cell lines without phosphorylated CDK4 presented very high p16CDKN2A levels, which were associated with proliferative activity. Absence of CDK4 phosphorylation in cell lines was associated with CDK4/6i insensitivity. RB1 defects (the primary cause of intrinsic CDK4/6i resistance) were not found in 5/7 tumors without detectable phosphorylated CDK4. A previously developed 11-gene expression signature identified the likely unresponsive tumors, lacking CDK4 phosphorylation. In cell lines, palbociclib synergized with dabrafenib/trametinib by completely and permanently arresting proliferation. These combinations prevented resistance mechanisms induced by palbociclib, most notably Cyclin E1-CDK2 activation and a paradoxical stabilization of phosphorylated CDK4 complexes. Conclusion Our study supports further clinical evaluation of CDK4/6i and their combination with anti-BRAF/MEK therapies as a novel effective treatment against advanced thyroid tumors. Moreover, the complementary use of our 11 genes predictor with p16/KI67 evaluation could represent a prompt tool for recognizing the intrinsically CDK4/6i insensitive patients, who are potentially better candidates to immediate chemotherapy.
Collapse
Affiliation(s)
- Jaime M. Pita
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Eric Raspé
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Katia Coulonval
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Maxime Tarabichi
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Geneviève Dom
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Frederick Libert
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
- BRIGHTCore, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Ligia Craciun
- Tumor Bank of the Institut Jules Bordet Comprehensive Cancer Center – Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Guy Andry
- Department of Head & Neck and Thoracic Surgery, Institut Jules Bordet Comprehensive Cancer Center – Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Laurence Wicquart
- Tumorothèque du Groupement de Coopération Sanitaire-Centre Régional de Référence en Cancérologie (C2RC) de Lille, Lille, France
| | - Emmanuelle Leteurtre
- Department of Pathology, Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Inserm, Centre Hospitalo-Universitaire (CHU) Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Christophe Trésallet
- Department of General and Endocrine Surgery - Pitié-Salpêtrière Hospital, Sorbonne University, Assistance Publique des Hôpitaux de Paris, Paris, France
- Department of Digestive, Bariatric and Endocrine Surgery - Avicenne University Hospital, Paris Nord - Sorbonne University, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Laura A. Marlow
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - John A. Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| | - Cosimo Durante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Carine Maenhaut
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Branca M. Cavaco
- Molecular Endocrinology Group, Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Lisbon, Portugal
| | - Jacques E. Dumont
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Giuseppe Costante
- Departments of Endocrinology and Medical Oncology, Institut Jules Bordet Comprehensive Cancer Center – Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pierre P. Roger
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM) and Université Libre de Bruxelles (ULB)-Cancer Research Center (U-CRC), Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
3
|
Xing X, Liu M, Wang X, Guo Q, Wang H. Promoting effects of calponin 3 on the growth of diffuse large B‑cell lymphoma cells. Oncol Rep 2023; 49:46. [PMID: 36660952 PMCID: PMC9868891 DOI: 10.3892/or.2023.8483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Diffuse large B‑cell lymphoma (DLBCL) is one of the most common types of lymphoma. Calponin 3 (CNN3) is a thin filament‑associated protein previously known to regulate smooth muscle contraction. Recent evidence illustrates its involvement in carcinogenesis; however, its roles in DLBCL remain unknown. CNN3 was found to be highly expressed in DLBCL specimens according to the online Gene Expression Profiling Interactive Analysis data. The aim of the present study was to investigate the roles of CNN3 in the progression of DLBCL. In vitro, the ectopic expression of CNN3 promoted the proliferation and G1/S transition of DLBCL cells, while its silencing led to opposite alterations. A similar tumor‑promoting role of CNN3 was also demonstrated by injecting nude mice with DLBCL cells over‑ or underexpressing CNN3. The results of dual‑luciferase reporter and chromatin immunoprecipitation assays revealed that forkhead box O3 (FOXO3), a known tumor suppressor in DLBCL, bound to the CNN3 promoter at ‑1955/‑1948 and ‑1190/‑1183, and suppressed the transcription of CNN3. The alterations induced by FOXO3 were partly blocked by CNN3 overexpression. On the whole, the present study demonstrates that CNN3, whose transcriptional activity is negatively regulated by FOXO3, contributes to the malignant behavior of DLBCL cells. The findings of the present study may provide novel diagnostic or therapeutic insight for DLBCL in clinical practice.
Collapse
Affiliation(s)
- Xiaojing Xing
- Department of Hematology and Breast Cancer, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute), Shenyang, Liaoning 110042, P.R. China,Correspondence to: Dr Xiaojing Xing, Department of Hematology and Breast Cancer, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute), 44 Xiaoheyan Road, Shenyang, Liaoning 110042, P.R. China, E-mail:
| | - Meichen Liu
- Department of Hematology and Breast Cancer, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute), Shenyang, Liaoning 110042, P.R. China
| | - Xuguang Wang
- Department of Pathology, Shenyang Medical College, Shenyang, Liaoning 110034, P.R. China
| | - Qianxue Guo
- Department of Hematology and Breast Cancer, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute), Shenyang, Liaoning 110042, P.R. China
| | - Hongyue Wang
- Department of Scientific Research and Academic, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital and Institute), Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
4
|
Kohoutova K, Dočekal V, Ausserlechner MJ, Kaiser N, Tekel A, Mandal R, Horvath M, Obsilova V, Vesely J, Hagenbuchner J, Obsil T. Lengthening the Guanidine-Aryl Linker of Phenylpyrimidinylguanidines Increases Their Potency as Inhibitors of FOXO3-Induced Gene Transcription. ACS OMEGA 2022; 7:34632-34646. [PMID: 36188303 PMCID: PMC9521028 DOI: 10.1021/acsomega.2c04613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/07/2022] [Indexed: 06/16/2023]
Abstract
Increased FOXO3 nuclear localization is involved in neuroblastoma chemoresistance and tumor angiogenesis. Accordingly, FOXO3 inhibition is a promising strategy for boosting antitumor immune responses and suppressing FOXO3-mediated therapy resistance in cancer cells. However, no FOXO3 inhibitors are currently available for clinical use. Nevertheless, we have recently identified (4-propoxy)phenylpyrimidinylguanidine as a FOXO3 inhibitor in cancer cells in the low micromolar range. Here, we report the synthesis and structure-activity relationship study of a small library of its derivatives, some of which inhibit FOXO3-induced gene transcription in cancer cells in a submicromolar range and are thus 1 order of magnitude more potent than their parent compound. By NMR and molecular docking, we showed that these compounds differ in their interactions with the DNA-binding domain of FOXO3. These results may provide a foundation for further optimizing (4-propoxy)phenylpyrimidinylguanidine and developing therapeutics for inhibiting the activity of forkhead box (FOX) transcription factors and their interactions with other binding partners.
Collapse
Affiliation(s)
- Klara Kohoutova
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
- Institute
of Physiology of the Czech Academy of Sciences, Laboratory of Structural
Biology of Signaling Proteins, Division
BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic
| | - Vojtěch Dočekal
- Department
of Organic Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | | | - Nora Kaiser
- Department
of Pediatrics I, Medical University Innsbruck, Innrain 66, Innsbruck 6020, Austria
| | - Andrej Tekel
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | - Raju Mandal
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | - Matej Horvath
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | - Veronika Obsilova
- Institute
of Physiology of the Czech Academy of Sciences, Laboratory of Structural
Biology of Signaling Proteins, Division
BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic
| | - Jan Vesely
- Department
of Organic Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
| | - Judith Hagenbuchner
- Department
of Pediatrics II, Medical University Innsbruck, Innrain 66, Innsbruck 6020, Austria
| | - Tomas Obsil
- Department
of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Albertov 6, Prague 12843, Czech Republic
- Institute
of Physiology of the Czech Academy of Sciences, Laboratory of Structural
Biology of Signaling Proteins, Division
BIOCEV, Prumyslova 595, Vestec 25250, Czech Republic
| |
Collapse
|
5
|
Datta N, Chakraborty S, Basu M, Ghosh MK. Tumor Suppressors Having Oncogenic Functions: The Double Agents. Cells 2020; 10:cells10010046. [PMID: 33396222 PMCID: PMC7824251 DOI: 10.3390/cells10010046] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/23/2020] [Accepted: 12/25/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer progression involves multiple genetic and epigenetic events, which involve gain-of-functions of oncogenes and loss-of-functions of tumor suppressor genes. Classical tumor suppressor genes are recessive in nature, anti-proliferative, and frequently found inactivated or mutated in cancers. However, extensive research over the last few years have elucidated that certain tumor suppressor genes do not conform to these standard definitions and might act as “double agents”, playing contrasting roles in vivo in cells, where either due to haploinsufficiency, epigenetic hypermethylation, or due to involvement with multiple genetic and oncogenic events, they play an enhanced proliferative role and facilitate the pathogenesis of cancer. This review discusses and highlights some of these exceptions; the genetic events, cellular contexts, and mechanisms by which four important tumor suppressors—pRb, PTEN, FOXO, and PML display their oncogenic potentials and pro-survival traits in cancer.
Collapse
Affiliation(s)
- Neerajana Datta
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India; (N.D.); (S.C.)
| | - Shrabastee Chakraborty
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India; (N.D.); (S.C.)
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24 Paraganas, West Bengal PIN-743372, India;
| | - Mrinal K. Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India; (N.D.); (S.C.)
- Correspondence:
| |
Collapse
|
6
|
Farhan M, Silva M, Li S, Yan F, Fang J, Peng T, Hu J, Tsao M, Little P, Zheng W. The role of FOXOs and autophagy in cancer and metastasis-Implications in therapeutic development. Med Res Rev 2020; 40:2089-2113. [PMID: 32474970 PMCID: PMC7586888 DOI: 10.1002/med.21695] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 04/21/2020] [Accepted: 05/16/2020] [Indexed: 12/17/2022]
Abstract
Autophagy is a highly conserved intracellular degradation process that plays a crucial role in cell survival and stress reactions as well as in cancer development and metastasis. Autophagy process involves several steps including sequestration, fusion of autophagosomes with lysosomes and degradation. Forkhead box O (FOXO) transcription factors regulate the expression of genes involved in cellular metabolic activity and signaling pathways of cancer growth and metastasis. Recent evidence suggests that FOXO proteins are also involved in autophagy regulation. The relationship among FOXOs, autophagy, and cancer has been drawing attention of many who work in the field. This study summarizes the role of FOXO proteins and autophagy in cancer growth and metastasis and analyzes their potential roles in cancer disease management.
Collapse
Affiliation(s)
- Mohd Farhan
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Marta Silva
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Shuai Li
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Fengxia Yan
- Department of MedicineJinan UniversityGuangzhouChina
| | - Jiankang Fang
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Tangming Peng
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Jim Hu
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Ming‐Sound Tsao
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Peter Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Wenhua Zheng
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| |
Collapse
|
7
|
Shah KK, Neff JL, Erickson LA, Jackson RA, Jenkins SM, Mansfield AS, Moser JC, Harris AL, Copland JA, Halling KC, Flotte TJ. Correlation of novel ALK ATI with ALK immunohistochemistry and clinical outcomes in metastatic melanoma. Histopathology 2020; 77:601-610. [PMID: 32564377 DOI: 10.1111/his.14191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/15/2020] [Indexed: 01/03/2023]
Abstract
AIMS Recently, a novel isoform of anaplastic lymphoma kinase, with alternative transcription initiation (ALKATI ), has been described in melanoma and is susceptible to targeted ALK-inhibitor therapy. Clinical outcomes of patients with ALKATI mutated melanoma as well as correlation with immunohistochemical (IHC) methods have not yet been described. METHODS AND RESULTS Clinicopathological characteristics were abstracted for 324 patients with metastatic melanoma (MM). IHC, fluorescence in-situ hybridisation and RNA-based digital molecular analysis assays were performed on archival tissue from 173 stage III and 192 stage IV tumours. ALKATI was identified in 12.7 and 4.8% stage III and IV tumours, respectively. Discrete presentations of the ALKATI are seen: isolated ALKATI (n = 20) and mixed ALKATI (combined ALKATI and ALKWT ; n = 7). Isolated ALKWT expression (n = 4) was seen with no ALK fusions. Stage III patients showed improved survival with ALKATI expression compared to those with ALKWT or no expression [5-year survival 80, 95% confidence interval (CI) = 57-100% versus 43%, 95% CI = 34-55%, P = 0.013]. Clinicopathological characteristics were not statistically significant. Strong diffuse cytoplasmic staining of ALK IHC (n = 12) has a sensitivity of 52.2%, specificity 100%, PPV of 100% and NPV of 92.5% of detecting isolated ALKATI . CONCLUSION Presence of ALKATI is a good prognostic indicator in MM. ALK IHC and digital molecular analysis can be incorporated into MM evaluation to identify patients with ALKATI for targeted therapy.
Collapse
Affiliation(s)
- Kabeer K Shah
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Dermatopathology, Mayo Clinic, Rochester, MN, USA
| | - Jadee L Neff
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Laboratory Genetics, Mayo Clinic, Rochester, MN, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Lori A Erickson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Dermatopathology, Mayo Clinic, Rochester, MN, USA
| | - Rory A Jackson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Laboratory Genetics, Mayo Clinic, Rochester, MN, USA
| | - Sarah M Jenkins
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Aaron S Mansfield
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - John A Copland
- Department of Cancer Biology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Kevin C Halling
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Laboratory Genetics, Mayo Clinic, Rochester, MN, USA
| | - Thomas J Flotte
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Division of Dermatopathology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
8
|
Myoepithelioma-like Hyalinizing Epithelioid Tumors of the Hand With Novel OGT-FOXO3 Fusions. Am J Surg Pathol 2020; 44:387-395. [PMID: 31567281 DOI: 10.1097/pas.0000000000001380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Myoepithelial tumors of soft tissue are uncommon neoplasms characterized histologically by spindle to epithelioid cells arranged in cords, nests, and/or reticular pattern with chondromyxoid to hyaline stroma, and genetically by rearrangement involving EWSR1 (among other less common genes) in about half of the cases. The diagnosis often requires immunostaining to confirm myoepithelial differentiation, most importantly the expression of epithelial markers and S100 protein and/or GFAP. However, there are cases wherein the morphology is reminiscent of myoepithelial tumors, while the immunophenotype falls short. Here, we report 2 highly similar myoepithelioma-like tumors arising in the hands of young adults. Both tumors were well-demarcated and composed of alternating cellular areas with palely eosinophilic hyaline stroma and scattered acellular zones of densely eosinophilic collagen deposition. The tumor cells were mainly epithelioid cells and arranged in cords or small nests. Vacuolated cells encircling hyaline matrix globules were focally prominent. A minor component of nonhyaline fibrous nodular areas composed of bland spindle cells and rich vasculature was also observed. Perivascular concentric spindle cell proliferation and perivascular hyalinization were present in some areas. The tumor cells were positive for CD34 and epithelial membrane antigen (focal) by immunostaining, while largely negative for cytokeratin, S100, GFAP, p63, GLUT1, and claudin-1. By RNA sequencing, a novel OGT-FOXO3 fusion gene was identified in case 1 and confirmed by reverse transcription polymerase chain reaction and fluorescence in situ hybridization in both cases. Sharing the unusual clinicopathologic features and the novel fusion, these 2 cases probably represent a distinct tumor entity, whose relationship with myoepithelial tumors and tumorigenic mechanisms exerted by the OGT-FOXO3 fusion remain to be studied.
Collapse
|
9
|
Yao LZ, Zhu YL, Liu JJ. Inhibition of PTEN Gene Expression by Small Interfering RNA on PI3K/ Akt/ FoxO3a Signaling Pathway in Human Nasopharyngeal Carcinoma. Technol Cancer Res Treat 2020; 19:1533033820917959. [PMID: 32281513 PMCID: PMC7155238 DOI: 10.1177/1533033820917959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/01/2020] [Accepted: 03/13/2020] [Indexed: 12/15/2022] Open
Abstract
The objective of this article is to study the effect of inhibiting phosphatase and tensin homolog deleted chromatosome 10 gene on phosphoinositide 3-kinase/protein kinase B (Akt)/Forkhead homeobox O3a signaling pathway in human nasopharyngeal carcinoma HK-1 cells. Nasopharyngeal carcinoma HK-1 cell lines were divided into PTEN gene interference group (siPTEN), nonspecific small interfering RNA group (siNC), empty vector group (Vector), and no transfection control group (Normal). The mRNA and protein expression levels of PTEN, PI3K, p-Akt, and FoxO3a were detected by real-time fluorescence quantitative polymerase chain reaction and Western blot. Immunofluorescence was used to detect the subcellular localization of PTEN, PI3K, p-Akt, and FoxO3a in HK-1 cells. The proliferation of HK-1 cells was detected by MTT assay, and the apoptosis of HK-1 cells was detected by flow cytometry. Compared with the siNC group, the expression levels of PTEN, FoxO3a messenger RNA, and protein in the siPTEN group were significantly decreased (P < .05), while the expression levels of PI3K, p-Akt messenger RNA, and protein were significantly increased (P < .05). The growth rate of HK-1 cells in the siPTEN group was significantly higher than the siNC group (P < .05), while the apoptosis rate was significantly lower than that of the siNC group (P < .05). Small interfering RNA can inhibit the expression of PTEN in HK-1 cells, and PTEN can participate in the development of NPC by affecting PI3K/Akt/FoxO3a signaling pathway.
Collapse
Affiliation(s)
- Liang Zhong Yao
- Department of Otorhinolaryngology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Yan Li Zhu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jun Jie Liu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
He X, Li S, Shi W, Lin Q, Ma J, Liu Y, Feng T, Cao X. Cyclin A1 is associated with poor prognosis in oesophageal squamous cell carcinoma. Oncol Lett 2019; 18:706-712. [PMID: 31289545 PMCID: PMC6546994 DOI: 10.3892/ol.2019.10377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/02/2019] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of cyclin A1 (CCNA1) is implicated in the carcinogenesis, progression and metastasis of many types of solid tumours. In the present study, an mRNA single-channel expression profile chip experiment revealed that the CCNA1 mRNA levels in oesophageal squamous cell carcinoma (ESCC) were increased >10-fold compared with those in the adjacent non-cancer tissues. Reverse transcription-quantitative polymerase chain reaction and immunohistochemistry analyses were performed to additionally investigate the role of CCNA1 in the development and progression of ESCC in patients treated by radical resection of the oesophagus. The association between CCNA1 mRNA expression and the clinicopathological parameters of patients with ESCC was statistically analysed. The results indicated that upregulation of CCNA1 occurred in ~70% of patients with ESCC, and increased CCNA1 mRNA expression was significantly associated with advanced clinical stage, lymph node metastasis, invasiveness and poor clinical outcome, including disease-free survival and overall survival rates. Taken together, the data suggested that CCNA1 had an important function in ESCC development and progression, and may serve as a prognostic biomarker and therapeutic target in ESCC.
Collapse
Affiliation(s)
- Xiaoting He
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China.,Department of Medical Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214043, P.R. China
| | - Suqing Li
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Weihong Shi
- Department of Clinical Medicine, Yancheng Vocational Institute of Health Sciences, Yancheng, Jiangsu 224005, P.R. China
| | - Qingfeng Lin
- Department of Medical Oncology, Jiangyin People's Hospital, Jiangyin, Jiangsu 214400, P.R. China
| | - Jian Ma
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Yang Liu
- Department of Pharmacy, No. 401 Hospital of Chinese People's Liberation Army, Qingdao, Shandong 266071, P.R. China
| | - Tingting Feng
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Xiufeng Cao
- Department of Surgical Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China.,Department of Surgery, Nanjing Medical University, Taikang Xianlin Drum Tower Hospital, Jiangsu 210000, P.R. China
| |
Collapse
|
11
|
Chintakuntlawar AV, Foote RL, Kasperbauer JL, Bible KC. Diagnosis and Management of Anaplastic Thyroid Cancer. Endocrinol Metab Clin North Am 2019; 48:269-284. [PMID: 30717908 DOI: 10.1016/j.ecl.2018.10.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Anaplastic thyroid cancer (ATC) is a devastating and usually incurable diagnosis. Clinical and pathologic diagnosis is best assessed at a tertiary center with concentrated ATC expertise. Expeditious multidisciplinary management is recommended for optimal patient outcomes. Based on multiinstitutional and population-based studies, multimodal therapy that includes chemoradiotherapy with surgery (when feasible) is the preferred initial treatment because it is associated with incrementally improved overall survival. In ATC that carries a BRAF V600E somatic mutation, combination therapy with BRAF and MEK inhibitors has shown promise but needs further study. Immunotherapeutic agents in neoadjuvant and metastatic settings are being investigated.
Collapse
Affiliation(s)
| | - Robert L Foote
- Department of Radiation Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Jan L Kasperbauer
- Division of Head and Neck Surgery, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Keith C Bible
- Division of Medical Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
12
|
Luo P, Tian LP, Ruan J, Wu FX. Disease Gene Prediction by Integrating PPI Networks, Clinical RNA-Seq Data and OMIM Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:222-232. [PMID: 29990218 DOI: 10.1109/tcbb.2017.2770120] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Disease gene prediction is a challenging task that has a variety of applications such as early diagnosis and drug development. The existing machine learning methods suffer from the imbalanced sample issue because the number of known disease genes (positive samples) is much less than that of unknown genes which are typically considered to be negative samples. In addition, most methods have not utilized clinical data from patients with a specific disease to predict disease genes. In this study, we propose a disease gene prediction algorithm (called dgSeq) by combining protein-protein interaction (PPI) network, clinical RNA-Seq data, and Online Mendelian Inheritance in Man (OMIN) data. Our dgSeq constructs differential networks based on rewiring information calculated from clinical RNA-Seq data. To select balanced sets of non-disease genes (negative samples), a disease-gene network is also constructed from OMIM data. After features are extracted from the PPI networks and differential networks, the logistic regression classifiers are trained. Our dgSeq obtains AUC values of 0.88, 0.83, and 0.80 for identifying breast cancer genes, thyroid cancer genes, and Alzheimer's disease genes, respectively, which indicates its superiority to other three competing methods. Both gene set enrichment analysis and predicted results demonstrate that dgSeq can effectively predict new disease genes.
Collapse
|
13
|
Ishii K, Hatori K, Takeichi O, Makino K, Himi K, Komiya H, Ogiso B. Expression of the Forkhead box transcription factor Foxo3a in human periapical granulomas. J Oral Sci 2018; 60:479-483. [PMID: 30429437 DOI: 10.2334/josnusd.17-0439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
It has been reported that Forkhead box transcription factor class O3a (Foxo3a) is expressed in rheumatoid arthritis, a chronic inflammatory condition accompanied by bone resorption, and plays a role in its pathology. However, it has remained unclear whether Foxo3a is involved in the pathogenesis of periapical granulomas. The present study was performed to compare the expression of Foxo3a in periapical granulomas and healthy gingival tissues. Samples were obtained surgically from patients, and subjected to hematoxylin-eosin staining for histopathologic diagnosis. Two-color immunofluorescence staining was also performed using antibodies against Foxo3a and markers for three types of inflammatory cells: neutrophils, T lymphocytes, and B lymphocytes. This revealed that Foxo3a was expressed in all three cell types in periapical granulomas but not in healthy gingival tissues. Foxo3a was expressed in 82.1%, 78.3%, and 77.5% of neutrophils, T lymphocytes, and B lymphocytes, respectively, and statistical analysis using the Kruskal-Wallis test followed by the Steel-Dwass test showed no significant difference of Foxo3a expression among the three cell types. Our results suggest that Foxo3a transcription factors may be involved in the pathogenesis of periapical granulomas.
Collapse
Affiliation(s)
- Kae Ishii
- Division of Applied Oral Sciences, Nihon University Graduate School of Dentistry.,Department of Endodontics, Nihon University School of Dentistry
| | - Keisuke Hatori
- Department of Endodontics, Nihon University School of Dentistry.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School Dentistry
| | - Osamu Takeichi
- Department of Endodontics, Nihon University School of Dentistry.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School Dentistry
| | - Kosuke Makino
- Department of Endodontics, Nihon University School of Dentistry
| | - Kazuma Himi
- Division of Applied Oral Sciences, Nihon University Graduate School of Dentistry.,Department of Endodontics, Nihon University School of Dentistry
| | - Hiroki Komiya
- Division of Applied Oral Sciences, Nihon University Graduate School of Dentistry.,Department of Endodontics, Nihon University School of Dentistry
| | - Bunnai Ogiso
- Department of Endodontics, Nihon University School of Dentistry.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School Dentistry
| |
Collapse
|
14
|
Liu Y, Ao X, Ding W, Ponnusamy M, Wu W, Hao X, Yu W, Wang Y, Li P, Wang J. Critical role of FOXO3a in carcinogenesis. Mol Cancer 2018; 17:104. [PMID: 30045773 PMCID: PMC6060507 DOI: 10.1186/s12943-018-0856-3] [Citation(s) in RCA: 339] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
FOXO3a is a member of the FOXO subfamily of forkhead transcription factors that mediate a variety of cellular processes including apoptosis, proliferation, cell cycle progression, DNA damage and tumorigenesis. It also responds to several cellular stresses such as UV irradiation and oxidative stress. The function of FOXO3a is regulated by a complex network of processes, including post-transcriptional suppression by microRNAs (miRNAs), post-translational modifications (PTMs) and protein-protein interactions. FOXO3a is widely implicated in a variety of diseases, particularly in malignancy of breast, liver, colon, prostate, bladder, and nasopharyngeal cancers. Emerging evidences indicate that FOXO3a acts as a tumor suppressor in cancer. FOXO3a is frequently inactivated in cancer cell lines by mutation of the FOXO3a gene or cytoplasmic sequestration of FOXO3a protein. And its inactivation is associated with the initiation and progression of cancer. In experimental studies, overexpression of FOXO3a inhibits the proliferation, tumorigenic potential, and invasiveness of cancer cells, while silencing of FOXO3a results in marked attenuation in protection against tumorigenesis. The role of FOXO3a in both normal physiology as well as in cancer development have presented a great challenge to formulating an effective therapeutic strategy for cancer. In this review, we summarize the recent findings and overview of the current understanding of the influence of FOXO3a in cancer development and progression.
Collapse
Affiliation(s)
- Ying Liu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Xiang Ao
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Wei Ding
- Department of comprehensive internal medicine, Affiliated Hospital, Qingdao University, Qingdao, 266003 China
| | - Murugavel Ponnusamy
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Wei Wu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Xiaodan Hao
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Wanpeng Yu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Yifei Wang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Peifeng Li
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| | - Jianxun Wang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021 China
| |
Collapse
|
15
|
Ahn H, Kim H, Abdul R, Kim Y, Sim J, Choi D, Paik SS, Shin SJ, Kim DH, Jang K. Overexpression of Forkhead Box O3a and Its Association With Aggressive Phenotypes and Poor Prognosis in Human Hepatocellular Carcinoma. Am J Clin Pathol 2018; 149:117-127. [PMID: 29365018 DOI: 10.1093/ajcp/aqx132] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVES Recent research has demonstrated that forkhead box O3a (FoxO3a) may function as an oncogenic transcription factor. We sought to validate the clinicopathologic significance of FoxO3a expression in hepatocellular carcinoma (HCC). METHODS Western blotting and immunohistochemistry were used to determine FoxO3a expression. In vitro cell proliferation and migration assays were performed in a HepG2 cell line. RESULTS FoxO3a was overexpressed in 121 (64.71%) cases of HCC. FoxO3a overexpression was associated with aggressive phenotypes of HCC, such as histologic grade (P < .001), stage (P = .031), and small vessel invasion (P < .001). FoxO3a overexpression was also correlated with poor disease-free survival in both univariate and multivariate survival analyses (P = .001 and P = .018, respectively). Downregulation of FoxO3a in a HepG2 cell line inhibited cell proliferation and migration. CONCLUSIONS These results suggest a role for FoxO3a in HCC progression and support the potential use as a prognostic biomarker.
Collapse
Affiliation(s)
- Hyein Ahn
- Department of Pathology, Konyang University College of Medicine, Daejeon, Korea
| | - Hyunsung Kim
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Rehman Abdul
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Yesul Kim
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Jongmin Sim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dongho Choi
- Departments of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Seung Sam Paik
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Su-Jin Shin
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Dong-Hoon Kim
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kiseok Jang
- Departments of Pathology, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
16
|
van Doeselaar S, Burgering BMT. FOXOs Maintaining the Equilibrium for Better or for Worse. Curr Top Dev Biol 2018; 127:49-103. [PMID: 29433740 DOI: 10.1016/bs.ctdb.2017.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A paradigm shift is emerging within the FOXO field and accumulating evidence indicates that we need to reappreciate the role of FOXOs, at least in cancer development. Here, we discuss the possibility that FOXOs are both tumor suppressors as well as promoters of tumor progression. This is mostly dependent on the biological context. Critical to this dichotomous role is the notion that FOXOs are central in preserving cellular homeostasis in redox control, genomic stability, and protein turnover. From this perspective, a paradoxical role in both suppressing and enhancing tumor progression can be reconciled. As many small molecules targeting the PI3K pathway are developed by big pharmaceutical companies and/or are in clinical trial, we will discuss what the consequences may be for the context-dependent role of FOXOs in tumor development in treatment options based on active PI3K signaling in tumors.
Collapse
Affiliation(s)
- Sabina van Doeselaar
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
17
|
Jiang Z, Jiang J, Zhao B, Yang H, Wang Y, Guo S, Deng Y, Lu D, Ma T, Wang H, Wang J. CPNE1 silencing inhibits the proliferation, invasion and migration of human osteosarcoma cells. Oncol Rep 2017; 39:643-650. [PMID: 29207139 DOI: 10.3892/or.2017.6128] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 11/13/2017] [Indexed: 01/26/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignancy of the bone affecting children and adolescents. Copine 1 (CPNE1) is a highly conserved calcium-dependent phospholipid-binding protein and may function in regulating signal transduction and membrane trafficking. In the present study, we investigated CPNE1 expression in osteosarcoma tissues and cells, and studied the effects of small interfering RNA (siRNA)-targeting CPNE1 on proliferation, metastasis and chemosensitivity of the osteosarcoma cells. The results demonstrated that CPNE1 was highly expressed in the osteosarcoma tissues and cell lines. Moreover, functional investigations confirmed that CPNE1 knockdown significantly inhibited cell proliferation, colony formation, invasion and metastasis in Saos-2 and HOS cells. Western blot analysis indicated that CPNE1 silencing downregulated the expression of many proteins associated with tumorigenesis and development, including Ras, MEK-1/2, WNT1, β-catenin, cyclin A1, IRAK2 and cIAP2. In addition, CPNE1 downregulation enhanced the sensitivity of Saos-2 cells towards cisplatin and adriamycin. The present study provides deep insight into the clinical use of lentiviral-mediated CPNE1 silencing for osteosarcoma therapy.
Collapse
Affiliation(s)
- Zhenhuan Jiang
- Department of Orthopaedics, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214200, P.R. China
| | - Jiannong Jiang
- Department of Orthopaedics, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214200, P.R. China
| | - Bizeng Zhao
- Department of Orthopedics, Affiliated Sixth People's Hospital of Shanghai JiaoTong University, Shanghai 20023, P.R. China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215200, P.R. China
| | - Yunliang Wang
- Department of Neurology, The 148 Central Hospital of PLA, Zibo, Shangdong 255000, P.R. China
| | - Shang Guo
- Department of Orthopedics, Affiliated Sixth People's Hospital of Shanghai JiaoTong University, Shanghai 20023, P.R. China
| | - Youping Deng
- Bioinformatics Core, Department of Complementary and Integrative Medicine, University of Hawaii John A. Burns School of Medicine, Honolulu, HI 96813, USA
| | - Deyi Lu
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Tieliang Ma
- Central Laboratory, Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214200, P.R. China
| | - Hongwei Wang
- Shanghai Realgen Biotech Inc., Pudong New District, Shanghai 203215, P.R. China
| | - Jinzhi Wang
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou, Jiangsu 215007, P.R. China
| |
Collapse
|
18
|
Zhang L, Boufraqech M, Lake R, Kebebew E. Carfilzomib potentiates CUDC-101-induced apoptosis in anaplastic thyroid cancer. Oncotarget 2017; 7:16517-28. [PMID: 26934320 PMCID: PMC4941332 DOI: 10.18632/oncotarget.7760] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/08/2016] [Indexed: 12/19/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is one of the most aggressive human malignancies, with no effective treatment currently available. Previously, we identified agents active against ATC cells, both in vitro and in vivo, using quantitative high-throughput screening of 3282 clinically approved drugs and small molecules. Here, we report that combining two of these active agents, carfilzomib, a second-generation proteasome inhibitor, and CUDC-101, a histone deacetylase and multi-kinase inhibitor, results in increased, synergistic activity in ATC cells. The combination of carfilzomib and CUDC-101 synergistically inhibited cellular proliferation and caused cell death in multiple ATC cell lines harboring various driver mutations observed in human ATC tumors. This increased anti-ATC effect was associated with a synergistically enhanced G2/M cell cycle arrest and increased caspase 3/7 activity induced by the drug combination. Mechanistically, treatment with carfilzomib and CUDC-101 increased p21 expression and poly (ADP-ribose) polymerase protein cleavage. Our results suggest that combining carfilzomib and CUDC-101 would offer an effective therapeutic strategy to treat ATC.
Collapse
Affiliation(s)
- Lisa Zhang
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Myriem Boufraqech
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ross Lake
- Laboratory of Genitourinary Cancer Pathogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Electron Kebebew
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Qian Z, Ren L, Wu D, Yang X, Zhou Z, Nie Q, Jiang G, Xue S, Weng W, Qiu Y, Lin Y. Overexpression of FoxO3a is associated with glioblastoma progression and predicts poor patient prognosis. Int J Cancer 2017; 140:2792-2804. [PMID: 28295288 DOI: 10.1002/ijc.30690] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/09/2017] [Accepted: 03/01/2017] [Indexed: 12/28/2022]
Abstract
Forkhead transcription factor FoxO3a has been reported to have ambiguous functions and distinct mechanisms in various solid tumors, including glioblastoma (GBM). Although a preliminary analysis of a small sample of patients indicated that FoxO3a aberrations in glioma might be related to aggressive clinical behavior, the clinical significance of FoxO3a in glioblastoma remains unclear. We investigated the expression of FoxO3a in a cohort of 91 glioblastoma specimens and analyzed the correlations of protein expression with patient prognosis. Furthermore, the functional impact of FoxO3a on GBM progression and the underlying mechanisms of FoxO3a regulation were explored in a series of in vitro and in vivo assays. FoxO3a expression was elevated in glioblastoma tissues, and high nuclear FoxO3a expression in human GBM tissues was associated with poor prognosis. Moreover, knockdown of FoxO3a significantly reduced the colony formation and invasion ability of GBM cells, whereas overexpression of FoxO3a promoted the colony formation and invasion ability. The results of in vivo GBM models further confirmed that FoxO3a knockdown inhibited GBM progression. More, the pro-oncogenic effects of FoxO3a in GBM were mediated by the activation of c-Myc, microtubule-associated protein 1 light chain 3 beta (LC3B) and Beclin1 in a mixed-lineage leukemia 2 (MLL2)-dependent manner. These findings suggest that high FoxO3a expression is associated with glioblastoma progression and that FoxO3a independently indicates poor prognosis in patients. FoxO3a might be a novel prognostic biomarker or a potential therapeutic target in glioblastoma.
Collapse
Affiliation(s)
- Zhongrun Qian
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Ren
- Department of Neurosurgery, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Dingchang Wu
- Department of Clinical Laboratory, Longyan First Hospital, Fujian Medical University, Longyan, China
| | - Xi Yang
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiyi Zhou
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Quanmin Nie
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gan Jiang
- Department of Pharmacology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuanglin Xue
- Department of Neurosurgery, Longyan First Hospital, Fujian Medical University, Longyan, China
| | - Weiji Weng
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongming Qiu
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingying Lin
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
20
|
Huang Y, Tao Y, Li X, Chang S, Jiang B, Li F, Wang ZM. Bioinformatics analysis of key genes and latent pathway interactions based on the anaplastic thyroid carcinoma gene expression profile. Oncol Lett 2016; 13:167-176. [PMID: 28428828 PMCID: PMC5396846 DOI: 10.3892/ol.2016.5447] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/10/2016] [Indexed: 01/03/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is an aggressive malignant disease in older adults
with a high mortality rate. The present study aimed to examine several key genes and
pathways, which are associated with ATC. The GSE33630 gene expression profile was
downloaded from the Gene Expression Omnibus database, which included 11 ATC and 45
normal thyroid samples. The differentially expressed genes (DEGs) in ATC were
identified using the Limma package in R. The Gene Ontology functions and Kyoto
Encyclopedia of Genes and Genomes pathways of the selected DEGs were enriched using
the Database for Annotation, Visualization and Integrated Discovery. A
protein-protein interaction (PPI) network of the DEGs was constructed to select
significant modules. Furthermore, a latent pathway interactive network was
constructed to select the significant pathways associated with ATC. A total of 665
DEGs in the ATC samples were screened, and four significant modules were selected
from the PPI network. The DEGs in the four modules were enriched in several functions
and pathways. In addition, 29 significant pathways associated with ATC were selected,
and he Toll-like receptor (TLR) signaling pathway, extracellular matrix
(ECM)-receptor interaction and cytokine-cytokine interaction pathway were identified
as important pathways. FBJ murine osteosarcoma viral oncogene homolog (FOS),
chemokine C-X-C motif ligand 10 (CXCL10), collagen type V α1 (COL5A1) and
chemokine (C-C motif) ligand 28 (CCL28) were the key DEGs involved in these
significant pathways. The data obtained in the present study revealed that the TLR
signaling pathway, ECM-receptor interaction and cytokine-cytokine receptor
interaction pathway, and the FOS, CXCL10, COL5A1, COL11A1 and CCL28 genes have
different roles in the progression of ATC, and these may be used as therapeutic
targets for ATC.
Collapse
Affiliation(s)
- Yun Huang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yiming Tao
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Xinying Li
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Shi Chang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Bo Jiang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Feng Li
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Zhi-Ming Wang
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
21
|
Zhang L, Zhang Y, Mehta A, Boufraqech M, Davis S, Wang J, Tian Z, Yu Z, Boxer MB, Kiefer JA, Copland JA, Smallridge RC, Li Z, Shen M, Kebebew E. Dual inhibition of HDAC and EGFR signaling with CUDC-101 induces potent suppression of tumor growth and metastasis in anaplastic thyroid cancer. Oncotarget 2016; 6:9073-85. [PMID: 25940539 PMCID: PMC4496203 DOI: 10.18632/oncotarget.3268] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 01/31/2015] [Indexed: 12/20/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is one of the most lethal human malignancies that currently has no effective therapy. We performed quantitative high-throughput screening (qHTS) in three ATC cell lines using 3,282 clinically approved drugs and drug candidates, and identified 100 active agents. Enrichment analysis of active compounds showed that inhibitors of EGFR and histone deacetylase (HDAC) were most active. Of these, the first-in-class dual inhibitor of EGFR, HER2 and HDACs, CUDC-101, had the highest efficacy and lower IC50 than established drugs. We validated that CUDC-101 inhibited cellular proliferation and resulted in cell death by inducing cell cycle arrest and caspase-dependent apoptosis. CUDC-101 also inhibited cellular migration in vitro. Mechanistically, CUDC-101 inhibited MAPK signaling and histone deacetylation in ATC cell lines with multiple driver mutations present in human ATC. The anticancer effect of CUDC-101 was associated with increased expression of p21 and E-cadherin, and reduced expression of survivin, XIAP, β-catenin, N-cadherin, and Vimentin. In an in vivo mouse model of metastatic ATC, CUDC-101 inhibited tumor growth and metastases, and significantly prolonged survival. Response to CUDC-101 treatment in vivo was associated with increased histone 3 acetylation and reduced survivin expression. Our findings provide a preclinical basis to evaluate CUDC-101 therapy in ATC.
Collapse
Affiliation(s)
- Lisa Zhang
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yaqin Zhang
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Amit Mehta
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Myriem Boufraqech
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sean Davis
- Cancer Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jing Wang
- Curis Inc., Translational Science, Lexington, MA, USA
| | - Ze Tian
- Curis Inc., Translational Science, Lexington, MA, USA
| | - Zhiya Yu
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Matthew B Boxer
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey A Kiefer
- Division of Information Sciences, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - John A Copland
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Robert C Smallridge
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.,Endocrinology Division, Internal Medicine Department, Mayo Clinic, Jacksonville, FL, USA
| | - Zhuyin Li
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Min Shen
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Electron Kebebew
- Endocrine Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
22
|
Arsenic R, Braicu EI, Letsch A, Dietel M, Sehouli J, Keilholz U, Ochsenreither S. Cancer-testis antigen cyclin A1 is broadly expressed in ovarian cancer and is associated with prolonged time to tumor progression after platinum-based therapy. BMC Cancer 2015; 15:784. [PMID: 26499264 PMCID: PMC4619521 DOI: 10.1186/s12885-015-1824-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 10/16/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Cyclin A1 is essential for male gametopoiesis. In acute myeloid leukemia, it acts as a leukemia-associated antigen. Cyclin A1 expression has been reported in several epithelial malignancies, including testicular, endometrial, and epithelial ovarian cancer (EOC). We analyzed Cyclin A1 expression in EOC and its correlation with clinical features to evaluate Cyclin A1 as a T-cell target in EOC. METHODS Cyclin A1 mRNA expression in EOC and healthy tissues was quantified by microarray analysis and quantitative real-time PCR (qRT-PCR). Protein expression in clinical samples was assessed by immunohistochemistry (IHC) and was correlated to clinical features. RESULTS Cyclin A1 protein was homogeneously expressed in 43 of 62 grade 3 tumor samples and in 1 of 10 grade 2 specimens (p < 0.001). Survival analysis showed longer time to progression (TTP) among patients with at least moderate Cyclin A1 expression (univariate: p = 0.018, multivariate: p = 0.035). FIGO stage, grading, age, macroscopic residual tumor after debulking, and peritoneal carcinomatosis / distant metastasis had no impact on TTP or overall survival (OS). CONCLUSION Cyclin A1 is highly expressed in most EOCs. The mechanism behind the prolonged TTP in patients with high Cyclin A1 expression warrants further investigation. The frequent, selectively high expression of Cyclin A1 in EOC makes it a promising target for T-cell therapies.
Collapse
Affiliation(s)
- Ruza Arsenic
- Department of Pathology, Institute of Pathology, Charité - University Hospital Berlin, 10117, Berlin, Germany.
| | - Elena Ilona Braicu
- Departement of Gynecology, University Hospital Berlin, 13353, Berlin, Germany.
| | - Anne Letsch
- Department of Hematology, Oncology and Tumor Immunology - University Hospital Berlin, 12200, Berlin, Germany.
| | - Manfred Dietel
- Department of Pathology, Institute of Pathology, Charité - University Hospital Berlin, 10117, Berlin, Germany.
| | - Jalid Sehouli
- Departement of Gynecology, University Hospital Berlin, 13353, Berlin, Germany.
| | - Ulrich Keilholz
- Charité Cancer Comprehensive Center, Charité, 10117, Berlin, Germany.
| | - Sebastian Ochsenreither
- Department of Hematology, Oncology and Tumor Immunology - University Hospital Berlin, 12200, Berlin, Germany.
| |
Collapse
|
23
|
Han J, Zhao J, Jiang J, Ma X, Liu X, Wang C, Jiang S, Wan C. Zinc deficiency impairs the renewal of hippocampal neural stem cells in adult rats: involvement of FoxO3a activation and downstream p27(kip1) expression. J Neurochem 2015; 134:879-91. [PMID: 26086369 DOI: 10.1111/jnc.13199] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/25/2015] [Accepted: 05/29/2015] [Indexed: 12/30/2022]
Abstract
Zinc plays an important role in the development and maintenance of central neural system. Zinc deficiency has been known to alter normal brain function, whose molecular mechanism remains largely elusive. In the present study, we established a zinc deficiency-exposed rat model, and, using western blot and immunohistochemical analyses, found that the expression of FoxO3a and p27(kip1) was remarkably up-regulated in the rat brain hippocampus. Immunofluorescence assay showed that FOXO3a and p27(kip1) were significantly co-localized with nestin, the marker of neural stem cells (NSCs). Furthermore, we identified that the proportion of proliferating NSCs was markedly decreased in zinc-deficient rat hippocampaus. Using C17.2 neural stem cells, it was revealed that exposure to zinc chelator N,N,N',N'-tetrakis-(2-pyridylmethy) ethylenediamine induced the expression of FoxO3a and p27(kip1) , which coincided with reduced NSC proliferation. Furthermore, depletion of FoxO3a inhibited p27(kip1) expression and restored the growth of NSCs. On the basis of these data, we concluded that FoxO3a/p27(kip1) signaling might play a significant role in zinc deficiency-induced growth impairment of NSCs and consequent neurological disorders. We describe here that zinc deficiency induces the proliferative impairment of hippocampal neural stem cells partially through the activation of FOXO3a-p27 axis in rats. Neural progenitor cells exhibited significantly up-regulated expression of FOXO3a and p27 after zinc deficiency in vivo and in vitro. Depletion of FOXO3a ameliorates zinc deficiency-induced expression of p27 and growth impairment of neural stem cells. We provide novel insight into the mechanisms underlying zinc deficiency-induced neurological deficits.
Collapse
Affiliation(s)
- Jingling Han
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Occupational Medicine and Environmental Toxicity, School of Public Health, Nantong University, Nantong, Jiangsu Province, China
| | - Jianya Zhao
- Department of Nutrition and Food hygiene, School of Public Health, Nantong University, Nantong, Jiangsu Province, China
| | - Junkang Jiang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Occupational Medicine and Environmental Toxicity, School of Public Health, Nantong University, Nantong, Jiangsu Province, China
| | - Xia Ma
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Occupational Medicine and Environmental Toxicity, School of Public Health, Nantong University, Nantong, Jiangsu Province, China
| | - Xinhang Liu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Occupational Medicine and Environmental Toxicity, School of Public Health, Nantong University, Nantong, Jiangsu Province, China
| | - Cheng Wang
- Department of Nutrition and Food hygiene, School of Public Health, Nantong University, Nantong, Jiangsu Province, China
| | - Shengyang Jiang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Department of Occupational Medicine and Environmental Toxicity, School of Public Health, Nantong University, Nantong, Jiangsu Province, China
| | - Chunhua Wan
- Department of Nutrition and Food hygiene, School of Public Health, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
24
|
Champa D, Di Cristofano A. Modeling anaplastic thyroid carcinoma in the mouse. Discov Oncol 2014; 6:37-44. [PMID: 25420535 DOI: 10.1007/s12672-014-0208-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023] Open
Abstract
Anaplastic thyroid carcinoma is the least common form of thyroid cancer; however, it accounts for the majority of deaths associated with this family of malignancies. A number of genetically engineered immunocompetent mouse models recapitulating the genetic and histological features of anaplastic thyroid cancer have been very recently generated and represent an invaluable tool to dissect the mechanisms involved in the progression from indolent, well-differentiated tumors to aggressive, undifferentiated carcinomas and to identify novel therapeutic targets. In this review, we focus on the relevant characteristics associated with these models and on what we have learned in terms of anaplastic thyroid cancer biology, genetics, and response to targeted therapy.
Collapse
Affiliation(s)
- Devora Champa
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Room 302, Bronx, NY, 10461, USA
| | | |
Collapse
|
25
|
Singh D, Fox SM, Tal-Singer R, Bates S, Riley JH, Celli B. Altered gene expression in blood and sputum in COPD frequent exacerbators in the ECLIPSE cohort. PLoS One 2014; 9:e107381. [PMID: 25265030 PMCID: PMC4179270 DOI: 10.1371/journal.pone.0107381] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 08/14/2014] [Indexed: 11/27/2022] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) who are defined as frequent exacerbators suffer with 2 or more exacerbations every year. The molecular mechanisms responsible for this phenotype are poorly understood. We investigated gene expression profile patterns associated with frequent exacerbations in sputum and blood cells in a well-characterised cohort. Samples from subjects from the ECLIPSE COPD cohort were used; sputum and blood samples from 138 subjects were used for microarray gene expression analysis, while blood samples from 438 subjects were used for polymerase chain reaction (PCR) testing. Using microarray, 150 genes were differentially expressed in blood (>±1.5 fold change, p≤0.01) between frequent compared to non-exacerbators. In sputum cells, only 6 genes were differentially expressed. The differentially regulated genes in blood included downregulation of those involved in lymphocyte signalling and upregulation of pro-apoptotic signalling genes. Multivariate analysis of the microarray data followed by confirmatory PCR analysis identified 3 genes that predicted frequent exacerbations; B3GNT, LAF4 and ARHGEF10. The sensitivity and specificity of these 3 genes to predict the frequent exacerbator phenotype was 88% and 33% respectively. There are alterations in systemic immune function associated with frequent exacerbations; down-regulation of lymphocyte function and a shift towards pro-apoptosis mechanisms are apparent in patients with frequent exacerbations.
Collapse
Affiliation(s)
- Dave Singh
- University of Manchester, Medicines Evaluation Unit, Manchester, United Kingdom
- * E-mail:
| | - Steven M. Fox
- GlaxoSmithKline, Medicines Research Centre, Stevenage, United Kingdom
| | - Ruth Tal-Singer
- GlaxoSmithKline, King of Prussia, Pennsylvania, United States of America
| | - Stewart Bates
- GlaxoSmithKline, Medicines Research Centre, Stevenage, United Kingdom
| | - John H. Riley
- GlaxoSmithKline, Stockley Park, Uxbridge, United Kingdom
| | - Bartolome Celli
- Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
26
|
Yu DA, Yoon J, Ko YS, Park J, Kim SY, Kim MA, Kim JH, Jung J, Cheon Y, Lee HS, Kim WH, Lee BL. Forkhead transcription factor FOXO1 inhibits nuclear factor-κB in gastric cancer. APMIS 2014; 122:848-55. [DOI: 10.1111/apm.12247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 12/18/2013] [Indexed: 12/28/2022]
Affiliation(s)
- Da-Ae Yu
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
| | - Jiyeon Yoon
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
| | - Young San Ko
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
| | - Jinju Park
- Cancer Research Institute; Department of Tumor Biology; Seoul National University College of Medicine; Seoul South Korea
| | - Sue Youn Kim
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
| | - Min A Kim
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Ji Hun Kim
- Department of Pathology; Asan Medical Center; University of Ulsan College of Medicine; Seoul South Korea
| | - Jieun Jung
- Department of Nanobiomedical Science; Dankook University; Cheonan South Korea
| | - Younghee Cheon
- Department of Pediatrics; College of Medicine; Yeungnam University; Daegu South Korea
| | - Hye Seung Lee
- Department of Pathology; Seoul National University Bundang Hospital; Seongnam South Korea
| | - Woo Ho Kim
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Byung Lan Lee
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
- Cancer Research Institute; Department of Tumor Biology; Seoul National University College of Medicine; Seoul South Korea
- Ischemic/Hypoxic Disease Institute Medical Research Center; Seoul National University College of Medicine; Seoul South Korea
| |
Collapse
|
27
|
Liu Y, Huang Y, Zhu GZ. Cyclin A1 is a transcriptional target of PITX2 and overexpressed in papillary thyroid carcinoma. Mol Cell Biochem 2013; 384:221-7. [PMID: 24002705 DOI: 10.1007/s11010-013-1801-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/23/2013] [Indexed: 10/26/2022]
Abstract
Physiological expression of cyclin A1, a unique cell cycle regulator essential for spermatogenesis, is predominantly restricted in male germ cells. Outstandingly, previous studies have also demonstrated the abnormal expression of cyclin A1 in various human tumors. How male germ cell-specific cyclin A1 is transcriptionally activated in tumor cells, however, is elusive. To begin to understand the molecular mechanisms governing the ectopic expression of cyclin A1, we searched for transcription factors and cis-regulatory DNA elements. We found that overexpression of PITX2, a paired-like homeodomain transcription factor and a downstream effector of Wnt/β-catenin signaling, resulted in upregulation of cyclin A1 in HEK293 cells and TPC-1 thyroid cancer cells. On the other hand, PITX2 knockdown in TPC-1 cells caused reduced cyclin A1. Promoter reporter assays with a series of deletion constructs determined that the DNA element from -102 to -96 bp of the cyclin A1 promoter is responsible for PITX2-induced gene expression. The result of chromatin immunoprecipitation revealed the occupancy of PITX2 on the cyclin A1 promoter. Taken together, these findings demonstrate that cyclin A1 is a transcriptional target of PITX2. Consistently, our immunohistochemistry result showed up-regulation of cyclin A1 in human papillary thyroid carcinoma, where overexpressed PITX2 has been endorsed in our recent report. Thus, our study provides new evidence on the regulation of cyclin A1 gene expression and offers a PITX2-cycin A1 pathway for cell cycle regulation.
Collapse
Affiliation(s)
- Yan Liu
- Department of Ophthalmology, Huashan Hospital Affiliated to Fudan University, 12 Middle Wulumuqi Road, Shanghai, 200040, People's Republic of China
| | | | | |
Collapse
|
28
|
Grunewald TGP, Willier S, Janik D, Unland R, Reiss C, da Costa OP, Buch T, Dirksen U, Richter GH, Neff F, Burdach S, Butt E. The Zyxin-related protein thyroid receptor interacting protein 6 (TRIP6) is overexpressed in Ewing's sarcoma and promotes migration, invasion and cell growth. Biol Cell 2013; 105:535-47. [DOI: 10.1111/boc.201300041] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 08/16/2013] [Indexed: 01/29/2023]
Affiliation(s)
- Thomas G. P. Grunewald
- Children's Cancer Research Center and Roman Herzog Comprehensive Cancer Center; Laboratory of Functional Genomics and Transplantation Biology; Klinikum rechts der Isar; Technische Universität München; Munich 80804 Germany
| | - Semjon Willier
- Institute for Clinical Biochemistry and Pathobiochemistry; University Clinic of Würzburg; Würzburg 97080 Germany
| | - Dirk Janik
- Institute of Pathology; Helmholtz Center Munich; Neuherberg 85764 Germany
| | - Rebekka Unland
- Department of Pediatric Hematology and Oncology; University Hospital Muenster; Westphalian Wilhelms University; Muenster 48149 Germany
| | - Cora Reiss
- Institute for Clinical Biochemistry and Pathobiochemistry; University Clinic of Würzburg; Würzburg 97080 Germany
- Center for Thrombosis and Hemostasis; University Medical Center Mainz; Mainz 55131 Germany
| | - Olivia Prazeres da Costa
- Institute for Medical Microbiology, Immunology, and Hygiene; Technische Universität München; Munich 81675 Germany
| | - Thorsten Buch
- Institute for Medical Microbiology, Immunology, and Hygiene; Technische Universität München; Munich 81675 Germany
| | - Uta Dirksen
- Department of Pediatric Hematology and Oncology; University Hospital Muenster; Westphalian Wilhelms University; Muenster 48149 Germany
| | - Günther H.S. Richter
- Children's Cancer Research Center and Roman Herzog Comprehensive Cancer Center; Laboratory of Functional Genomics and Transplantation Biology; Klinikum rechts der Isar; Technische Universität München; Munich 80804 Germany
| | - Frauke Neff
- Institute of Pathology; Helmholtz Center Munich; Neuherberg 85764 Germany
| | - Stefan Burdach
- Children's Cancer Research Center and Roman Herzog Comprehensive Cancer Center; Laboratory of Functional Genomics and Transplantation Biology; Klinikum rechts der Isar; Technische Universität München; Munich 80804 Germany
| | - Elke Butt
- Institute for Clinical Biochemistry and Pathobiochemistry; University Clinic of Würzburg; Würzburg 97080 Germany
| |
Collapse
|
29
|
Cadmium modifies the cell cycle and apoptotic profiles of human breast cancer cells treated with 5-fluorouracil. Int J Mol Sci 2013; 14:16600-16. [PMID: 23941782 PMCID: PMC3759927 DOI: 10.3390/ijms140816600] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 07/15/2013] [Accepted: 07/22/2013] [Indexed: 12/20/2022] Open
Abstract
Industrialisation, the proximity of factories to cities, and human work activities have led to a disproportionate use of substances containing heavy metals, such as cadmium (Cd), which may have deleterious effects on human health. Carcinogenic effects of Cd and its relationship with breast cancer, among other tumours, have been reported. 5-Fluorouracil (5-FU) is a fluoropyrimidine anticancer drug used to treat solid tumours of the colon, breast, stomach, liver, and pancreas. The purpose of this work was to study the effects of Cd on cell cycle, apoptosis, and gene and protein expression in MCF-7 breast cancer cells treated with 5-FU. Cd altered the cell cycle profile, and its effects were greater when used either alone or in combination with 5-FU compared with 5-FU alone. Cd significantly suppressed apoptosis of MCF-7 cells pre-treated with 5-FU. Regarding gene and protein expression, bcl2 expression was mainly upregulated by all treatments involving Cd. The expression of caspase 8 and caspase 9 was decreased by most of the treatments and at all times evaluated. C-myc expression was increased by all treatments involving Cd, especially 5-FU plus Cd at the half time of treatment. Cd plus 5-FU decreased cyclin D1 and increased cyclin A1 expression. In conclusion, our results indicate that exposure to Cd blocks the anticancer effects of 5-FU in MCF-7 cells. These results could have important clinical implications in patients treated with 5-FU-based therapies and who are exposed to high levels of Cd.
Collapse
|