1
|
Wang B, Li D, Ilnytskyy Y, Khachigian LM, Zhong N, Rodriguez-Juarez R, Kovalchuk I, Kovalchuk O. A Positive Feedback DNA-PK/MYT1L-CXCR1-ERK1/2 Proliferative Signaling Loop in Glioblastoma. Int J Mol Sci 2025; 26:4398. [PMID: 40362634 PMCID: PMC12072392 DOI: 10.3390/ijms26094398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Glioblastoma is the most common primary brain tumor in adults. Our previous studies revealed a functional interplay of myelin transcription factor 1-like (MYT1L) with the DNA-dependent protein kinase (DNA-PK) in the regulation of p21 transcription. However, the contributing role of this functional interplay in glioblastoma remains largely unknown. Here, we used cell lines with normal DNA-PK (HEK293 and M059K) or deficient DNA-PK (M059J) as a model system to demonstrate the importance of the DNA-PK-dependent activation of MYT1L in controlling the transcription of CXC chemokine receptor 1 (CXCR1) in a positive-feedback proliferative signaling loop in glioblastoma with numerous conventional techniques. In normal DNA-PK cells, MYT1L acted as an oncogene by promoting cell proliferation, inhibiting apoptosis, and shortening a cell cycle S phase. However, in DNA-PK-deficient cells, MYT1L functioned as a tumor suppressor by inhibiting cell proliferation and inducing a G1 arrest. The enforced expression of MYT1L promoted CXCR1 transcription in DNA-PK-normal cells but attenuated transcription in DNA-PK-deficient cells. Bioinformatics analysis predicted a MYT1L-binding sequence at the CXCR1 promoter. The functional dependence of MYT1L on DNA-PK in CXCR1 transcription was validated by luciferase assay. Although the expression of CXCR1 was lower in M059J cells as compared to M059K cells, it was higher than in normal brain tissue. The CXCR1 ligands interleukin 8 (IL-8) and GRO protein alpha (GROα) expressed in M059J and M059K cells may signal through the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway that can be blocked by CXCR1 siRNA. Our findings demonstrate the existence of a positive feedback DNA-PK/MYT1L-CXCR1-ERK1/2 proliferation loop in glioblastoma cells that may represent a pharmacological target loop for therapeutic intervention.
Collapse
Affiliation(s)
- Bo Wang
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Dongping Li
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Levon M. Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia;
| | - Nuanying Zhong
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Rocio Rodriguez-Juarez
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (B.W.); (D.L.); (Y.I.); (N.Z.); (R.R.-J.)
| |
Collapse
|
2
|
Zhang Y, Duan W, Chen L, Chen J, Xu W, Fan Q, Li S, Liu Y, Wang S, He Q, Li X, Huang Y, Peng H, Zhao J, Zhang Q, Qiu Z, Shao Z, Zhang B, Wang Y, Tian Y, Shu Y, Qin Z, Chi Y. Potassium ion channel modulation at cancer-neural interface enhances neuronal excitability in epileptogenic glioblastoma multiforme. Neuron 2025; 113:225-243.e10. [PMID: 39532103 DOI: 10.1016/j.neuron.2024.10.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 08/12/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
The central nervous system (CNS) is increasingly recognized as a critical modulator in the oncogenesis of glioblastoma multiforme (GBM), with interactions between cancer and local neuronal circuits frequently leading to epilepsy; however, the relative contributions of these factors remain unclear. Here, we report a coordinated intratumor shift among distinct cancer subtypes within progenitor-like families of epileptic GBM patients, revealing an accumulation of oligodendrocyte progenitor (OPC)-like subpopulations at the cancer-neuron interface along with heightened electrical signaling activity in the surrounding neuronal networks. The OPC-like cells associated with epilepsy express KCND2, which encodes the voltage-gated K+ channel KV4.2, enhancing neuronal excitability via accumulation of extracellular K+, as demonstrated in patient-derived ex vivo slices, xenografting models, and engineering organoids. Together, we uncovered the essential local circuitry, cellular components, and molecular mechanisms facilitating cancer-neuron interaction at peritumor borders. KCND2 plays a crucial role in mediating nervous system-cancer electrical communication, suggesting potential targets for intervention.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Wei Duan
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Lingchao Chen
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Junrui Chen
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Wei Xu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Qi Fan
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Shuwei Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Yuandong Liu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Shidi Wang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China
| | - Quansheng He
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Xiaohui Li
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Yang Huang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Haibao Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Jiaxu Zhao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Qiangqiang Zhang
- Advanced Model Animal Research Center, Department of Biotechnology and Biomedicine, Yangtze Delta Region Institute, Tsinghua University, Zhejiang 314006, China; Zhejiang Key Laboratory of Multiomics and Molecular Enzymology, Yangtze Delta Region Institute, Tsinghua University, Zhejiang 314006, China
| | - Zhixin Qiu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China; Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhicheng Shao
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Bo Zhang
- Novel Bioinformatics Co., Ltd., Shanghai, China
| | - Yihua Wang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China
| | - Yang Tian
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China.
| | - Yousheng Shu
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China.
| | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China.
| | - Yudan Chi
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200032, China.
| |
Collapse
|
3
|
Bastola S, Pavlyukov MS, Sharma N, Ghochani Y, Nakano MA, Muthukrishnan SD, Yu SY, Kim MS, Sohrabi A, Biscola NP, Yamashita D, Anufrieva KS, Kovalenko TF, Jung G, Ganz T, O'Brien B, Kawaguchi R, Qin Y, Seidlits SK, Burlingame AL, Oses-Prieto JA, Havton LA, Goldman SA, Hjelmeland AB, Nakano I, Kornblum HI. Endothelial-secreted Endocan activates PDGFRA and regulates vascularity and spatial phenotype in glioblastoma. Nat Commun 2025; 16:471. [PMID: 39773984 PMCID: PMC11707362 DOI: 10.1038/s41467-024-55487-1] [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] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Extensive neovascularization is a hallmark of glioblastoma (GBM). In addition to supplying oxygen and nutrients, vascular endothelial cells provide trophic support to GBM cells via paracrine signaling. Here we report that Endocan (ESM1), an endothelial-secreted proteoglycan, confers enhanced proliferative, migratory, and angiogenic properties to GBM cells and regulates their spatial identity. Mechanistically, Endocan exerts at least part of its functions via direct binding and activation of the PDGFRA receptor. Subsequent downstream signaling enhances chromatin accessibility of the Myc promoter and upregulates Myc expression inducing stable phenotypic changes in GBM cells. Furthermore, Endocan confers radioprotection on GBM cells in vitro and in vivo. Inhibition of Endocan-PDGFRA signaling with ponatinib increases survival in the Esm1 wild-type but not in the Esm1 knock-out mouse GBM model. Our findings identify Endocan and its downstream signaling axis as a potential target to subdue GBM recurrence and highlight the importance of vascular-tumor interactions for GBM development.
Collapse
Affiliation(s)
- Soniya Bastola
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Marat S Pavlyukov
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Neel Sharma
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yasmin Ghochani
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Mayu A Nakano
- Precision Medicine Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sree Deepthi Muthukrishnan
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sang Yul Yu
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Min Soo Kim
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alireza Sohrabi
- Department of Bioengineering, University of Texas at Austin, Austin, TX, USA
| | - Natalia P Biscola
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daisuke Yamashita
- Department of Neurosurgery, Ehime University Graduate School of Medicine, Shitsukawa 454, Toon, Ehime, Japan
| | - Ksenia S Anufrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine of Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical and Biological Agency, Moscow, Russia
| | | | - Grace Jung
- Department of Medicine, Center for Iron Disorders, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tomas Ganz
- Department of Medicine, Center for Iron Disorders, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Beatrice O'Brien
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Riki Kawaguchi
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Interdepartmental Program in Bioinformatics, Program in Neurogenetics, Department of Neurology and Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yue Qin
- Interdepartmental Program in Bioinformatics, Program in Neurogenetics, Department of Neurology and Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Leif A Havton
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, James J Peters VA Medical Center, Bronx, NY, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anita B Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ichiro Nakano
- Department of Neurosurgery, Harada Hospital, Iruma, Saitama, Japan.
| | - Harley I Kornblum
- The Intellectual and Developmental Disabilities Research Center, The Semel Institute for Neuroscience and Human Behavior, and The Broad Stem Cell Research Center, The Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Yoshida K, Chambers JK, Uchida K. Chromosomal gain and mutations of platelet-derived growth factor receptor-α gene in canine high-grade oligodendroglioma. Vet Pathol 2025:3009858241309396. [PMID: 39757746 DOI: 10.1177/03009858241309396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Canine high-grade oligodendrogliomas (HGOGs) exhibit a high expression of platelet-derived growth factor receptor-α (PDGFRA). We examined PDGFRA mutations and gain of PDGFRA and their association with the PDGFRA expression and proliferation of tumor cells in canine HGOG cases and cell lines. Polymerase chain reaction and sequence analysis revealed expected pathogenic mutations in PDGFRA exons 7 and 8 in 16/34 (47%) cases. However, these mutations were not associated with PDGFRA expression, as examined by mRNA in situ hybridization (ISH) and immunohistochemistry, or proliferation activity, as examined by the Ki-67 labeling index (LI). Chromosomal ISH performed in 16 cases revealed PDGFRA and endoplasmic reticulum membrane protein complex subunit 2 (EMC2) gains in 15 cases (94%). PDGFRA gain was moderately correlated with PDGFRA mRNA expression (ρ = 0.54, P = .04) and were moderately correlated with PDGFRA H-score, which is the score based on immunolabeling intensity (ρ = 0.44, P = .09). However, PDGFRA gain was not correlated with the Ki-67 LI (ρ = 0.23, P = .38). The canine HGOG cell line with PDGFRA gain showed higher PDGFRA mRNA expression (P < .01), H-score (P < .01), and Ki-67 LI (P < .01) than the cell line without PDGFRA gain in vitro. The gain of PDGFRA and EMC2 suggests polysomy of canine chromosome 13, where both genes are located. The in vitro analysis results suggested that chromosome 13 polysomy is associated with increased PDGFRA expression and cell proliferation in canine HGOG. Chromosome 13 polysomy may be involved in canine gliomagenesis by increasing PDGFRA expression and inducing tumor cell proliferation.
Collapse
|
5
|
Yoshida K, Chambers JK, Uchida K. The relationships of platelet-derived growth factor, microvascular proliferation, and tumor cell proliferation in canine high-grade oligodendrogliomas: Immunohistochemistry of 45 tumors and an AFOB-01 xenograft mouse model. Vet Pathol 2024; 61:732-742. [PMID: 38577818 DOI: 10.1177/03009858241241793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
High-grade oligodendroglioma (HGOG) is the most common type of glioma in dogs and expresses platelet-derived growth factor receptor-α (PDGFR-α). Microvascular proliferation is often observed in HGOG. Therefore, the present study investigated the functional relationships between PDGFR-α, microvascular proliferation, and tumor cell proliferation in canine HGOG. The expression of PDGFR-α and PDGF-subunit A (PDGF-A) in tumor cells, as well as endothelial cells and pericytes of tumor-associated microvascular proliferations, in 45 canine HGOGs were examined immunohistochemically. Microvascular proliferation was observed in 24/45 cases (53%). PDGFR-α expression in tumor cells and microvascular proliferations was observed in 45/45 (100%) and 2/24 cases (8%), respectively. Furthermore, PDGF-A expression in tumor cells and microvascular proliferations was detected in 13/45 (29%) and 24/24 cases (100%), respectively. In vitro, stimulation of the canine HGOG cell line AOFB-01 with PDGF-A showed that the doubling time of AOFB-01 cells was significantly shorter with PDGF-A than without PDGF-A. Crenolanib (a PDGFR inhibitor) inhibited AOFB-01 cell proliferation. In vivo, the AOFB-01 xenograft mouse model was treated with crenolanib. Tumor xenografts were smaller in crenolanib-treated mice than in untreated control mice. PDGFR-α expression in tumor cells and PDGF-A expression in microvascular proliferations and tumor cells suggest autocrine and paracrine effects of PDGF-A in canine HGOG. The results of in vitro assays indicate that canine HGOG expresses functional PDGFR-α, which responds to PDGF-A. Therefore, PDGF-A produced by microvascular proliferations and tumor cells may promote the proliferation of PDGFR-α-expressing tumor cells in canine HGOG. PDGFR-α signaling has potential as a therapeutic target.
Collapse
|
6
|
Srivastava R, Dodda M, Zou H, Li X, Hu B. Tumor Niches: Perspectives for Targeted Therapies in Glioblastoma. Antioxid Redox Signal 2023; 39:904-922. [PMID: 37166370 PMCID: PMC10654996 DOI: 10.1089/ars.2022.0187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/12/2023]
Abstract
Significance: Glioblastoma (GBM), the most common and lethal primary brain tumor with a median survival rate of only 15 months and a 5-year survival rate of only 6.8%, remains largely incurable despite the intensive multimodal treatment of surgical resection and radiochemotherapy. Developing effective new therapies is an unmet need for patients with GBM. Recent Advances: Targeted therapies, such as antiangiogenesis therapy and immunotherapy, show great promise in treating GBM based upon increasing knowledge about brain tumor biology. Single-cell transcriptomics reveals the plasticity, heterogeneity, and dynamics of tumor cells during GBM development and progression. Critical Issues: While antiangiogenesis therapy and immunotherapy have been highly effective in some types of cancer, the disappointing results from clinical trials represent continued challenges in applying these treatments to GBM. Molecular and cellular heterogeneity of GBM is developed temporally and spatially, which profoundly contributes to therapeutic resistance and tumor recurrence. Future Directions: Deciphering mechanisms of tumor heterogeneity and mapping tumor niche trajectories and functions will provide a foundation for the development of more effective therapies for GBM patients. In this review, we discuss five different tumor niches and the intercellular and intracellular communications among these niches, including the perivascular, hypoxic, invasive, immunosuppressive, and glioma-stem cell niches. We also highlight the cellular and molecular biology of these niches and discuss potential strategies to target these tumor niches for GBM therapy. Antioxid. Redox Signal. 39, 904-922.
Collapse
Affiliation(s)
- Rashmi Srivastava
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Meghana Dodda
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Han Zou
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Changsha, China
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- John G. Rangos Sr. Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Liu X, Hu Y, Xue Z, Zhang X, Liu X, Liu G, Wen M, Chen A, Huang B, Li X, Yang N, Wang J. Valtrate, an iridoid compound in Valeriana, elicits anti-glioblastoma activity through inhibition of the PDGFRA/MEK/ERK signaling pathway. J Transl Med 2023; 21:147. [PMID: 36829235 PMCID: PMC9960449 DOI: 10.1186/s12967-023-03984-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/13/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Valtrate, a natural compound isolated from the root of Valeriana, exhibits antitumor activity in many cancers through different mechanisms. However, its efficacy for the treatment of glioblastoma (GBM), a tumor type with a poor prognosis, has not yet been rigorously investigated. METHODS GBM cell lines were treated with valtrate and CCK-8, colony formation and EdU assays, flow cytometry, and transwell, 3D tumor spheroid invasion and GBM-brain organoid co-culture invasion assays were performed to assess properties of proliferation, viability, apoptosis and invasion/migration. RNA sequencing analysis on valtrate-treated cells was performed to identify putative target genes underlying the antitumor activity of the drug in GBM cells. Western blot analysis, immunofluorescence and immunohistochemistry were performed to evaluate protein levels in valtrate-treated cell lines and in samples obtained from orthotopic xenografts. A specific activator of extracellular signal-regulated kinase (ERK) was used to identify the pathways mediating the effect. RESULTS Valtrate significantly inhibited the proliferation of GBM cells in vitro by inducing mitochondrial apoptosis and suppressed invasion and migration of GBM cells by inhibiting levels of proteins associated with epithelial mesenchymal transition (EMT). RNA sequencing analysis of valtrate-treated GBM cells revealed platelet-derived growth factor receptor A (PDGFRA) as a potential target downregulated by the drug. Analysis of PDGFRA protein and downstream mediators demonstrated that valtrate inhibited PDGFRA/MEK/ERK signaling. Finally, treatment of tumor-bearing nude mice with valtrate led to decreased tumor volume (fivefold difference at day 28) and enhanced survival (day 27 vs day 36, control vs valtrate-treated) relative to controls. CONCLUSIONS Taken together, our study demonstrated that the natural product valtrate elicits antitumor activity in GBM cells through targeting PDGFRA and thus provides a candidate therapeutic compound for the treatment of GBM.
Collapse
Affiliation(s)
- Xuemeng Liu
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Yaotian Hu
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Zhiyi Xue
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Xun Zhang
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Xiaofei Liu
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Guowei Liu
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Muzi Wen
- grid.284723.80000 0000 8877 7471School of Public Health, Southern Medical University, Foushan, 528000 China
| | - Anjing Chen
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Bin Huang
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Xingang Li
- grid.452402.50000 0004 1808 3430Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117 China
| | - Ning Yang
- Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012, China. .,Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117, China. .,Department of Epidemiology and Health Statistics, School of Public Health, Shandong University, Jinan, 250012, China.
| | - Jian Wang
- Department of Neurosurgery, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Qilu Hospital, Shandong University, Jinan, 250012, China. .,Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250117, China. .,Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009, Bergen, Norway.
| |
Collapse
|
8
|
McLaughlin A, Lakshmanan R, Dyke J, Warne R, Saha S, Lind C, Bynevelt M. Diffuse paediatric cerebellar glioma: two identical imaging phenotypes of an extremely rare entity with disparate pathology. Childs Nerv Syst 2023; 39:857-861. [PMID: 36658366 DOI: 10.1007/s00381-023-05836-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023]
Abstract
Although the posterior fossa is a common location for paediatric brain tumours [1], diffuse glioma isolated to the cerebellum is an extremely rare imaging entity. Only two cases of isolated diffuse paediatric cerebellar glioma have been reported in the English language to the best of our knowledge [2, 3], and only one of these cases had a similar imaging phenotype to our cases [3]. Although somewhat similar to Lhermitte-Duclos (dysplastic gangliocytoma of the cerebellum), the appearances are distinct from other neoplastic entities of the paediatric posterior fossa. Clinical presentation and neurological examination findings are vital however to help differentiate other diffuse pathologies involving the cerebellum such as rhombencephalitis. Presented here are two diffuse cerebellar gliomas in children under the age of 3 with near identical imaging phenotypes demonstrating differing histological and molecular genetic profiles.
Collapse
Affiliation(s)
- Aden McLaughlin
- Department of Medical Imaging, Perth Children's Hospital, Nedlands, Perth, WA, 6009, Australia.
| | - Rahul Lakshmanan
- Department of Medical Imaging, Perth Children's Hospital, Nedlands, Perth, WA, 6009, Australia
| | - Jason Dyke
- PathWest Neuropathology, Royal Perth Hospital, Perth, WA, 6000, Australia
- School of Medicine and Pharmacology, University of Western Australia, Crawley, WA, 6009, Australia
| | - Richard Warne
- Department of Medical Imaging, Perth Children's Hospital, Nedlands, Perth, WA, 6009, Australia
| | - Snigdha Saha
- Perth Children's Hospital Department of Neurosurgery, Nedlands, Perth, WA, 6009, Australia
| | - Christopher Lind
- Perth Children's Hospital Department of Neurosurgery, Nedlands, Perth, WA, 6009, Australia
| | - Michael Bynevelt
- Department of Medical Imaging, Perth Children's Hospital, Nedlands, Perth, WA, 6009, Australia
- Neurological Intervention and Imaging Service of Western Australia (NIISwa), Nedlands, Perth, WA, 6009, Australia
| |
Collapse
|
9
|
Li Y, Yu H, Ma Q, Wei M, Liu X, Qi Y, Li C, Dong L, Zhang H. si-PDGFR β-Loaded Exosomes Suppress the Progression of Glioma by Inhibiting the Oxidative Associated PI3K/Akt/EZH2 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5081439. [PMID: 36275907 PMCID: PMC9586723 DOI: 10.1155/2022/5081439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/19/2022] [Accepted: 09/29/2022] [Indexed: 11/24/2022]
Abstract
This study investigated the possibility of exosomes loaded with si-PDGFRβ ability to suppress the progression of glioma. Common gliomas develop from neuroglial progenitor cells. Many variables affect the survival rate and occurrence of gliomas. Understanding oxidative stress processes and creating new, efficient treatments are crucial because oxidative stress is linked to the development of brain tumors. For this purpose, selected clinical samples were subjected to various tests like quantitative real-time PCR, Cignal Finder RTK signaling 7-pathway reporter array analysis, CCK-8 analysis, flow cytometry, and immunoblotting. Here, we demonstrated that PDGFRβ expression was increased in glioma patients. Following that, cell-derived exosomes were extracted and collected and traced in vivo, and selected tissue samples were subjected to immunohistochemical analysis. The results indicated that the knockdown of PDGFRβ (si-PDGFRβ) inhibited the proliferation of glioma cells. Besides this, si-PDGFRβ-loaded exosomes induced a similar antitumor effect in glioma cells. The anticancer effect of si-PDGFRβ-loaded exosomes was mediated by the inactivation of the PI3K/Akt/EZH2 pathway. Finally, we verified that this exosome delivery system, si-PDGFRβ-loaded exosomes, had robust targeting and no associated toxicity. In conclusion, the study confirmed that si-PDGFRβ-loaded exosomes inhibit glioma progression via inactivating the PI3K/Akt/EZH2 signaling pathway.
Collapse
Affiliation(s)
- Yuping Li
- Neuro Intensive Care Unit, Clinical Medical College, Yangzhou University, Yangzhou, China
- Department of Neurosurgery, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Hailong Yu
- Neuro Intensive Care Unit, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Qiang Ma
- Neuro Intensive Care Unit, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Min Wei
- Department of Neurosurgery, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Xiaoguang Liu
- Neuro Intensive Care Unit, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yajie Qi
- Neuro Intensive Care Unit, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Chen Li
- Department of Neurosurgery, Changzhou No. 2 People's Hospital, Changzhou, Jiangsu, China
| | - Lun Dong
- Neuro Intensive Care Unit, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Hengzhu Zhang
- Department of Neurosurgery, Clinical Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
10
|
Kaissarian NM, Meyer D, Kimchi-Sarfaty C. Synonymous Variants: Necessary Nuance in our Understanding of Cancer Drivers and Treatment Outcomes. J Natl Cancer Inst 2022; 114:1072-1094. [PMID: 35477782 PMCID: PMC9360466 DOI: 10.1093/jnci/djac090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/24/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
Once called "silent mutations" and assumed to have no effect on protein structure and function, synonymous variants are now recognized to be drivers for some cancers. There have been significant advances in our understanding of the numerous mechanisms by which synonymous single nucleotide variants (sSNVs) can affect protein structure and function by affecting pre-mRNA splicing, mRNA expression, stability, folding, miRNA binding, translation kinetics, and co-translational folding. This review highlights the need for considering sSNVs in cancer biology to gain a better understanding of the genetic determinants of human cancers and to improve their diagnosis and treatment. We surveyed the literature for reports of sSNVs in cancer and found numerous studies on the consequences of sSNVs on gene function with supporting in vitro evidence. We also found reports of sSNVs that have statistically significant associations with specific cancer types but for which in vitro studies are lacking to support the reported associations. Additionally, we found reports of germline and somatic sSNVs that were observed in numerous clinical studies and for which in silico analysis predicts possible effects on gene function. We provide a review of these investigations and discuss necessary future studies to elucidate the mechanisms by which sSNVs disrupt protein function and are play a role in tumorigeneses, cancer progression, and treatment efficacy. As splicing dysregulation is one of the most well recognized mechanisms by which sSNVs impact protein function, we also include our own in silico analysis for predicting which sSNVs may disrupt pre-mRNA splicing.
Collapse
Affiliation(s)
- Nayiri M Kaissarian
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation & Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Douglas Meyer
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation & Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Chava Kimchi-Sarfaty
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation & Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
11
|
Li SY, Johnson R, Smyth LC, Dragunow M. Platelet-derived growth factor signalling in neurovascular function and disease. Int J Biochem Cell Biol 2022; 145:106187. [PMID: 35217189 DOI: 10.1016/j.biocel.2022.106187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 11/25/2022]
Abstract
Platelet-derived growth factors are critical for cerebrovascular development and homeostasis. Abnormalities in this signalling pathway are implicated in neurological diseases, especially those where neurovascular dysfunction and neuroinflammation plays a prominent role in disease pathologies, such as stroke and Alzheimer's disease; the angiogenic nature of this pathway also draws its significance in brain malignancies such as glioblastoma where tumour angiogenesis is profuse. In this review, we provide an updated overview of the actions of the platelet-derived growth factors on neurovascular function, their role in the regulation of perivascular cell types expressing the cognate receptors, neurological diseases associated with aberrance in signalling, and highlight the clinical relevance and therapeutic potentials of this pathway for central nervous system diseases.
Collapse
Affiliation(s)
- Susan Ys Li
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Rebecca Johnson
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Leon Cd Smyth
- Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University in St Louis, MO, USA.
| | - Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
12
|
Vuong HG, Le HT, Dunn IF. The prognostic significance of further genotyping H3G34 diffuse hemispheric gliomas. Cancer 2022; 128:1907-1912. [PMID: 35195909 DOI: 10.1002/cncr.34156] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/31/2022] [Accepted: 02/08/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND H3G34-mutant diffuse hemispheric glioma (DHG) is recognized as a new, distinct entity in the latest World Health Organization classification for central nervous system tumors and is associated with a particularly aggressive course. The authors performed a systematic review and pooled analysis to investigate the frequency of genetic events in these tumors and to determine whether these events were associated with survival trends. METHODS Two electronic databases were accessed to search for relevant data. Included criteria were studies that had individual patient data on H3.3 G34-mutant gliomas. To analyze the impact of genetic events on overall survival, Kaplan-Meier analysis and Cox regression models were used, and corresponding hazard ratios and 95% confidence intervals were computed. RESULTS In total, 20 studies with 257 H3G34-mutant DHGs were included for integrated analyses. The H3 glycine-to-valine (H3G34V) mutation showed a significantly worse prognosis than the glycine-to-arginine (H3G34R) mutation (median overall survival, 9.9 vs 14.8 months; hazard ratio, 3.040; 95% confidence interval, 1.208-7.651; P = .018), and this result remained statistically significant in the multivariate Cox regression model. Among H3G34 DHGs, TP53 mutation was the most common genetic alteration (94.9%), followed by ATRX alterations (87.5%), MGMT methylation (79.5%), and PDGFRA alterations (33.2%). The presence of PDGFRA amplification or EGFR amplification conferred poor survival. After adjusting for age and sex, these alterations were still independent indicators for adverse outcomes. CONCLUSIONS The authors highlight the important role of molecular stratification of H3G34 DHGs, which may help refine our understanding of the natural history of this group of malignant tumors.
Collapse
Affiliation(s)
- Huy Gia Vuong
- Department of Neurosurgery, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma
| | - Hieu Trong Le
- Department of Pathology, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Ian F Dunn
- Department of Neurosurgery, Oklahoma University Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
13
|
Sun KH(M, Wong YT(H, Cheung KM(C, Yuen C(M, Chan YT(T, Lai WY(J, Chao C(D, Fan WS(K, Chow YK(K, Law MF, Tam HC(T. Update on Molecular Diagnosis in Extranodal NK/T-Cell Lymphoma and Its Role in the Era of Personalized Medicine. Diagnostics (Basel) 2022; 12:diagnostics12020409. [PMID: 35204500 PMCID: PMC8871212 DOI: 10.3390/diagnostics12020409] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK)/T-cell lymphoma (NKTCL) is an aggressive malignancy with unique epidemiological, histological, molecular, and clinical characteristics. It occurs in two pathological forms, namely, extranodal NKTCL (ENKTCL) and aggressive NK leukemia, according to the latest World Health Organization (WHO) classification. Epstein–Barr virus (EBV) infection has long been proposed as the major etiology of lymphomagenesis. The adoption of high-throughput sequencing has allowed us to gain more insight into the molecular mechanisms of ENKTCL, which largely involve chromosome deletion and aberrations in Janus kinase (JAK)-signal transducer and activator of transcription (STAT), programmed cell death protein-1 (PD-1)/PD-ligand 1 (PD-L1) pathways, as well as mutations in tumor suppressor genes. The molecular findings could potentially influence the traditional chemoradiotherapy approach, which is known to be associated with significant toxicity. This article will review the latest molecular findings in NKTCL and recent advances in the field of molecular diagnosis in NKTCL. Issues of quality control and technical difficulties will also be discussed, along with future prospects in the molecular diagnosis and treatment of NKTCL.
Collapse
Affiliation(s)
- Ka-Hei (Murphy) Sun
- Division of Hematopathology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, Hong Kong; (K.-H.S.); (C.Y.)
| | | | - Ka-Man (Carmen) Cheung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Carmen (Michelle) Yuen
- Division of Hematopathology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, Hong Kong; (K.-H.S.); (C.Y.)
| | - Yun-Tat (Ted) Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Wing-Yan (Jennifer) Lai
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Chun (David) Chao
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Wing-Sum (Katie) Fan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Yuen-Kiu (Karen) Chow
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| | - Man-Fai Law
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
- Correspondence:
| | - Ho-Chi (Tommy) Tam
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Hong Kong; (K.-M.C.); (Y.-T.C.); (W.-Y.L.); (C.C.); (W.-S.F.); (Y.-K.C.); (H.-C.T.)
| |
Collapse
|
14
|
Chen S, Zhang Z, Zhang B, Huang Q, Liu Y, Qiu Y, Long X, Wu M, Zhang Z. CircCDK14 Promotes Tumor Progression and Resists Ferroptosis in Glioma by Regulating PDGFRA. Int J Biol Sci 2022; 18:841-857. [PMID: 35002529 PMCID: PMC8741855 DOI: 10.7150/ijbs.66114] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/02/2021] [Indexed: 12/28/2022] Open
Abstract
CircRNAs have garnered significant interest in recent years due to their regulation in human tumorigenesis, yet, the function of most glioma-related circRNAs remains unclear. In this study, using RNA-Seq, we screened differentially regulated circRNAs in glioma, in comparison to non-tumor brain tissue. Loss- and gain-of-function strategies were used to assess the effect of circCDK14 on tumor progression both in vitro and in vivo. Luciferase reporter, RNA pull-down and fluorescence in situ hybridization assays were carried out to validate interactions between circCDK14 and miR-3938 as well as miR-3938 and PDGFRA. Transmission electron microscopic observation of mitochondria, iron and reactive oxygen species assays were employed for the detection of circCDK14 effect on glioma cells' sensitivity to erastin-induced ferroptosis (Fp). Our findings indicated that circCDK14 was overexpressed in glioma tissues and cell lines, and elevated levels of circCDK14 induced poor prognosis of glioma patients. CircCDK14 promotes the migration, invasion and proliferation of glioma cells in vitro as well as tumorigenesis in vivo. An evaluation of the underlying mechanism revealed that circCDK14 sponged miR-3938 to upregulate oncogenic gene PDGFRA expression. Moreover, we also found that circCDK14 reduced glioma cells' sensitivity to Fp by regulating PDGFRA expression. In conclusion, circCDK14 induces tumor in glioma and increases malignant tumor behavior via the miR-3938/PDGFRA axis. Hence, the miR-3938/PDGFRA axis may be an excellent candidate of anti-glioma therapy.
Collapse
Affiliation(s)
- Simin Chen
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Department of Clinical Laboratory, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Zhaoyu Zhang
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Cancer Research Institute, Central South University, Changsha 410013, Hunan, China
| | - Baoxin Zhang
- Armed Police Hospital of Hunan Province, Changsha 410013, Hunan, China
| | - Qing Huang
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Department of Clinical Laboratory, Yueyang Central Hospital, Yueyang 414000, Hunan, China
| | - Yi Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yi Qiu
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Cancer Research Institute, Central South University, Changsha 410013, Hunan, China
| | - Xinmiao Long
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China.,Cancer Research Institute, Central South University, Changsha 410013, Hunan, China
| | - Minghua Wu
- Cancer Research Institute, Central South University, Changsha 410013, Hunan, China
| | - Zuping Zhang
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
15
|
Higa N, Akahane T, Yokoyama S, Yonezawa H, Uchida H, Takajo T, Otsuji R, Hamada T, Matsuo K, Kirishima M, Hata N, Hanaya R, Tanimoto A, Yoshimoto K. Prognostic impact of PDGFRA gain/amplification and MGMT promoter methylation status in patients with IDH wild-type glioblastoma. Neurooncol Adv 2022; 4:vdac097. [PMID: 35911637 PMCID: PMC9332894 DOI: 10.1093/noajnl/vdac097] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background Platelet-derived growth factor receptor alpha (PDGFRA) is the second most frequently mutated tyrosine kinase receptor in glioblastoma (GBM). However, the prognostic impact of PDGFRA amplification on GBM patients remains unclear. Herein, we evaluated this impact by retrospectively analyzing outcomes of patients with IDH wild-type GBM. Methods Using a custom-made oncopanel, we evaluated PDGFRA gain/amplification in 107 GBM samples harboring wild-type IDH, along with MGMT promoter (MGMTp) methylation status. Results We detected PDGFRA gain/amplification in 31 samples (29.0%). PDGFRA gain/amplification predicted poor prognosis (P = .003). Compared to unamplified PDGFRA, PDGFRA gain/amplification in GBM was associated with higher patient age (P = .031), higher Ki-67 score (P = .019), and lower extent of surgical resection (P = .033). Unmethylated MGMTp also predicted poor prognosis (P = .005). As PDGFRA gain/amplification and unmethylated MGMTp were independent factors for poor prognosis in multivariate analyses, we grouped GBM cases based on PDGFRA and MGMTp status: poor (PDGFRA gain/amplification and unmethylated MGMTp), intermediate (PDGFRA gain/amplification or unmethylated MGMTp), and good (PDGFRA intact and methylated MGMTp) prognosis. The Kaplan-Meier survival analysis indicated that these groups significantly correlated with the OS of GBM patients (P < .001). Conclusions Here we report that PDGFRA gain/amplification is a predictor of poor prognosis in IDH wild-type GBM. Combining PDGFRA gain/amplification with MGMTp methylation status improves individual prognosis prediction in patients with IDH wild-type GBM.
Collapse
Affiliation(s)
- Nayuta Higa
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
| | - Toshiaki Akahane
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
- Center for Human Genome and Gene Analysis, Kagoshima University Hospital , Kagoshima-City, Kagoshima , Japan
| | - Seiya Yokoyama
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
| | - Hajime Yonezawa
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
| | - Hiroyuki Uchida
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
| | - Tomoko Takajo
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
| | - Ryosuke Otsuji
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University , Fukuoka , Japan
| | - Taiji Hamada
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
| | - Kei Matsuo
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
| | - Mari Kirishima
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
| | - Nobuhiro Hata
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University , Fukuoka , Japan
| | - Ryosuke Hanaya
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
| | - Akihide Tanimoto
- Department of Pathology, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
- Center for Human Genome and Gene Analysis, Kagoshima University Hospital , Kagoshima-City, Kagoshima , Japan
| | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima-City, Kagoshima , Japan
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University , Fukuoka , Japan
| |
Collapse
|
16
|
Aldaz P, Arozarena I. Tyrosine Kinase Inhibitors in Adult Glioblastoma: An (Un)Closed Chapter? Cancers (Basel) 2021; 13:5799. [PMID: 34830952 PMCID: PMC8616487 DOI: 10.3390/cancers13225799] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal form of malignant brain tumor. GBM patients normally undergo surgery plus adjuvant radiotherapy followed by chemotherapy. Numerous studies into the molecular events driving GBM highlight the central role played by the Epidermal Growth Factor Receptor (EGFR), as well as the Platelet-derived Growth Factor Receptors PDGFRA and PDGFRB in tumor initiation and progression. Despite strong preclinical evidence for the therapeutic potential of tyrosine kinase inhibitors (TKIs) that target EGFR, PDGFRs, and other tyrosine kinases, clinical trials performed during the last 20 years have not led to the desired therapeutic breakthrough for GBM patients. While clinical trials are still ongoing, in the medical community there is the perception of TKIs as a lost opportunity in the fight against GBM. In this article, we review the scientific rationale for the use of TKIs targeting glioma drivers. We critically analyze the potential causes for the failure of TKIs in the treatment of GBM, and we propose alternative approaches to the clinical evaluation of TKIs in GBM patients.
Collapse
Affiliation(s)
- Paula Aldaz
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), 31008 Pamplona, Spain
| | - Imanol Arozarena
- Cancer Signaling Unit, Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain
- Health Research Institute of Navarre (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
17
|
Cui M, Gao X, Chi Y, Zhang M, Lin H, Chen H, Sun C, Ma X. Molecular Alterations and Their Correlation With the Survival of Glioblastoma Patients With Corpus Callosum Involvement. Front Neurosci 2021; 15:701426. [PMID: 34393714 PMCID: PMC8361605 DOI: 10.3389/fnins.2021.701426] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/08/2021] [Indexed: 12/21/2022] Open
Abstract
Purpose: To explore molecular alterations and their correlation with the survival of patients with glioblastoma (GBM) with corpus callosum (CC) involvement (ccGBM). Methods: Electronic medical records were reviewed for glioma patients tested for molecular alterations and treated at our hospital between January 2016 and July 2020. ccGBM was compared to GBM without CC involvement (non-ccGBM) to identify differences in molecular alterations. Clinical outcomes and survival were compared between ccGBM and non-ccGBM patients, as well as among patients with ccGBM with different molecular alteration statuses. ccGBM was also compared to diffuse midline glioma (DMG) to clarify their correlation in molecular alterations, the progression-free survival (PFS), and overall survival (OS). Results: Thirty ccGBM and 88 non-ccGBM patients were included. PDGFRA amplification (PDGFRAamp, 33.3 vs. 9.1%, P = 0.004) and missense mutation (PDGFRAmut, 20.0 vs. 3.4%, P = 0.011) both had higher incidences in ccGBM than in non-ccGBM. PDGFRA alteration was associated with the occurrence of ccGBM (OR = 4.91 [95% CI: 1.55–15.52], P = 0.007). ccGBM with PDGFRAamp resulted in a shorter median PFS (8.6 vs. 13.5 months, P = 0.025) and OS (12.4 vs. 17.9 months, P = 0.022) than non-ccGBM with PDGFRAnon-amp. ccGBM with PDGFRAamp combined with PDGFRAmut (PDGFRAamp-mut) had a shorter median PFS (7.6 vs. 8.9 months, P = 0.022) and OS (9.6 vs. 17.8 months, P = 0.006) than non-ccGBM with wild-type PDGFRA and no amplification (PDGFRA-w, non-amp). Compared to ccGBM with PDGFRA-w, non-amp, ccGBM with PDGFRAamp and PDGFRAamp-mut both had a shorter median PFS and OS (P < 0.05). The hazard ratios (HRs) of PDGFRAamp for PFS and OS in ccGBM were 3.08 (95% CI: 1.02–9.35, P = 0.047) and 5.07 (1.52–16.89, P = 0.008), respectively, and the HRs of PDGFRAamp-mut for PFS and OS were 13.16 (95% CI: 3.19–54.40, P < 0.001) and 16.36 (2.66–100.70, P = 0.003). ccGBM may have similar incidences of PDGFRAamp or mut (PDGFRAamp/mut) as DMG, and they also had similar median PFS (10.9 vs. 9.0 months, P = 0.558) and OS (16.8 vs. 11.5 months, P = 0.510). Conclusion:PDGFRA alterations are significantly associated with the occurrence and poor prognosis of ccGBM. ccGBM with PDGFRAamp/mut may be classified as a single subtype of GBM that has a similar survival rate to DMG. PDGFR inhibitors may be a promising treatment method for ccGBM.
Collapse
Affiliation(s)
- Meng Cui
- Medical School of Chinese PLA, Beijing, China.,Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xin Gao
- Medical School of Chinese PLA, Beijing, China.,Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yihong Chi
- Department of Information Technology, Xian Janssen Pharmaceutical Ltd., Beijing, China
| | - Meng Zhang
- Medical School of Chinese PLA, Beijing, China.,Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Hepu Lin
- Department of Neurosurgery, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Hewen Chen
- Medical School of Chinese PLA, Beijing, China.,Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Caihong Sun
- Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xiaodong Ma
- Medical School of Chinese PLA, Beijing, China.,Department of Neurosurgery, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
18
|
Pienkowski T, Kowalczyk T, Kretowski A, Ciborowski M. A review of gliomas-related proteins. Characteristics of potential biomarkers. Am J Cancer Res 2021; 11:3425-3444. [PMID: 34354853 PMCID: PMC8332856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/15/2021] [Indexed: 06/13/2023] Open
Abstract
Brain tumors are one of the most commonly diagnosed cancers of the central nervous system. Of all diagnosed malignant tumors, 80% are gliomas. An unequivocal diagnosis of gliomas is not always simple, and there is a great need for research to find new treatment options and diagnostic approaches. This paper is focused on the glioma-related protein profiles as compared to healthy brain tissue, which is reflected in multiple correlations between biological aspects that influence proliferation, apoptosis evasion and the invasiveness of neoplastic cells. The work presents the possibilities of facilitating clinical practice with proteomic biomarkers, which offer a wider diagnostic spectrum and reduce the margin of mistake in histopathological or imaging diagnostic methods. In fact, many changes in the body's homeostasis can be overlooked due to the lack of symptoms or their non-specificity. Nevertheless, a single marker has limited reliability in distinguishing a particular tumor subtype, since the increased or decreased level of the protein of interest may differ between the stages or locations of the tumor. Moreover, the correlations between proposed proteins - presented in this paper - may help clinicians to choose the most optimal therapy, and estimate its effectiveness, or indicate new therapeutic targets affecting disrupted biochemical pathways.
Collapse
Affiliation(s)
- Tomasz Pienkowski
- Clinical Research Center, Medical University of Bialystok M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Tomasz Kowalczyk
- Clinical Research Center, Medical University of Bialystok M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Center, Medical University of Bialystok M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Center, Medical University of Bialystok M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| |
Collapse
|
19
|
Makino Y, Arakawa Y, Yoshioka E, Shofuda T, Kawauchi T, Terada Y, Tanji M, Kanematsu D, Mineharu Y, Miyamoto S, Kanemura Y. Prognostic stratification for IDH-wild-type lower-grade astrocytoma by Sanger sequencing and copy-number alteration analysis with MLPA. Sci Rep 2021; 11:14408. [PMID: 34257410 PMCID: PMC8277860 DOI: 10.1038/s41598-021-93937-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/30/2021] [Indexed: 12/21/2022] Open
Abstract
The characteristics of IDH-wild-type lower-grade astrocytoma remain unclear. According to cIMPACT-NOW update 3, IDH-wild-type astrocytomas with any of the following factors show poor prognosis: combination of chromosome 7 gain and 10 loss (+ 7/- 10), and/or EGFR amplification, and/or TERT promoter (TERTp) mutation. Multiplex ligation-dependent probe amplification (MLPA) can detect copy number alterations at reasonable cost. The purpose of this study was to identify a precise, cost-effective method for stratifying the prognosis of IDH-wild-type astrocytoma. Sanger sequencing, MLPA, and quantitative methylation-specific PCR were performed for 42 IDH-wild-type lower-grade astrocytomas surgically treated at Kyoto University Hospital, and overall survival was analysed for 40 patients who underwent first surgery. Of the 42 IDH-wild-type astrocytomas, 21 were classified as grade 4 using cIMPACT-NOW update 3 criteria and all had either TERTp mutation or EGFR amplification. Kaplan-Meier analysis confirmed the prognostic significance of cIMPACT-NOW criteria, and World Health Organization grade was also prognostic. Cox regression hazard model identified independent significant prognostic indicators of PTEN loss (risk ratio, 9.75; p < 0.001) and PDGFRA amplification (risk ratio, 13.9; p = 0.002). The classification recommended by cIMPACT-NOW update 3 could be completed using Sanger sequencing and MLPA. Survival analysis revealed PTEN and PDGFRA were significant prognostic factors for IDH-wild-type lower-grade astrocytoma.
Collapse
Affiliation(s)
- Yasuhide Makino
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan.
| | - Ema Yoshioka
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan
| | - Tomoko Shofuda
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan
| | - Takeshi Kawauchi
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan
| | - Yukinori Terada
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Masahiro Tanji
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Daisuke Kanematsu
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Yonehiro Kanemura
- Department of Biomedical Research and Innovation, Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan.
- Department of Neurosurgery, National Hospital Organization Osaka National Hospital, Osaka, Japan.
| |
Collapse
|
20
|
Liang M, Wang B, Schneider A, Vainshtein I, Roskos L. A Novel Pharmacodynamic Biomarker and Mechanistic Modeling Facilitate the Development of Tovetumab, a Monoclonal Antibody Directed Against Platelet-Derived Growth Factor Receptor Alpha, for Cancer Therapy. AAPS JOURNAL 2020; 23:4. [PMID: 33210183 DOI: 10.1208/s12248-020-00523-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/09/2020] [Indexed: 11/30/2022]
Abstract
Tovetumab (MEDI-575) is a fully human IgG2κ monoclonal antibody that specifically binds to human platelet-derived growth factor receptor alpha (PDGFRα) and blocks receptor signal transduction by PDGF ligands. The affinity of tovetumab determined using surface plasmon resonance technology and flow cytometry demonstrated comparable binding affinity for human and monkey PDGFRα. In single and repeat-dose monkey pharmacokinetic-pharmacodynamic (PK-PD) studies, tovetumab administration resulted in dose-dependent elevation of circulating levels of PDGF-AA, a member of the PDGF ligand family, due to displacement of PDGF-AA from PDGFRα by tovetumab and subsequent blockade of PDGFRα-mediated PDGF-AA degradation. As such, PDGF-AA accumulation is an indirect measurement of receptor occupancy and is a novel PD biomarker for tovetumab. The nonlinear PK of tovetumab and dose-dependent increase in circulating PDGF-AA profiles were well described by a novel mechanistic model, in which tovetumab and PDGF-AA compete for the binding to PDGFRα. To facilitate translational simulation, the internalization half-lives of PDGF-AA and tovetumab upon binding to PDGFRα were determined using confocal imaging to be 14 ± 4 min and 30 ± 8 min, respectively. By incorporating PDGFRα internalization kinetics, the model not only predicted the target receptor occupancy by tovetumab, but also the biologically active agonistic ligand-receptor complex. This work described a novel PD biomarker approach applicable for anti-receptor therapeutics and the first mechanistic model to delineate the in vivo tri-molecular system of a drug, its target receptor, and a competing endogenous ligand, which collectively have been used for optimal dose recommendation supporting clinical development of tovetumab.
Collapse
Affiliation(s)
- Meina Liang
- Clinical and Quantitative Pharmacology, BioPharmaceuticals Research and Development, AstraZeneca, 121 Oyster Point Blvd., South San Francisco, California, 94080, USA.
| | - Bing Wang
- Clinical and Quantitative Pharmacology, BioPharmaceuticals Research and Development, AstraZeneca, 121 Oyster Point Blvd., South San Francisco, California, 94080, USA.,Amador Bioscience, Pleasanton, California, 94588, USA
| | - Amy Schneider
- Clinical and Quantitative Pharmacology, BioPharmaceuticals Research and Development, AstraZeneca, 121 Oyster Point Blvd., South San Francisco, California, 94080, USA.,The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Inna Vainshtein
- Clinical and Quantitative Pharmacology, BioPharmaceuticals Research and Development, AstraZeneca, 121 Oyster Point Blvd., South San Francisco, California, 94080, USA.,Exelixis, Alameda, California, 94502, USA
| | - Lorin Roskos
- Clinical and Quantitative Pharmacology, BioPharmaceuticals Research and Development, AstraZeneca, 121 Oyster Point Blvd., South San Francisco, California, 94080, USA. .,Exelixis, Alameda, California, 94502, USA.
| |
Collapse
|
21
|
Caruso FP, Garofano L, D'Angelo F, Yu K, Tang F, Yuan J, Zhang J, Cerulo L, Pagnotta SM, Bedognetti D, Sims PA, Suvà M, Su XD, Lasorella A, Iavarone A, Ceccarelli M. A map of tumor-host interactions in glioma at single-cell resolution. Gigascience 2020; 9:giaa109. [PMID: 33155039 PMCID: PMC7645027 DOI: 10.1093/gigascience/giaa109] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/08/2020] [Accepted: 09/17/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Single-cell RNA sequencing is the reference technique for characterizing the heterogeneity of the tumor microenvironment. The composition of the various cell types making up the microenvironment can significantly affect the way in which the immune system activates cancer rejection mechanisms. Understanding the cross-talk signals between immune cells and cancer cells is of fundamental importance for the identification of immuno-oncology therapeutic targets. RESULTS We present a novel method, single-cell Tumor-Host Interaction tool (scTHI), to identify significantly activated ligand-receptor interactions across clusters of cells from single-cell RNA sequencing data. We apply our approach to uncover the ligand-receptor interactions in glioma using 6 publicly available human glioma datasets encompassing 57,060 gene expression profiles from 71 patients. By leveraging this large-scale collection we show that unexpected cross-talk partners are highly conserved across different datasets in the majority of the tumor samples. This suggests that shared cross-talk mechanisms exist in glioma. CONCLUSIONS Our results provide a complete map of the active tumor-host interaction pairs in glioma that can be therapeutically exploited to reduce the immunosuppressive action of the microenvironment in brain tumor.
Collapse
Affiliation(s)
- Francesca Pia Caruso
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples “Federico II”, Via Claudio 21, 80128 Naples, Italy
- Bioinformatics Lab, BIOGEM, Via Camporeale, 83031 Ariano Irpino, Italy
| | - Luciano Garofano
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples “Federico II”, Via Claudio 21, 80128 Naples, Italy
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Fulvio D'Angelo
- Bioinformatics Lab, BIOGEM, Via Camporeale, 83031 Ariano Irpino, Italy
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Kai Yu
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, 5 Yiheyuan Rd, Haidian District, 100871 Beijing, China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, 5 Yiheyuan Rd, Haidian District, 100871 Beijing, China
| | - Jinzhou Yuan
- Department of Science and Technologies, Università degli Studi del Sannio, Via de Sanctis, 82100 Benevento, Italy
- Cancer Program, Sidra Medicine, Al Luqta Street, Zone 52, Education City, 26999, Doha Qatar
| | - Jing Zhang
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Luigi Cerulo
- Bioinformatics Lab, BIOGEM, Via Camporeale, 83031 Ariano Irpino, Italy
- Department of Science and Technologies, Università degli Studi del Sannio, Via de Sanctis, 82100 Benevento, Italy
| | - Stefano M Pagnotta
- Department of Science and Technologies, Università degli Studi del Sannio, Via de Sanctis, 82100 Benevento, Italy
| | - Davide Bedognetti
- Cancer Program, Sidra Medicine, Al Luqta Street, Zone 52, Education City, 26999, Doha Qatar
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Viale Benedetto XV 10, 16132 Genoa, Italy
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 St Nicholas Ave, New York , NY 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Mario Suvà
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St, Boston, MA 02114, USA
- Broad Institute of Harvard and MIT, 415 Main St, Cambridge, MA 02142, USA
| | - Xiao-Dong Su
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, 5 Yiheyuan Rd, Haidian District, 100871 Beijing, China
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, 1130 St Nicholas Ave, New York , NY 10032 USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, 1130 St Nicholas Ave, New York , NY 10032 USA
- Department of Neurology, Columbia University Medical Center, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Michele Ceccarelli
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples “Federico II”, Via Claudio 21, 80128 Naples, Italy
- Bioinformatics Lab, BIOGEM, Via Camporeale, 83031 Ariano Irpino, Italy
| |
Collapse
|
22
|
Whitehouse JP, Howlett M, Hii H, Mayoh C, Wong M, Barahona P, Ajuyah P, White CL, Buntine MK, Dyke JM, Lee S, Valvi S, Stanley J, Andradas C, Carline B, Kuchibhotla M, Ekert PG, Cowley MJ, Gottardo NG, Endersby R. A Novel Orthotopic Patient-Derived Xenograft Model of Radiation-Induced Glioma Following Medulloblastoma. Cancers (Basel) 2020; 12:cancers12102937. [PMID: 33053751 PMCID: PMC7600047 DOI: 10.3390/cancers12102937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Radiation-induced glioma (RIG) is a highly aggressive brain cancer arising as a consequence of radiation therapy. We report a case of RIG that arose in the brain stem following treatment for paediatric medulloblastoma, and the development and characterisation of a matched orthotopic patient-derived xenograft (PDX) model (TK-RIG915). Patient and PDX tumours were analysed using DNA methylation profiling, whole genome sequencing (WGS) and RNA sequencing. While initially thought to be a diffuse intrinsic pontine glioma (DIPG) based on disease location, results from methylation profiling and WGS were not consistent with this diagnosis. Furthermore, clustering analyses based on RNA expression suggested the tumours were distinct from primary DIPG. Additional gene expression analysis demonstrated concordance with a published RIG expression profile. Multiple genetic alterations that enhance PI3K/AKT and Ras/Raf/MEK/ERK signalling were discovered in TK-RIG915 including an activating mutation in PIK3CA, upregulation of PDGFRA and AKT2, inactivating mutations in NF1, and a gain-of-function mutation in PTPN11. Additionally, deletion of CDKN2A/B, increased IDH1 expression, and decreased ARID1A expression were observed. Detection of phosphorylated S6, 4EBP1 and ERK via immunohistochemistry confirmed PI3K pathway and ERK activation. Here, we report one of the first PDX models for RIG, which recapitulates the patient disease and is molecularly distinct from primary brain stem glioma. Genetic interrogation of this model has enabled the identification of potential therapeutic vulnerabilities in this currently incurable disease.
Collapse
Affiliation(s)
- Jacqueline P. Whitehouse
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Meegan Howlett
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Hilary Hii
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
| | - Chelsea Mayoh
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
| | - Marie Wong
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
| | - Paulette Barahona
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
| | - Pamela Ajuyah
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
| | - Christine L. White
- Genetics and Molecular Pathology Laboratory, Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia; (C.L.W.); (M.K.B.)
- Department of Molecular and Translational Science, Monash University, Melbourne 3168, Victoria, Australia
| | - Molly K. Buntine
- Genetics and Molecular Pathology Laboratory, Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia; (C.L.W.); (M.K.B.)
- Department of Molecular and Translational Science, Monash University, Melbourne 3168, Victoria, Australia
| | - Jason M. Dyke
- Department of Neuropathology, PathWest Laboratory Medicine, Royal Perth Hospital, Perth 6000, Australia;
- Pathology and Laboratory Medicine, University of Western Australia, Nedlands 6009, Australia
| | - Sharon Lee
- Department of Neurosurgery, Perth Children’s Hospital, Nedlands 6009, Australia;
| | - Santosh Valvi
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Department of Paediatric and Adolescent Oncology/Haematology, Perth Children’s Hospital, Nedlands 6009, Australia
- Division of Paediatrics, University of Western Australia Medical School, Nedlands 6009, Australia
| | - Jason Stanley
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Clara Andradas
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Brooke Carline
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
| | - Mani Kuchibhotla
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
| | - Paul G. Ekert
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville 3052, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne 3000, Victoria, Australia
| | - Mark J. Cowley
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
| | - Nicholas G. Gottardo
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
- Department of Paediatric and Adolescent Oncology/Haematology, Perth Children’s Hospital, Nedlands 6009, Australia
| | - Raelene Endersby
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
- Correspondence:
| |
Collapse
|
23
|
Kim JY, Jo Y, Oh HK, Kim EH. Sorafenib increases tumor treating fields-induced cell death in glioblastoma by inhibiting STAT3. Am J Cancer Res 2020; 10:3475-3486. [PMID: 33163284 PMCID: PMC7642661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/22/2020] [Indexed: 06/11/2023] Open
Abstract
A newly diagnosed or recurrent Glioblastoma multiforme (GBM) can be treated with Tumor-treating fields (TTFields), an emerging type of alternative electric field-based therapy using low-intensity electric fields. TTFields have a penchant to arrest mitosis, eventually leading to apoptosis. Therefore, it is regarded as a potential anticancer therapy. However, in this study, we confirmed the combined efficacy of sorafenib and TTFields to improve the treatment efficiency of malignant GBM. Experimentation revealed the ability of sorafenib to decrease the signal transducer and activator of transcription 3 (STAT3) and this inhibition increased the sensitivity of TTFields in preventing tumor expansion. It was found that both combinatorial as well as monotherapy aimed to inhibit or reduce the level of STAT3, but the extent was different and based upon the reaction conditions. This drug is also capable of arresting multiple kinase pathways along with STAT3-related proteins (Mcl-1 and Survivin). STAT3 silencing can also be accomplished by RNA interference and can increase the TTFields-sensitizing effect of sorafenib. If the effects are reversed and gene regulating STAT3 is expressed more, it annihilates the effects of treatment. Moreover, sorafenib plus TTFields significantly inhibited xenograft tumor growth and combinatorial treatment reduced STAT3 expression more effectively in vivo. These in vitro and in vivo results indicate that sorafenib tends to sensitize GBM cells to TTFields-induced apoptosis by inhibiting STAT3.
Collapse
Affiliation(s)
- Jeong-Yub Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Yunhui Jo
- Department of Bio-Convergence Engineering, Korea UniversitySeoul 02842, Republic of Korea
| | - Hoon-Kyu Oh
- Department of Pathology, School of Medicine, Daegu Catholic UniversityNam-gu, Daegu, Republic of Korea
| | - Eun Ho Kim
- Department of Biochemistry, School of Medicine, Daegu Catholic UniversityNam-gu, Daegu, Republic of Korea
| |
Collapse
|
24
|
Wang L, Li LR, Zhang L, Wang JW. The landscape of new drugs in extranodal NK/T-cell lymphoma. Cancer Treat Rev 2020; 89:102065. [PMID: 32653806 DOI: 10.1016/j.ctrv.2020.102065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/11/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022]
Abstract
To date, much progress has been made in early-stage extranodal NK/T-cell lymphoma (ENKTCL), and risk-adapted therapy with radiotherapy (RT) alone for the low-risk group and RT combined with asparaginase-based chemotherapy (CT) for the high-risk group yields favorable outcomes. However, optimal treatment strategies have not been defined yet for advanced-stage ENKTCL. Historically, ENKTCL responded poorly to conventional anthracycline-based chemotherapy probably because of inherent multidrug resistance (MDR). The fact that ENKTCL cells lack asparagine synthetase (ASNS) activity warranted the use of L-asparaginase or pegaspargase as frontline chemotherapies. Even though, due to high mortality of the disease, approximately 50% patients failing the frontline therapy arrived at dismal clinical outcomes with a median progression-free survival (PFS) less than 8 months. As distinctive molecular and biological subgroups are increasingly discovered within the disease entity of ENKTCL, novel targeted therapies and immunotherapy are of the urgent need for those heterogeneous subgroups. In this review, we sought to summarize the preclinical and clinical results of 6 categories of promising targeted therapy and immunotherapy for the treatment of ENKTCL, including monoclonal antibodies, immune checkpoint inhibitors, small-molecular inhibitors, epigenetic therapy, immunomodulatory drugs, and adoptive T-cell therapy, and these might change the landscape of treatment for ENKTCL in the near future.
Collapse
Affiliation(s)
- Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing Tongren Hospital, Beijing 100730, China.
| | - Lin-Rong Li
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing 100032, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Beijing Key Laboratory of Nasal Disease, Beijing Institute of Otolaryngology, Beijing 100730, China
| | - Jing-Wen Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
25
|
Ahir BK, Engelhard HH, Lakka SS. Tumor Development and Angiogenesis in Adult Brain Tumor: Glioblastoma. Mol Neurobiol 2020; 57:2461-2478. [PMID: 32152825 PMCID: PMC7170819 DOI: 10.1007/s12035-020-01892-8] [Citation(s) in RCA: 251] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
Angiogenesis is the growth of new capillaries from the preexisting blood vessels. Glioblastoma (GBM) tumors are highly vascularized tumors, and glioma growth depends on the formation of new blood vessels. Angiogenesis is a complex process involving proliferation, migration, and differentiation of vascular endothelial cells (ECs) under the stimulation of specific signals. It is controlled by the balance between its promoting and inhibiting factors. Various angiogenic factors and genes have been identified that stimulate glioma angiogenesis. Therefore, attention has been directed to anti-angiogenesis therapy in which glioma proliferation is inhibited by inhibiting the formation of new tumor vessels using angiogenesis inhibitory factors and drugs. Here, in this review, we highlight and summarize the various molecular mediators that regulate GBM angiogenesis with focus on recent clinical research on the potential of exploiting angiogenic pathways as a strategy in the treatment of GBM patients.
Collapse
Affiliation(s)
- Bhavesh K Ahir
- Section of Hematology and Oncology, University of Illinois College of Medicine at Chicago, Chicago, IL, 60612, USA
| | - Herbert H Engelhard
- Department of Neurosurgery, University of Illinois College of Medicine at Chicago, Chicago, IL, 60612, USA
| | - Sajani S Lakka
- Section of Hematology and Oncology, University of Illinois College of Medicine at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
26
|
Receptor Tyrosine Kinases: Principles and Functions in Glioma Invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:151-178. [PMID: 32034713 DOI: 10.1007/978-3-030-30651-9_8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Protein tyrosine kinases are enzymes that are capable of adding a phosphate group to specific tyrosines on target proteins. A receptor tyrosine kinase (RTK) is a tyrosine kinase located at the cellular membrane and is activated by binding of a ligand via its extracellular domain. Protein phosphorylation by kinases is an important mechanism for communicating signals within a cell and regulating cellular activity; furthermore, this mechanism functions as an "on" or "off" switch in many cellular functions. Ninety unique tyrosine kinase genes, including 58 RTKs, were identified in the human genome; the products of these genes regulate cellular proliferation, survival, differentiation, function, and motility. Tyrosine kinases play a critical role in the development and progression of many types of cancer, in addition to their roles as key regulators of normal cellular processes. Recent studies have revealed that RTKs such as epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFR), c-Met, Tie, Axl, discoidin domain receptor 1 (DDR1), and erythropoietin-producing human hepatocellular carcinoma (Eph) play a major role in glioma invasion. Herein, we summarize recent advances in understanding the role of RTKs in glioma pathobiology, especially the invasive phenotype, and present the perspective that RTKs are a potential target of glioma therapy.
Collapse
|
27
|
Chondroitin sulfate synthase 1 enhances proliferation of glioblastoma by modulating PDGFRA stability. Oncogenesis 2020; 9:9. [PMID: 32019907 PMCID: PMC7000683 DOI: 10.1038/s41389-020-0197-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/13/2019] [Accepted: 01/16/2020] [Indexed: 11/13/2022] Open
Abstract
Chondroitin sulfate synthases, a family of enzyme involved in chondroitin sulfate (CS) polymerization, are dysregulated in various human malignancies, but their roles in glioma remain unclear. We performed database analysis and immunohistochemistry on human glioma tissue, to demonstrate that the expression of CHSY1 was frequently upregulated in glioma, and that it was associated with adverse clinicopathologic features, including high tumor grade and poor survival. Using a chondroitin sulfate-specific antibody, we showed that the expression of CHSY1 was significantly associated with CS formation in glioma tissue and cells. In addition, overexpression of CHSY1 in glioma cells enhanced cell viability and orthotopic tumor growth, whereas CHSY1 silencing suppressed malignant growth. Mechanistic investigations revealed that CHSY1 selectively regulates PDGFRA activation and PDGF-induced signaling in glioma cells by stabilizing PDGFRA protein levels. Inhibiting PDGFR activity with crenolanib decreased CHSY1-induced malignant characteristics of GL261 cells and prolonged survival in an orthotopic mouse model of glioma, which underlines the critical role of PDGFRA in mediating the effects of CHSY1. Taken together, these results provide information on CHSY1 expression and its role in glioma progression, and highlight novel insights into the significance of CHSY1 in PDGFRA signaling. Thus, our findings point to new molecular targets for glioma treatment.
Collapse
|
28
|
González-Tablas M, Arandia D, Jara-Acevedo M, Otero Á, Vital AL, Prieto C, González-Garcia N, Nieto-Librero AB, Tao H, Pascual D, Ruiz L, Sousa P, Galindo-Villardón P, Orfao A, Tabernero MD. Heterogeneous EGFR, CDK4, MDM4, and PDGFRA Gene Expression Profiles in Primary GBM: No Association with Patient Survival. Cancers (Basel) 2020; 12:cancers12010231. [PMID: 31963499 PMCID: PMC7016708 DOI: 10.3390/cancers12010231] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The prognostic impact of the expression profile of genes recurrently amplified in glioblastoma multiforme (GBM) remains controversial. METHODS We investigated the RNA gene expression profile of epidermal growth factor receptor (EGFR), cyclin-dependent kinase 4 (CDK4), murine doble minute 4 (MDM4), and platelet derived growth factor receptor alpha (PDGFRA) in 83 primary GBM tumors vs. 42 normal brain tissue samples. Interphase FISH (iFISH) analysis for the four genes, together with analysis of intragenic deletions in EGFR and PDGFRA, were evaluated in parallel at the DNA level. As validation cohort, publicly available RNA gene expression data on 293 samples from 10 different GBM patient series were also studied. RESULTS At the RNA level, CDK4 was the most frequently overexpressed gene (90%) followed by EGFR (58%) and PDGFRA (58%). Chromosome 7 copy number alterations, i.e., trisomy (49%) and polysomy (44%), showed no clear association with EGFR gene expression levels. In turn, intragenic EGFR deletions were found in 39 patients (47%), including EGFRvIII (46%) in association with EGFRvIVa (4%), EGFRvII (2%) or other EGFR deletions (3%) and PDGFRA deletion of exons 8-9 was found in only two tumors (2%). CONCLUSIONS Overall, none of the gene expression profiles and/or intragenic EGFR deletions showed a significant impact on overall survival of GBM supporting the notion that other still unraveled features of the disease might play a more relevant prognostic role in GBM.
Collapse
Affiliation(s)
- María González-Tablas
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Centre for Cancer Research (CIC-IBMCC, CSIC/USAL, IBSAL) and Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Daniel Arandia
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | - María Jara-Acevedo
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Biomedical Research Networking Centre on Cancer–CIBER-CIBERONC, Institute of Health Carlos III, 28029 Madrid, Spain
- Sequencing DNA Service (NUCLEUS), University of Salamanca, 37007 Salamanca, Spain
| | - Álvaro Otero
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Ana-Luisa Vital
- Centre for Neuroscience and Cell Biology and Faculty of Pharmacy, University of Coimbra, 3004-561 Coimbra, Portugal;
| | - Carlos Prieto
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Bioinformatics Service (NUCLEUS), University of Salamanca, 37007 Salamanca, Spain
| | - Nerea González-Garcia
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Department of Statistics, University of Salamanca, 37007 Salamanca, Spain;
| | - Ana Belén Nieto-Librero
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Department of Statistics, University of Salamanca, 37007 Salamanca, Spain;
| | - Herminio Tao
- Neurosurgery Service, University Hospital of Coimbra, 3004-561 Coimbra, Portugal;
| | - Daniel Pascual
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Laura Ruiz
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Pablo Sousa
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | | | - Alberto Orfao
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Centre for Cancer Research (CIC-IBMCC, CSIC/USAL, IBSAL) and Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Biomedical Research Networking Centre on Cancer–CIBER-CIBERONC, Institute of Health Carlos III, 28029 Madrid, Spain
- Correspondence: (A.O.); (M.D.T.); Tel.: +34923-29-11-00 (M.D.T.)
| | - María Dolores Tabernero
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Centre for Cancer Research (CIC-IBMCC, CSIC/USAL, IBSAL) and Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Biomedical Research Networking Centre on Cancer–CIBER-CIBERONC, Institute of Health Carlos III, 28029 Madrid, Spain
- Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL-IBSAL), 37007 Salamanca, Spain
- Correspondence: (A.O.); (M.D.T.); Tel.: +34923-29-11-00 (M.D.T.)
| |
Collapse
|
29
|
Ding Y, Wang X, Pan J, Ji M, Luo Z, Zhao P, Zhang Y, Wang G. Aberrant expression of long non-coding RNAs (lncRNAs) is involved in brain glioma development. Arch Med Sci 2020; 16:177-188. [PMID: 32051722 PMCID: PMC6963149 DOI: 10.5114/aoms.2020.91290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/01/2017] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Aberrant expression of long non-coding RNAs (lncRNAs) has been implicated in various diseases, including cancer. However, little is known about lncRNAs in human brain gliomas. MATERIAL AND METHODS We examined lncRNA profiles from three glioma specimens using lncRNA expression profiling microarrays. Quantitative real-time RT-PCR was used to analyze the differential expression of raw intensities of lncRNA expression in glioma and peritumoral tissues. RESULTS We found 4858 lncRNAs to be differentially expressed between tumor tissue and peritumoral tissue. Of these, 2845 lncRNAs were up-regulated (fold change > 3.0) and 2013 were down-regulated (fold change < 1/3). A total of 4084 messenger RNAs were also differentially expressed, including 2280 up-regulated transcripts (fold change > 3.0) and 1804 that were down-regulated (fold change < 1/3). Consistent with the microarray data, qPCR confirmed differential expression of these 6 lncRNAs (ak125809, ak098473, uc002ehu.1, bc043564, NR_027322, and uc003qmb.2) between tumor and peritumoral tissue. We next established co-expression networks of differentially expressed lncRNAs and mRNAs. Many mRNAs, such as LOC729991, NUDCD1, SHC3, PDGFA, and MDM2, and lncRNAs, such as ENST00000425922, ENST00000455568, uc002ukz.1, ENST00000502715, and NR_027873, have been shown to play important roles in glioma development. Consistent with this, pathway analysis revealed that "GLIOMA" (KEGG Pathway ID: hsa05214) was significantly enriched in tumor tissue. CONCLUSIONS Our data suggest that altered expression of lncRNAs may be a critical determinant of tumorigenesis in glioma patients.
Collapse
Affiliation(s)
- Yi Ding
- Department of Neurosurgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xinfa Wang
- Department of Neurosurgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Junchen Pan
- Department of Neurosurgery, Nanjing BenQ hospital, Nanjing, China
| | - Minjun Ji
- Department of Neurosurgery, Nanjing Medical University, Nanjing, China
| | - Zhengxiang Luo
- Department of Neurosurgery, Nanjing Brian Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Penglai Zhao
- Department of Neurosurgery, Nanjing Brian Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yansong Zhang
- Department of Neurosurgery, Nanjing Brian Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Gang Wang
- Department of Neurosurgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
30
|
Yang RR, Shi ZF, Zhang ZY, Chan AKY, Aibaidula A, Wang WW, Kwan JSH, Poon WS, Chen H, Li WC, Chung NYF, Punchhi G, Chu WCY, Chan ISH, Liu XZ, Mao Y, Li KKW, Ng HK. IDH mutant lower grade (WHO Grades II/III) astrocytomas can be stratified for risk by CDKN2A, CDK4 and PDGFRA copy number alterations. Brain Pathol 2019; 30:541-553. [PMID: 31733156 DOI: 10.1111/bpa.12801] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/29/2019] [Indexed: 12/31/2022] Open
Abstract
In the 2016, WHO classification of tumors of the central nervous system, isocitrate dehydrogenase (IDH) mutation is a main classifier for lower grade astrocytomas and IDH-mutated astrocytomas is now regarded as a single group with longer survival. However, the molecular and clinical heterogeneity among IDH mutant lower grade (WHO Grades II/III) astrocytomas have only rarely been investigated. In this study, we recruited 160 IDH mutant lower grade (WHO Grades II/III) astrocytomas, and examined PDGFRA amplification, CDKN2A deletion and CDK4 amplification by FISH analysis, TERT promoter mutation by Sanger sequencing and ATRX loss and p53 expression by immunohistochemistry. We identified PDGFRA amplification, CDKN2A homozygous deletion and CDK4 amplification in 18.8%, 15.0% and 18.1% of our cohort respectively, and these alterations occurred in a mutually exclusive fashion. PDGFRA amplification was associated with shorter PFS (P = 0.0003) and OS (P < 0.0001). In tumors without PDGFRA amplification, CDKN2A homozygous deletion or CDK4 amplification was associated with a shorter OS (P = 0.035). Tumors were divided into three risk groups based on the presence of molecular alterations: high risk (PDGFRA amplification), intermediate risk (CDKN2A deletion or CDK4 amplification) and low risk (neither CDKN2A deletion and CDK4 amplification nor PDGFRA amplification). These three risk groups were significantly different in overall survival with mean survivals of 40.5, 62.9 and 71.5 months. The high-risk group also demonstrated a shorter PFS compared to intermediate- (P = 0.036) and low-risk (P < 0.0001) groups. One limitation of this study is the relatively short follow-up period, a common confounding factor for studies on low-grade tumors. Our data illustrate that IDH mutant lower grade astrocytomas is not a homogeneous group and should be molecularly stratified for risk.
Collapse
Affiliation(s)
- Rui Ryan Yang
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China.,Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| | - Zhi-Feng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen-Yu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Aden Ka-Yin Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| | | | - Wei-Wei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Johnny Sheung Him Kwan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| | - Wai Sang Poon
- Department of Neurosurgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| | - Hong Chen
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wen-Cai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Nellie Yuk-Fei Chung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| | - Gopika Punchhi
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| | - William Ching-Yuen Chu
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| | - Ivan Sik-Hei Chan
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| | - Xian-Zhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Kay Ka-Wai Li
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| | - Ho-Keung Ng
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China SAR
| |
Collapse
|
31
|
Yang Y, Dodbele S, Park T, Glass R, Bhat K, Sulman EP, Zhang Y, Abounader R. MicroRNA-29a inhibits glioblastoma stem cells and tumor growth by regulating the PDGF pathway. J Neurooncol 2019; 145:23-34. [PMID: 31482267 PMCID: PMC10880555 DOI: 10.1007/s11060-019-03275-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/24/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE microRNAs are small noncoding RNAs that play important roles in cancer regulation. In this study, we investigated the expression, functional effects and mechanisms of action of microRNA-29a (miR-29a) in glioblastoma (GBM). METHODS miR-29a expression levels in GBM cells, stem cells (GSCs) and human tumors as well as normal astrocytes and normal brain were measured by quantitative PCR. miR-29a targets were uncovered by target prediction algorithms, and verified by immunoblotting and 3' UTR reporter assays. The effects of miR-29a on cell proliferation, death, migration and invasion were assessed with cell counting, Annexin V-PE/7AAD flow cytometry, scratch assay and transwell assay, respectively. Orthotopic xenografts were used to determine the effects of miR-29a on tumor growth. RESULTS Mir-29a was downregulated in human GBM specimens, GSCs and GBM cell lines. Exogenous expression of miR-29a inhibited GSC and GBM cell growth and induced apoptosis. miR-29a also inhibited GBM cell migration and invasion. PDGFC and PDGFA were uncovered and validated as direct targets of miR-29a in GBM. miR-29a downregulated PDGFC and PDGFA expressions at the transcriptional and translational levels. PDGFC and PDGFA expressions in GBM tumors, GSCs, and GBM established cell lines were higher than in normal brain and human astrocytes. Mir-29a expression inhibited orthotopic GBM xenograft growth. CONCLUSIONS miR-29a is a tumor suppressor miRNA in GBM, where it inhibits cancer stem cells and tumor growth by regulating the PDGF pathway.
Collapse
Affiliation(s)
- Yanzhi Yang
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, PO Box 800168, Charlottesville, VA, 22908, USA
| | - Samantha Dodbele
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, PO Box 800168, Charlottesville, VA, 22908, USA
| | - Thomas Park
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, PO Box 800168, Charlottesville, VA, 22908, USA
| | - Rainer Glass
- Neurosurgical Research, University Clinics Munich, Munich, Germany
| | - Krishna Bhat
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erik P Sulman
- Department of Radiation Oncology, NYU Langone School of Medicine, New York, USA
| | - Ying Zhang
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, PO Box 800168, Charlottesville, VA, 22908, USA.
| | - Roger Abounader
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, PO Box 800168, Charlottesville, VA, 22908, USA.
- Department of Neurology, University of Virginia, Charlottesville, VA, USA.
- Cancer Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
32
|
Ganapathi Sankaran D, Stemm-Wolf AJ, Pearson CG. CEP135 isoform dysregulation promotes centrosome amplification in breast cancer cells. Mol Biol Cell 2019; 30:1230-1244. [PMID: 30811267 PMCID: PMC6724517 DOI: 10.1091/mbc.e18-10-0674] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
The centrosome, composed of two centrioles surrounded by pericentriolar material, is the cell's central microtubule-organizing center. Centrosome duplication is coupled with the cell cycle such that centrosomes duplicate once in S phase. Loss of such coupling produces supernumerary centrosomes, a condition called centrosome amplification (CA). CA promotes cell invasion and chromosome instability, two hallmarks of cancer. We examined the contribution of centriole overduplication to CA and the consequences for genomic stability in breast cancer cells. CEP135, a centriole assembly protein, is dysregulated in some breast cancers. We previously identified a short isoform of CEP135, CEP135mini, that represses centriole duplication. Here, we show that the relative level of full-length CEP135 (CEP135full) to CEP135mini (the CEP135full:mini ratio) is increased in breast cancer cell lines with high CA. Inducing expression of CEP135full in breast cancer cells increases the frequency of CA, multipolar spindles, anaphase-lagging chromosomes, and micronuclei. Conversely, inducing expression of CEP135mini reduces centrosome number. The differential expression of the CEP135 isoforms in vivo is generated by alternative polyadenylation. Directed genetic mutations near the CEP135mini alternative polyadenylation signal reduces the CEP135full:mini ratio and decreases CA. We conclude that dysregulation of CEP135 isoforms promotes centriole overduplication and contributes to chromosome segregation errors in breast cancer cells.
Collapse
Affiliation(s)
- Divya Ganapathi Sankaran
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045-2537
| | - Alexander J. Stemm-Wolf
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045-2537
| | - Chad G. Pearson
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045-2537
| |
Collapse
|
33
|
Lau CH, Ho JWT, Lo PK, Tin C. Targeted Transgene Activation in the Brain Tissue by Systemic Delivery of Engineered AAV1 Expressing CRISPRa. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:637-649. [PMID: 31108320 PMCID: PMC6526230 DOI: 10.1016/j.omtn.2019.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 04/14/2019] [Accepted: 04/14/2019] [Indexed: 01/10/2023]
Abstract
Targeted transcriptional modulation in the central nervous system (CNS) can be achieved by adeno-associated virus (AAV) delivery of CRISPR activation (CRISPRa) and interference (CRISPRi) transgenes. To enable AAV packaging, we constructed minimal CRISPRa and CRISPRi transgenes by fusing catalytically inactive Staphylococcus aureus Cas9 (dSaCas9) to the transcriptional activator (VP64 and VP160) and repressor (KRAB and SID4X) domains along with truncated regulatory elements. We then evaluated the performance of these constructs in two reporter assays (bioluminescent and fluorescent), five endogenous genes (Camk2a, Mycn, Nrf2, Keap1, and PDGFRA), and two cell lines (neuro-2a [N2a] and U87) by targeting the promoter and/or enhancer regions. To enable systemic delivery of AAVs to the CNS, we have also generated an AAV1-PHP.B by inserting a 7-mer PHP.B peptide on AAV1 capsid. We showed that AAV1-PHP.B can efficiently cross the blood-brain barrier (BBB) and be taken up by the brain tissue upon lateral tail vein injection in mice. Importantly, a single-dose intravenous administration of AAV1-PHP.B expressing CRISPRa was shown to achieve targeted transgene activation in the mouse brain. This proof-of-concept study will contribute to the development of a non-invasive, specific and potent AAV-CRISPR system for correcting transcriptional misregulation in broad brain areas and multiple neuroanatomical structures.
Collapse
Affiliation(s)
- Cia-Hin Lau
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Jonathan Weng-Thim Ho
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Pik Kwan Lo
- Department of Chemistry, City University of Hong Kong, Kowloon Tong, Hong Kong; CityU Shenzhen Research Institute, Shenzhen, China
| | - Chung Tin
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong; CityU Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
34
|
Kang YJ, Cutler EG, Cho H. Therapeutic nanoplatforms and delivery strategies for neurological disorders. NANO CONVERGENCE 2018; 5:35. [PMID: 30499047 PMCID: PMC6265354 DOI: 10.1186/s40580-018-0168-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/20/2018] [Indexed: 05/26/2023]
Abstract
The major neurological disorders found in a central nervous system (CNS), such as brain tumors, Alzheimer's diseases, Parkinson's diseases, and Huntington's disease, have led to devastating outcomes on the human public health. Of these disorders, early diagnostics remains poor, and no treatment has been successfully discovered; therefore, they become the most life-threatening medical burdens worldwide compared to other major diseases. The major obstacles for the drug discovery are the presence of a restrictive blood-brain barrier (BBB), limiting drug entry into brains and undesired neuroimmune activities caused by untargeted drugs, leading to irreversible neuronal damages. Recent advances in nanotechnology have contributed to the development of novel nanoplatforms and effective delivering strategies to improve the CNS disorder treatment while less disturbing brain systems. The nanoscale drug carriers, including liposomes, dendrimers, viral capsids, polymeric nanoparticles, silicon nanoparticles, and magnetic/metallic nanoparticles, enable the effective drug delivery penetrating across the BBB, the aforementioned challenges in the CNS. Moreover, drugs encapsulated by the nanocarriers can reach further deeper into targeting regions while preventing the degradation. In this review, we classify novel disease hallmarks incorporated with emerging nanoplatforms, describe promising approaches for improving drug delivery to the disordered CNS, and discuss their implications for clinical practice.
Collapse
Affiliation(s)
- You Jung Kang
- Department of Mechanical Engineering and Engineering Science, Center for Biomedical Engineering and Science, Department of Biological Sciences, The Nanoscale Science Program, University of North Carolina at Charlotte, Charlotte, NC USA
| | - Eric Gerard Cutler
- Department of Mechanical Engineering and Engineering Science, Center for Biomedical Engineering and Science, Department of Biological Sciences, The Nanoscale Science Program, University of North Carolina at Charlotte, Charlotte, NC USA
| | - Hansang Cho
- Department of Mechanical Engineering and Engineering Science, Center for Biomedical Engineering and Science, Department of Biological Sciences, The Nanoscale Science Program, University of North Carolina at Charlotte, Charlotte, NC USA
| |
Collapse
|
35
|
Sumiyoshi K, Koso H, Watanabe S. Spontaneous development of intratumoral heterogeneity in a transposon-induced mouse model of glioma. Cancer Sci 2018; 109:1513-1523. [PMID: 29575648 PMCID: PMC5980157 DOI: 10.1111/cas.13579] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/19/2018] [Accepted: 03/13/2018] [Indexed: 01/20/2023] Open
Abstract
Glioma is the most common form of malignant brain cancer in adults. The Sleeping Beauty (SB) transposon‐based glioma mouse model allows for effective in vivo analysis of candidate genes. In the present study, we developed a transposon vector that encodes the triple combination of platelet‐derived growth factor subunit A (PDGFA), and shRNAs against Nf1 and Trp53 (shNf1/shp53). Initiation and progression of glioma in the brain were monitored by expression of a fluorescent protein. Transduction of the vector into neural progenitor and stem cells (NPC) in the subventricular zone (SVZ) of the neonatal brain induced proliferation of oligodendrocyte precursor cells, and promoted formation of highly penetrant malignant gliomas within 2‐4 months. Cells isolated from the tumors were capable of forming secondary tumors. Two transposon vectors, encoding either PDGFA or shNf1/shp53 were co‐electroporated into NPC. Cells expressing PDGFA or shNf1/shp53 were labeled with unique fluorescent proteins allowing visualization of the spatial distribution of cells with different genetic alterations within the same tumor. Tumor cells located at the center of tumors expressed PDGFA at higher levels than those located at the periphery, indicating that intratumoral heterogeneity in PDGFA expression levels spontaneously developed within the same tumor. Tumor cells comprising the palisading necrosis strongly expressed PDGFA, suggesting that PDGFA signaling is involved in hypoxic responses in glioma. The transposon vectors developed are compatible with any genetically engineered mouse model, providing a useful tool for the functional analysis of candidate genes in glioma.
Collapse
Affiliation(s)
- Keisuke Sumiyoshi
- Division of Molecular and Developmental Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hideto Koso
- Division of Molecular and Developmental Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sumiko Watanabe
- Division of Molecular and Developmental Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
36
|
Li YX, Shi Z, Aibaidula A, Chen H, Tang Q, Li KKW, Chung NYF, Chan DTM, Poon WS, Mao Y, Wu J, Zhou L, Chan AKY, Ng HK. Not all 1p/19q non-codeleted oligodendroglial tumors are astrocytic. Oncotarget 2018; 7:64615-64630. [PMID: 27556304 PMCID: PMC5323103 DOI: 10.18632/oncotarget.11378] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/12/2016] [Indexed: 12/21/2022] Open
Abstract
Although 1p/19q codeletion is the genetic hallmark defining oligodendrogliomas, approximately 30-40% of oligodendroglial tumors have intact 1p/19q in the literature and they demonstrate a worse prognosis. This group of 1p/19q intact oligodendroglial tumors is frequently suggested to be astrocytic in nature with TP53 and ATRX mutations but actually remains under-investigated. In the present study, we provided evidence that not all 1p/19q intact oligodendroglial tumors are astrocytic through histologic and molecular approaches. We examined 1p/19q status by FISH in a large cohort of 337 oligodendroglial tumors and identified 39.8% lacking 1p/19q codeletion which was independently associated with poor prognosis. Among this 1p/19q intact oligodendroglial tumor cohort, 58 cases demonstrated classic oligodendroglial histology which showed older patient age, better prognosis, association with grade III histology, PDGFRA expression, TERTp mutation, as well as frequent IDH mutation. More than half of the 1p/19q intact oligodendroglial tumors showed lack of astrocytic defining markers, p53 expression and ATRX loss. TP53 mutational analysis was additionally conducted in 45 cases of the 1p/19q intact oligodendroglial tumors. Wild-type TP53 was detected in 71.1% of cases which was associated with classic oligodendroglial histology. Importantly, IDH and TERTp co-occurred in 75% of 1p/19q intact, TP53 wild-type oligodendrogliomas, highlighting the potential of the co-mutations in assisting diagnosis of oligodendrogliomas in tumors with clear cell morphology and non-codeleted 1p/19q status. In summary, our study demonstrated that not all 1p/19q intact oligodendroglial tumors are astrocytic and co-evaluation of IDH and TERTp mutation could potentially serve as an adjunct for diagnosing 1p/19q intact oligodendrogliomas.
Collapse
Affiliation(s)
- Yan-Xi Li
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China.,Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | | | - Hong Chen
- Department of Neuropathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Kay Ka-Wai Li
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China
| | - Nellie Yuk-Fei Chung
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China
| | - Danny Tat-Ming Chan
- Neurosurgery Division, Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Wai Sang Poon
- Neurosurgery Division, Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Aden Ka-Yin Chan
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China
| | - Ho-Keung Ng
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
37
|
Connolly NP, Shetty AC, Stokum JA, Hoeschele I, Siegel MB, Miller CR, Kim AJ, Ho CY, Davila E, Simard JM, Devine SE, Rossmeisl JH, Holland EC, Winkles JA, Woodworth GF. Cross-species transcriptional analysis reveals conserved and host-specific neoplastic processes in mammalian glioma. Sci Rep 2018; 8:1180. [PMID: 29352201 PMCID: PMC5775420 DOI: 10.1038/s41598-018-19451-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/02/2018] [Indexed: 01/03/2023] Open
Abstract
Glioma is a unique neoplastic disease that develops exclusively in the central nervous system (CNS) and rarely metastasizes to other tissues. This feature strongly implicates the tumor-host CNS microenvironment in gliomagenesis and tumor progression. We investigated the differences and similarities in glioma biology as conveyed by transcriptomic patterns across four mammalian hosts: rats, mice, dogs, and humans. Given the inherent intra-tumoral molecular heterogeneity of human glioma, we focused this study on tumors with upregulation of the platelet-derived growth factor signaling axis, a common and early alteration in human gliomagenesis. The results reveal core neoplastic alterations in mammalian glioma, as well as unique contributions of the tumor host to neoplastic processes. Notable differences were observed in gene expression patterns as well as related biological pathways and cell populations known to mediate key elements of glioma biology, including angiogenesis, immune evasion, and brain invasion. These data provide new insights regarding mammalian models of human glioma, and how these insights and models relate to our current understanding of the human disease.
Collapse
Affiliation(s)
- Nina P Connolly
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Amol C Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jesse A Stokum
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ina Hoeschele
- Virginia Bioinformatics Institute and Department of Statistics, Virginia Tech, Blacksburg, Virginia, USA
| | - Marni B Siegel
- Departments of Pathology and Laboratory Medicine, Neurology, and Pharmacology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - C Ryan Miller
- Departments of Pathology and Laboratory Medicine, Neurology, and Pharmacology, Lineberger Comprehensive Cancer Center and Neuroscience Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cheng-Ying Ho
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Eduardo Davila
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Scott E Devine
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John H Rossmeisl
- Department of Small Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, USA.,Wake Forest University Baptist Health Comprehensive Cancer Center, Brain Tumor Center of Excellence, Winston-Salem, North Carolina, USA
| | - Eric C Holland
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | - Jeffrey A Winkles
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA. .,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
38
|
Heldin CH, Lennartsson J, Westermark B. Involvement of platelet-derived growth factor ligands and receptors in tumorigenesis. J Intern Med 2018; 283:16-44. [PMID: 28940884 DOI: 10.1111/joim.12690] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Platelet-derived growth factor (PDGF) isoforms and their receptors have important roles during embryogenesis, particularly in the development of various mesenchymal cell types in different organs. In the adult, PDGF stimulates wound healing and regulates tissue homeostasis. However, overactivity of PDGF signalling is associated with malignancies and other diseases characterized by excessive cell proliferation, such as fibrotic conditions and atherosclerosis. In certain tumours, genetic or epigenetic alterations of the genes for PDGF ligands and receptors drive tumour cell proliferation and survival. Examples include the rare skin tumour dermatofibrosarcoma protuberance, which is driven by autocrine PDGF stimulation due to translocation of a PDGF gene, and certain gastrointestinal stromal tumours and leukaemias, which are driven by constitute activation of PDGF receptors due to point mutations and formation of fusion proteins of the receptors, respectively. Moreover, PDGF stimulates cells in tumour stroma and promotes angiogenesis as well as the development of cancer-associated fibroblasts, both of which promote tumour progression. Inhibitors of PDGF signalling may thus be of clinical usefulness in the treatment of certain tumours.
Collapse
Affiliation(s)
- C-H Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - J Lennartsson
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - B Westermark
- Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
39
|
Xu R, Ji J, Zhang X, Han M, Zhang C, Xu Y, Wei Y, Wang S, Huang B, Chen A, Zhang D, Zhang Q, Li W, Jiang Z, Wang J, Li X. PDGFA/PDGFRα-regulated GOLM1 promotes human glioma progression through activation of AKT. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:193. [PMID: 29282077 PMCID: PMC5745991 DOI: 10.1186/s13046-017-0665-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/14/2017] [Indexed: 01/25/2023]
Abstract
BACKGROUND Golgi Membrane Protein 1 (GOLM1), a protein involved in the trafficking of proteins through the Golgi apparatus, has been shown to be oncogenic in a variety of human cancers. Here, we examined the role of GOLM1 in the development of human glioma. METHODS qRT-PCR, immunohistochemistry, and western blot analysis were performed to evaluate GOLM1 levels in cell lines and a cohort of primary human glioma and non-neoplastic brain tissue samples. Glioma cell lines were modified with lentiviral constructs expressing short hairpin RNAs targeting GOLM1 or overexpressing the protein to assess function in proliferation, viability, and migration and invasion in vitro using EdU, CCK8, clone-forming, Transwell assays, 3D tumor spheroid invasion assay and in vivo in orthotopic implantations. Protein lysates were used to screen a membrane-based antibody array to identify kinases mediated by GOLM1. Specific inhibitors of PDGFRα (AG1296) and AKT (MK-2206) were used to examine the regulation of PDGFA/PDGFRα on GOLM1 and the underlying pathway respectively. RESULTS qRT-PCR, immunohistochemistry and western blot analysis revealed GOLM1 expression to be elevated in glioma tissues and cell lines. Silencing of GOLM1 attenuated proliferation, migration, and invasion of U251, A172 and P3#GBM (primary glioma) cells, while overexpression of GOLM1 enhanced malignant behavior of U87MG cells. We further demonstrated that activation of AKT is the driving force of GOLM1-promoted glioma progression. The last finding of this research belongs to the regulation of PDGFA/PDGFRα on GOLM1, while GOLM1 was also a key element of PDGFA/PDGFRα-mediated activation of AKT, as well as the progression of glioma cells. CONCLUSIONS PDGFA/PDGFRα-regulated GOLM1 promotes glioma progression possibly through activation of a key signaling kinase, AKT. GOLM1 interference may therefore provide a novel therapeutic target and improve the efficacy of glioma treatment, particularly in the case of the proneural molecular subtype of human glioma.
Collapse
Affiliation(s)
- Ran Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Jianxiong Ji
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Chao Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Yangyang Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Yuzhen Wei
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China.,Department of Neurosurgery, Jining No.1 People's Hospital, Jiankang Road, Jining, 272011, China
| | - Shuai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Qing Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Zheng Jiang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China. .,Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, #107 Wenhua Xi Road, Jinan, 250012, China.
| |
Collapse
|
40
|
PDGFR inhibition mediated intracellular signalling in C6 glioma growth and migration: role of ERK and ROCK pathway. Cytotechnology 2017; 70:465-477. [PMID: 29143227 DOI: 10.1007/s10616-017-0163-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 10/31/2017] [Indexed: 12/11/2022] Open
Abstract
Aberrant PDGFR (Platelet derived growth factor receptor) signalling in brain tumors and gliomas is one of the primary cause of tumor progression. PDGFR stimulation by its ligand and the role of its downstream mediators such as extracellular regulated kinases (ERK1/2), PI3K and ROCK pathways have not been thoroughly investigated. The present study sought to investigate the role of PDGF receptor signalling inhibition on suppression of rat C6 glioma growth and migration. Treatment of C6 cells with PDGFR inhibitor, AG1295 caused a significant reduction in migration and proliferation by regulating the ERK and ROCK signalling. Subsequently, PDGFR blocking was demonstrated to regulate cytoskeleton reorganization by modulating the Actin-pMLC reorganization and pERK-FAK-Paxillin complex formation which may further regulate the C6 glioma migration. Further, other malignant behaviour of C6 glioma such as anchorage independent growth, adhesion, invasion and sphere forming abilities were found to be impaired by PDGFR blocking. PDGFR inhibition further regulates the C6 glioma tumor behaviour by inducing gene expression of GFAP, BDNF, and MECP2 and down regulating FAK expression. In conclusion, our data elucidate novel mechanisms involve in PDGFR inhibition mediated inhibition of C6 glioma growth and migration which can be a future potential target for the treatment of glioma.
Collapse
|
41
|
Zucca LE, Morini Matushita MA, da Silva Oliveira RJ, Scapulatempo-Neto C, de Lima MA, Ribeiro GG, Viana CR, Cárcano FM, Reis RM. Expression of tyrosine kinase receptor AXL is associated with worse outcome of metastatic renal cell carcinomas treated with sunitinib. Urol Oncol 2017; 36:11.e13-11.e21. [PMID: 28986088 DOI: 10.1016/j.urolonc.2017.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 02/01/2023]
Abstract
BACKGROUND Renal cell carcinoma (RCC) represents 2%-3% of all cancers of the Western countries. Currently, sunitinib, a receptor tyrosine kinase inhibitor, particularly of PDGF and VEGF receptors, is the first-line therapy for metastatic RCC (mRCC), with significant improvement in clinical outcome. However, there is a lack of predictive biomarkers of sunitinib response. Recently, others and our group suggested that the receptor tyrosine kinase AXL may modify the response to sunitinib. OBJECTIVE To study the expression of AXL in a series patients with of mRCC treated with sunitinib and to correlate it with patient's clinic-pathological features and therapeutic response. MATERIAL AND METHODS Sixty-four patients with mRCC (51 clear cell carcinomas (CCCs) and 13 non-CCCs) were evaluated for AXL expression by immunohistochemistry in the primary tumor. RESULTS AXL positivity was observed in 47% (30/64) of cases, namely in 43% (22/51) of CCCs and 61% (8/13) of non-CCC. Considering only the clear cell subtype, the univariate analysis showed that AXL expression was statistically associated with a poor prognosis, with a median overall survival of 13 months vs. 43 months in patients with negative AXL. In this subtype, along with the AXL positivity, other prognostic factors were absence of nephrectomy, Karnofsky performance status, more than 1 site of metastasis and liver metastasis. Moreover, AXL expression was associated with shorter progression to sunitinib. Overall, the multivariate survival analysis showed that absence of nephrectomy (HR = 4.85, P = 0.001), more than 1 site of metastasis (HR = 2.99, P = 0.002), bone metastasis (HR = 2.95, P = 0.001), together with AXL expression (HR = 2.01, P = 0.048) were independent poor prognostic factor in patients with mRCC. CONCLUSION AXL expression was associated with worse clinical outcome and may be an important prognostic biomarker in sunitinib-treated patients with metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Luís Eduardo Zucca
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Department of Medical Oncology, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | | | | | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Department of Pathology, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Marcos Alves de Lima
- Nucleous of Epidemiology and Statistics, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | | | | | - Flavio Mavignier Cárcano
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Department of Medical Oncology, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, Barretos, São Paulo, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
42
|
Targeting neuronal activity-regulated neuroligin-3 dependency in high-grade glioma. Nature 2017; 549:533-537. [PMID: 28959975 PMCID: PMC5891832 DOI: 10.1038/nature24014] [Citation(s) in RCA: 373] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 08/22/2017] [Indexed: 12/14/2022]
Abstract
High-grade gliomas (HGG) are a devastating group of cancers, representing the leading cause of brain tumor-related death in both children and adults. Therapies aimed at mechanisms intrinsic to the glioma cell have translated to only limited success; effective therapeutic strategies will need to also target elements of the tumor microenvironment that promote glioma progression. We recently demonstrated that neuronal activity robustly promotes the growth of a range of molecularly and clinically distinct HGG types, including adult glioblastoma (GBM), anaplastic oligodendroglioma, pediatric GBM, and diffuse intrinsic pontine glioma (DIPG)1. An important mechanism mediating this neural regulation of brain cancer is activity-dependent cleavage and secretion of the synaptic molecule neuroligin-3 (NLGN3), which promotes glioma proliferation through the PI3K-mTOR pathway1. However, neuroligin-3 necessity to glioma growth, proteolytic mechanism of secretion and further molecular consequences in glioma remain to be clarified. Here, we demonstrate a striking dependence of HGG growth on microenvironmental neuroligin-3, elucidate signaling cascades downstream of neuroligin-3 binding in glioma and determine a therapeutically targetable mechanism of secretion. Patient-derived orthotopic xenografts of pediatric GBM, DIPG and adult GBM fail to grow in Nlgn3 knockout mice. Neuroligin-3 stimulates numerous oncogenic pathways, including early focal adhesion kinase activation upstream of PI3K-mTOR, and induces transcriptional changes including upregulation of numerous synapse-related genes in glioma cells. Neuroligin-3 is cleaved from both neurons and oligodendrocyte precursor cells via the ADAM10 sheddase. ADAM10 inhibitors prevent release of neuroligin-3 into the tumor microenvironment and robustly block HGG xenograft growth. This work defines a promising strategy for targeting neuroligin-3 secretion, which could prove transformative for HGG therapy.
Collapse
|
43
|
Abstract
Primary brain tumors, particularly glioblastoma, are associated with significant morbidity and are often recalcitrant to standard therapies. In recent years, brain tumors have been the focus of large-scale genomic sequencing efforts, providing unprecedented insight into the genomic aberrations and cellular signaling mechanisms that drive these cancers. Discoveries from these efforts have translated into novel diagnostic algorithms, biomarkers, and therapeutic strategies in Neuro-Oncology. However, the cellular mechanisms that drive brain tumors are heterogeneous and complex: applying this new knowledge to improve patient outcomes remains a challenge. Efforts to characterize and target these molecular vulnerabilities are evolving.
Collapse
Affiliation(s)
- Rebecca A Harrison
- Department of Neuro-Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
44
|
Phase II study of MEDI-575, an anti-platelet-derived growth factor-α antibody, in patients with recurrent glioblastoma. J Neurooncol 2016; 131:185-191. [PMID: 27844311 DOI: 10.1007/s11060-016-2287-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/09/2016] [Indexed: 10/20/2022]
Abstract
MEDI-575, an immunoglobulin G2κ monoclonal antibody, selectively binds to platelet-derived growth factor-α receptor (PDGFR-α) with high specificity. This multicenter, single-arm, open-label, phase II study evaluated the efficacy and safety of MEDI-575 in patients with recurrent glioblastoma. Adults with first recurrence of glioblastoma following surgery, temozolomide, and radiation received MEDI-575 25 mg/kg intravenously over 60 min every 21 days until disease progression or unacceptable toxicity. Six-month progression-free survival rate (PFS-6) was the primary end point; secondary measures included response rate, overall survival (OS), and safety/tolerability. PDGFR-α expression was evaluated by immunohistochemistry. Fifty-six patients were enrolled; median age was 56.5 years (range 23-79), 66 % were male, and 66 % were aged ≥65 years. PFS-6 was 15.4 % [90 % confidence interval (CI) 8.1-24.9]. No complete or partial responses were observed; 23 (41.1 %) patients had stable disease as best response. Median PFS was 1.4 months (90 % CI 1.4, 1.8); median OS was 9.7 months (90 % CI 6.5, 11.8). The most common treatment-related adverse events (AEs) were diarrhea (16 %), nausea (13 %), and fatigue (13 %). Twelve (21 %) patients reported grade ≥3 AEs, with hydrocephalus (n = 3), dysphagia (n = 2), and convulsion (n = 2) reported in more than 1 patient. Two patients had treatment-related Grade ≥3 AEs of decreased lymphocyte count and asthenia (n = 1 each). Seven patients (13 %) discontinued MEDI-575 owing to AEs. Labeling of PDGFRα in glioblastoma cells and tumor-associated stromal cells was highly variable, with no correlation with PFS. MEDI-575, although well tolerated, had limited clinical activity in recurrent glioblastoma.
Collapse
|
45
|
Zhang RQ, Shi Z, Chen H, Chung NYF, Yin Z, Li KKW, Chan DTM, Poon WS, Wu J, Zhou L, Chan AKY, Mao Y, Ng HK. Biomarker-based prognostic stratification of young adult glioblastoma. Oncotarget 2016; 7:5030-41. [PMID: 26452024 PMCID: PMC4826263 DOI: 10.18632/oncotarget.5456] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/25/2015] [Indexed: 11/25/2022] Open
Abstract
While the predominant elderly and the pediatric glioblastomas have been extensively investigated, young adult glioblastomas were understudied. In this study, we sought to stratify young adult glioblastomas by BRAF, H3F3A and IDH1 mutations and examine the clinical relevance of the biomarkers. In 107 glioblastomas aged from 17 to 35 years, mutually exclusive BRAF-V600E (15%), H3F3A-K27M (15.9%), H3F3A-G34R/V (2.8%) and IDH1-R132H (16.8%) mutations were identified in over half of the cases. EGFR amplification and TERTp mutation were only detected in 3.7% and 8.4% in young adult glioblastomas, respectively. BRAF-V600E identified a clinically favorable subset of glioblastomas with younger age, frequent CDKN2A homozygous deletion, and was more amendable to surgical resection. H3F3A-K27M mutated glioblastomas were tightly associated with midline locations and showed dismal prognosis. IDH1-R132H was associated with older age and favorable outcome. Interestingly, tumors with positive PDGFRA immunohistochemical expression exhibited poorer prognosis and identified an aggressive subset of tumors among K27M mutated glioblastomas. Combining BRAF, H3F3A and IDH1 mutations allowed stratification of young adult glioblastomas into four prognostic subgroups. In summary, our study demonstrates the clinical values of stratifying young adult glioblastomas with BRAF, H3F3A and IDH1 mutations, which has important implications in refining prognostic classification of glioblastomas.
Collapse
Affiliation(s)
- Rui-Qi Zhang
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China.,Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Hong Chen
- Department of Neuropathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Nellie Yuk-Fei Chung
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China
| | - Zi Yin
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China
| | - Kay Ka-Wai Li
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China
| | - Danny Tat-Ming Chan
- Neurosurgery Division, Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Wai Sang Poon
- Neurosurgery Division, Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Aden Ka-Yin Chan
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ho-Keung Ng
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
46
|
Sharma VK, Singh A, Srivastava SK, Kumar V, Gardi NL, Nalwa A, Dinda AK, Chattopadhyay P, Yadav S. Increased expression of platelet-derived growth factor associated protein-1 is associated with PDGF-B mediated glioma progression. Int J Biochem Cell Biol 2016; 78:194-205. [PMID: 27448842 DOI: 10.1016/j.biocel.2016.07.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 07/16/2016] [Accepted: 07/18/2016] [Indexed: 12/30/2022]
Abstract
The current treatment therapies available for malignant gliomas are inadequate. There is an urgent need to develop more effective therapies by characterizing the molecular pathogenesis of the disease. Over expression of platelet-derived growth factor (PDGF) ligands and receptors have been reported in malignant gliomas. Platelet-derived growth factor associated protein-1 (PDAP-1) is reported to modulate the mitogenic activity of PDGF ligands, but to date, there is no information concerning its role in PDGF-mediated glioma cell proliferation. This study aimed to characterize the role of PDAP-1 in PDGF-mediated glioma proliferation. The expression of PDAP-1 was observed to be significantly increased (p<0.05) in grade IV glioma tissue and cell lines compared to grade III. siRNA-mediated knockdown of PDAP-1 reduced the expression of PDGF-B and its downstream genes (Akt1/Protein kinase B (PKB) and phosphoinositide-dependent kinase-1 (PDK1) by up to 50%. In PDAP-1 knockdown glioma cells, more than a twofold reduction was also observed in the level of phosphorylated Akt. Interestingly, knockdown of PDAP-1 in combination with PDGF-B antibody inhibited glioma cell proliferation through activation of Caspase 3/7 and 9. We also demonstrate that PDAP-1 co-localizes with PDGF-B in the cytoplasm of glioma cells, and an interaction between both of the proteins was established. Collectively, these findings suggest that the expression of PDAP-1 is associated with disease malignancy, and its inhibition reduced the proliferation of malignant glioma cells through down-regulation of PDGF-B/Akt/PDK1 signaling. Thus, this study establishes PDAP-1 as an effecter of PDGF signaling in glioma cells and suggests that it could also be a promising therapeutic target.
Collapse
Affiliation(s)
- Vinay Kumar Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Anand Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | - Vignesh Kumar
- Proteomics and Structural Biology Unit, Institute of genomics and Integrative Biology, New Delhi 110025, India
| | - Nilesh Laxman Gardi
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Mumbai, India
| | - Aasma Nalwa
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Amit Kumar Dinda
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | - Savita Yadav
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
47
|
Cárcano FM, Lengert AH, Vidal DO, Scapulatempo Neto C, Queiroz L, Marques H, Baltazar F, Berardinelli GN, Martinelli CMS, da Silva ECA, Reis RM, Lopes LF. Absence of microsatellite instability and BRAF (V600E) mutation in testicular germ cell tumors. Andrology 2016; 4:866-72. [PMID: 27153176 DOI: 10.1111/andr.12200] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 03/16/2016] [Accepted: 03/18/2016] [Indexed: 12/27/2022]
Abstract
Testicular germ cell tumors (TGCT) are the most common malignant neoplasm in young men. DNA mismatch repair deficiency can lead to microsatellite instability (MSI), an important mechanism of genetic instability. A mutation of the BRAF gene has been implicated in the pathogenesis of several solid tumors and has recently become an important therapeutic target. The role of MSI and BRAF gene mutation in TGCT, particularly in refractory disease, is poorly understood and reported findings are controversial. In this study, we aimed to determine the frequency and clinical impact of MSI status and BRAF mutations in TGCT. DNA was isolated from formalin-fixed paraffin embedded (FFPE) tissue from 150 TGCT cases. The MSI phenotype was evaluated using multiplex PCR for five quasimonomorphic mononucleotide repeat markers. Exon 15 of the BRAF oncogene (V600E) was analyzed by PCR, followed by direct sequencing. Sixteen percent of cases were considered to have refractory disease. In a small subset of cases (17 for MSI and 18 for BRAF), the quantity and quality of DNA recovery were poor and therefore, were unable to be analyzed. The remaining 133 TGCT cases showed a complete absence of MSI. Of the 132 cases successfully evaluated for BRAF mutations, all were V600E wild-type. In conclusion, despite a distinct response of testicular germ cell tumors to therapy, microsatellite instability, and the BRAF V600E mutation were absent in all testicular germ cell tumors tested in this study.
Collapse
Affiliation(s)
- F M Cárcano
- Department of Medical Oncology, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Health Sciences, Dr. Paulo Prata - FACISB, Barretos, Brazil
| | - A H Lengert
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos Children's Cancer Hospital, Barretos, Brazil
| | - D O Vidal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos Children's Cancer Hospital, Barretos, Brazil
| | - C Scapulatempo Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil
| | - L Queiroz
- Department of Medical Oncology, Hospital de Braga, Braga, Guimarães, Portugal
| | - H Marques
- Department of Medical Oncology, Hospital de Braga, Braga, Guimarães, Portugal
| | - F Baltazar
- Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - G N Berardinelli
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - C M S Martinelli
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - E C A da Silva
- Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil
| | - R M Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - L F Lopes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos Children's Cancer Hospital, Barretos, Brazil
| |
Collapse
|
48
|
Vazquez VDL, Vicente AL, Carloni A, Berardinelli G, Soares P, Scapulatempo C, Martinho O, Reis RM. Molecular profiling, including TERT promoter mutations, of acral lentiginous melanomas. Melanoma Res 2016; 26:93-9. [PMID: 26709572 DOI: 10.1097/cmr.0000000000000222] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Acral lentiginous melanoma (ALM) is the less common subtype with singular characterization. TERT (human telomerase reverse transcriptase) promoter mutations have being described as recurrent in melanomas and infrequent in ALM, but their real incidence and clinical relevance is unclear. The objectives of this study were to describe the prevalence of TERT promoter mutations in ALM, and correlate with the molecular profile of other drive genes and clinical features. Sixty-one samples from 48 patients with ALM were analyzed. After DNA isolation, the mutation profiles of the hotspot region of BRAF, NRAS, KIT, PDGFRA, and TERT genes were determined by PCR amplification followed by direct Sanger sequencing. KIT, PDGFRA, and VEGFR2 gene amplification was performed by quantitative PCR. Clinical information such as survival, clinical stage, and Breslow tumor classification were obtained from medical records. TERT promoter mutations were found in 9.3% of the cases, BRAF in 10.3%, NRAS in 7.5%, KIT in 20.7%, and PDGFRA in 14.8% of ALM. None of the cases showed KIT, PDGFRA, or VEGFR2 gene amplification. We found an association between KIT mutations and advanced Clark level (IV and V, P=0.043) and TERT promoter mutations with low mitotic index. No other significant associations were observed between mutation profile and patients' clinical features nor survival rates. Oncogenic TERT promoter mutations are present in a fraction of ALMs. No relevant associations were found between TERT mutation status and clinical/molecular features nor survival. Mutations of KIT and PDGFRA are the most common genetic alterations, and they can be therapeutic targets for these patients.
Collapse
Affiliation(s)
- Vinicius de Lima Vazquez
- aMolecular Oncology Research Center bDepartment of Surgery, Melanoma and Sarcoma Unity cDepartment of Pathology, Barretos Cancer Hospital, Barretos, São Paulo, Brazil dInstitute of Pathology and Molecular Immunology of University of Porto, (IPATIMUP), Porto eLife and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho fICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Purkait S, Mallick S, Sharma V, Kumar A, Pathak P, Jha P, Biswas A, Julka PK, Gupta D, Suri A, Upadhyay AD, Suri V, Sharma MC, Sarkar C. A simplified approach for molecular classification of glioblastomas (GBMs): experience from a tertiary care center in India. Brain Tumor Pathol 2016; 33:183-90. [PMID: 26865311 DOI: 10.1007/s10014-016-0251-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/25/2016] [Indexed: 01/04/2023]
Abstract
This study aims to establish a simplified molecular classification of glioblastomas (GBMs) based on molecular genetic alterations. GBM cases (n-114) were evaluated for IDH-1 and TP53 mutation by Sanger sequencing, PDGFRA and EGFR amplification by FISH, NF1 and YKL40 expression by qRT-PCR. Subsequently they were classified into four subgroups: classical like (CL), proneural like (PN), mesenchymal like (MES) and neural like (NEU). CL subtype was most frequent (39 %), followed by PN (32 %) and MES (20 %) subtypes. PN subtype had significantly younger age at presentation and longest survival (median PFS-82.5 weeks; 1 and 2 years OS-90.6 and 71.3 %). Other three subgroups had equally poor prognosis and hence, clubbed together as non-proneural (Non-PN) (median PFS-39 weeks; 1 and 2 years OS-66 and 0 %). Hence, we recommended this relatively easy method of subclassifying GBMs into PN and Non-PN which are statistically different in prognosis (both OS and PFS on uni and multivariate analysis). Although evaluation of six molecular alterations for identifying these two subgroups is still cumbersome, we propose segregation of PN subtype alone based on assessment of IDH1, TP53 and PDGFRA status, which is relatively easy and may be amenable to routine practice.
Collapse
Affiliation(s)
- Suvendu Purkait
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Supriya Mallick
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Vikas Sharma
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Anupam Kumar
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Pankaj Pathak
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Prerana Jha
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Ahitagni Biswas
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Pramod Kumar Julka
- Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Deepak Gupta
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Suri
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Datt Upadhyay
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Mehar C Sharma
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India.
| |
Collapse
|
50
|
Cárcano FM, Vidal DO, van Helvoort Lengert A, Neto CS, Queiroz L, Marques H, Baltazar F, da Silva Martinelli CM, Soares P, da Silva ECA, Lopes LF, Reis RM. Hotspot TERT promoter mutations are rare events in testicular germ cell tumors. Tumour Biol 2015; 37:4901-7. [PMID: 26526580 DOI: 10.1007/s13277-015-4317-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/22/2015] [Indexed: 12/25/2022] Open
Abstract
The abnormal activation of telomerase, codified by the telomerase reverse transcriptase (TERT) gene, is related to one of cancer hallmarks. Hotspot somatic mutations in the promoter region of TERT, specifically the c.-124:C>T and c.-146:C>T, were recently identified in a range of human cancers and have been associated with a more aggressive behavior. Testicular germ cell tumors frequently exhibit a good prognosis; however, the development of refractory disease is still a clinical challenge. In this study, we aim to evaluate for the first time the presence of the hotspot telomerase reverse transcriptase gene promoter mutations in testicular germ cell tumors. A series of 150 testicular germ cell tumor cases and four germ cell tumor cell lines were evaluated by PCR followed by direct Sanger sequencing and correlated with patient's clinical pathological features. Additionally, we genotyped the telomerase reverse transcriptase gene promoter single nucleotide polymorphism rs2853669 (T>C) located at -245 position. We observed the presence of the TERT promoter mutation in four patients, one exhibited the c.-124:C>T and three the c.-146:C>T. No association between TERT mutation status and clinicopathological features could be identified. The analysis of the rs2853669 showed that variant C was present in 22.8 % of the cases. In conclusion, we showed for the first time that TERT promoter mutations occur in a small subset (~3 %) of testicular germ cell tumors.
Collapse
Affiliation(s)
- Flavio Mavignier Cárcano
- Department of Clinical Oncology, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, Barretos, Brazil
| | - Daniel Onofre Vidal
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Rua Antenor Duarte Villela St, CEP 14784 400, Barretos, Sao Paulo, Brazil.,Barretos Children's Cancer Hospital, 3025, Avenida João Baroni, Barretos, Brazil
| | - André van Helvoort Lengert
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Rua Antenor Duarte Villela St, CEP 14784 400, Barretos, Sao Paulo, Brazil.,Barretos Children's Cancer Hospital, 3025, Avenida João Baroni, Barretos, Brazil
| | - Cristovam Scapulatempo Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Rua Antenor Duarte Villela St, CEP 14784 400, Barretos, Sao Paulo, Brazil.,Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil
| | - Luisa Queiroz
- Department of Medical Oncology, Hospital de Braga, Braga, Portugal
| | | | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Camila Maria da Silva Martinelli
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Rua Antenor Duarte Villela St, CEP 14784 400, Barretos, Sao Paulo, Brazil
| | - Paula Soares
- Institute of Molecular Pathology and Immunology of the University of Porto-IPATIMUP, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Medical Faculty, University of Porto, 4200-319, Porto, Portugal
| | | | - Luiz Fernando Lopes
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Rua Antenor Duarte Villela St, CEP 14784 400, Barretos, Sao Paulo, Brazil. .,Barretos Children's Cancer Hospital, 3025, Avenida João Baroni, Barretos, Brazil.
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Rua Antenor Duarte Villela St, CEP 14784 400, Barretos, Sao Paulo, Brazil. .,Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|