51
|
Burgy O, Crestani B, Bonniaud P. Targeting the nasty nestin to shoot lung fibrosis. Eur Respir J 2022; 59:59/5/2103146. [PMID: 35512809 DOI: 10.1183/13993003.03146-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/05/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Olivier Burgy
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France .,Constitutive Reference Center for Rare Pulmonary Diseases - OrphaLung, Dijon-Bourgogne University Hospital, Dijon, France
| | - Bruno Crestani
- Université Paris Cité, Inserm, U1152, laboratoire d'excellence INFLAMEX, Paris, France.,APHP, Service de Pneumologie A, Constitutive Reference Center for Rare Pulmonary Diseases - OrphaLung, FHU APOLLO, Hôpital Bichat, Paris, France
| | - Philippe Bonniaud
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France.,Constitutive Reference Center for Rare Pulmonary Diseases - OrphaLung, Dijon-Bourgogne University Hospital, Dijon, France.,Dept of Pulmonary Medicine and Intensive Care Unit, Dijon-Bourgogne University Hospital, Dijon, France
| |
Collapse
|
52
|
Sementino E, Kadariya Y, Cheung M, Menges CW, Tan Y, Kukuyan AM, Shrestha U, Karchugina S, Cai KQ, Peri S, Duncan JS, Chernoff J, Testa JR. Inactivation of p21-Activated Kinase 2 (Pak2) Inhibits the Development of Nf2-Deficient Tumors by Restricting Downstream Hedgehog and Wnt Signaling. Mol Cancer Res 2022; 20:699-711. [PMID: 35082167 PMCID: PMC9081258 DOI: 10.1158/1541-7786.mcr-21-0837] [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] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/02/2021] [Accepted: 01/13/2022] [Indexed: 11/16/2022]
Abstract
Because loss of the NF2 tumor suppressor gene results in p21-activated kinase (Pak) activation, PAK inhibitors hold promise for the treatment of NF2-deficient tumors. To test this possibility, we asked if loss of Pak2, a highly expressed group I PAK member, affects the development of malignant mesothelioma in Nf2;Cdkn2a-deficient (NC) mice and the growth properties of NC mesothelioma cells in culture. In vivo, deletion of Pak2 resulted in a markedly decreased incidence and delayed onset of both pleural and peritoneal malignant mesotheliomas in NC mice. In vitro, Pak2 deletion decreased malignant mesothelioma cell viability, migration, clonogenicity, and spheroid formation. RNA-sequencing analysis demonstrated downregulated expression of Hedgehog and Wnt pathway genes in NC;Pak2-/- mesothelioma cells versus NC;Pak2+/+ mesothelioma cells. Targeting of the Hedgehog signaling component Gli1 or its target gene Myc inhibited cell viability and spheroid formation in NC;P+/+ mesothelioma cells. Kinome profiling uncovered kinase changes indicative of EMT in NC;Pak2-/- mesothelioma cells, suggesting that Pak2-deficient malignant mesotheliomas can adapt by reprogramming their kinome in the absence of Pak activity. The identification of such compensatory pathways offers opportunities for rational combination therapies to circumvent resistance to anti-PAK drugs. IMPLICATIONS We provide evidence supporting a role for PAK inhibitors in treating NF2-deficient tumors. NF2-deficient tumors lacking Pak2 eventually adapt by kinome reprogramming, presenting opportunities for combination therapies to bypass anti-PAK drug resistance.
Collapse
Affiliation(s)
- Eleonora Sementino
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yuwaraj Kadariya
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mitchell Cheung
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Craig W. Menges
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yinfei Tan
- Genomics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Anna-Mariya Kukuyan
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Ujjawal Shrestha
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Sofiia Karchugina
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Kathy Q. Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Suraj Peri
- Bioinformatics and Biostatistics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - James S. Duncan
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Jonathan Chernoff
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Joseph R. Testa
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Joseph R. Testa, Ph.D., Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 1911; Phone: (215) 728-2610; Fax: (215) 214-1619;
| |
Collapse
|
53
|
Marayati R, Stafman LL, Williams AP, Bownes LV, Quinn CH, Markert HR, Easlick JL, Stewart JE, Crossman DK, Mroczek-Musulman E, Beierle EA. CRISPR/Cas9-mediated knockout of PIM3 suppresses tumorigenesis and cancer cell stemness in human hepatoblastoma cells. Cancer Gene Ther 2022; 29:558-572. [PMID: 33864024 PMCID: PMC8521561 DOI: 10.1038/s41417-021-00334-4] [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: 11/14/2020] [Revised: 02/23/2021] [Accepted: 03/26/2021] [Indexed: 02/02/2023]
Abstract
Hepatoblastoma remains one of the most difficult childhood tumors to treat and is alarmingly understudied. We previously demonstrated that Proviral Insertion site in Maloney murine leukemia virus (PIM) kinases, specifically PIM3, are overexpressed in human hepatoblastoma cells and function to promote tumorigenesis. We aimed to use CRISPR/Cas9 gene editing with dual gRNAs to introduce large inactivating deletions in the PIM3 gene and achieve stable PIM3 knockout in the human hepatoblastoma cell line, HuH6. PIM3 knockout of hepatoblastoma cells led to significantly decreased proliferation, viability, and motility, inhibited cell-cycle progression, decreased tumor growth in a xenograft murine model, and increased animal survival. Analysis of RNA sequencing data revealed that PIM3 knockout downregulated expression of pro-migratory and pro-invasive genes and upregulated expression of genes involved in apoptosis and differentiation. Furthermore, PIM3 knockout decreased hepatoblastoma cancer cell stemness as evidenced by decreased tumorsphere formation, decreased mRNA abundance of stemness markers, and decreased cell surface expression of CD133, a marker of hepatoblastoma stem cell-like cancer cells. Reintroduction of PIM3 into PIM3 knockout cells rescued the malignant phenotype. Successful CRISPR/Cas9 knockout of PIM3 kinase in human hepatoblastoma cells confirmed the role of PIM3 in promoting hepatoblastoma tumorigenesis and cancer cell stemness.
Collapse
Affiliation(s)
- Raoud Marayati
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Laura L. Stafman
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Adele P. Williams
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Laura V. Bownes
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Colin H. Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Hooper R. Markert
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Juliet L. Easlick
- Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jerry E. Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - David K. Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | | | - Elizabeth A. Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
54
|
Helweg LP, Windmöller BA, Burghardt L, Storm J, Förster C, Wethkamp N, Wilkens L, Kaltschmidt B, Banz-Jansen C, Kaltschmidt C. The Diminishment of Novel Endometrial Carcinoma-Derived Stem-like Cells by Targeting Mitochondrial Bioenergetics and MYC. Int J Mol Sci 2022; 23:ijms23052426. [PMID: 35269569 PMCID: PMC8910063 DOI: 10.3390/ijms23052426] [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: 01/31/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer stem cells (CSCs) are a small subpopulation of tumor cells harboring properties that include self-renewal, multi-lineage differentiation, tumor reconstitution, drug resistance and invasiveness, making them key players in tumor relapse. In the present paper, we develop new CSC models and analyze the molecular pathways involved in survival to identify targets for the establishment of novel therapies. Endometrial carcinoma-derived stem-like cells (ECSCs) were isolated from carcinogenic gynecological tissue and analyzed regarding their expression of prominent CSC markers. Further, they were treated with the MYC-signaling inhibitor KJ-Pyr-9, chemotherapeutic agent carboplatin and type II diabetes medication metformin. ECSC populations express common CSC markers, such as Prominin-1 and CD44 antigen as well as epithelial-to-mesenchymal transition markers, Twist, Snail and Slug, and exhibit the ability to form free-floating spheres. The inhibition of MYC signaling and treatment with carboplatin as well as metformin significantly reduced the cell survival of ECSC-like cells. Further, treatment with metformin significantly decreased the mitochondrial membrane potential of ECSC-like cells, while the extracellular lactate concentration was increased. The established ECSC-like populations represent promising in vitro models to further study the contribution of ECSCs to endometrial carcinogenesis. Targeting MYC signaling as well as mitochondrial bioenergetics has shown promising results in the diminishment of ECSCs, although molecular signaling pathways need further investigations.
Collapse
Affiliation(s)
- Laureen P. Helweg
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Correspondence: ; Tel.: +49-0521-106-5619
| | - Beatrice A. Windmöller
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
| | - Leonie Burghardt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
| | - Jonathan Storm
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
| | - Christine Förster
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Institute of Pathology, KRH Hospital Nordstadt, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany;
| | - Nils Wethkamp
- Institute of Pathology, KRH Hospital Nordstadt, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany;
| | - Ludwig Wilkens
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Institute of Pathology, KRH Hospital Nordstadt, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany;
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Constanze Banz-Jansen
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Department of Gynecology and Obstetrics, and Perinatal Center, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, 33615 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
| |
Collapse
|
55
|
Farrokhfar S, Tiraihi T, Movahedin M, Azizi H. Morphine Induces Differential Gene Expression in Transdifferentiated Neuron-Like Cells from Adipose-Derived Stem Cells. BIOL BULL+ 2022. [DOI: 10.1134/s1062359022130052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
56
|
Kaltschmidt B, Witte KE, Greiner JFW, Weissinger F, Kaltschmidt C. Targeting NF-κB Signaling in Cancer Stem Cells: A Narrative Review. Biomedicines 2022; 10:biomedicines10020261. [PMID: 35203471 PMCID: PMC8869483 DOI: 10.3390/biomedicines10020261] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 12/14/2022] Open
Abstract
Among the cell populations existing within a tumor, cancer stem cells are responsible for metastasis formation and chemotherapeutic resistance. In the present review, we focus on the transcription factor NF-κB, which is present in every cell type including cancer stem cells. NF-κB is involved in pro-tumor inflammation by its target gene interleukin 1 (IL1) and can be activated by a feed-forward loop in an IL1-dependent manner. Here, we summarize current strategies targeting NF-κB by chemicals and biologicals within an integrated cancer therapy. Specifically, we start with a tyrosine kinase inhibitor targeting epidermal growth factor (EGF)-receptor-mediated phosphorylation. Furthermore, we summarize current strategies of multiple myeloma treatment involving lenalidomide, bortezomib, and dexamethasone as potential NF-κB inhibitors. Finally, we discuss programmed death-ligand 1 (PD-L1) as an NF-κB target gene and its role in checkpoint therapy. We conclude, that NF-κB inhibition by specific inhibitors of IκB kinase was of no clinical use but inhibition of upstream and downstream targets with drugs or biologicals might be a fruitful way to treat cancer stem cells.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany;
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
| | - Kaya E. Witte
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Johannes F. W. Greiner
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| | - Florian Weissinger
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
- Department of Hematology, Oncology, Internal Medicine, Bone Marrow and Stem Cell Transplantation, Palliative Medicine, and Tumor Center, Protestant Hospital of Bethel Foundation, University Hospital OWL of Bielefeld University, Schildescher Str. 99, 33611 Bielefeld, Germany
| | - Christian Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld FBMB e.V., Maraweg 21, 33617 Bielefeld, Germany; (K.E.W.); (J.F.W.G.); (F.W.)
- Department of Cell Biology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
- Correspondence: ; Tel.: +49-521-106-5625
| |
Collapse
|
57
|
Yamamoto Y, Hayashi Y, Sakaki H, Murakami I. Evaluation of Clinical and Immunohistochemical Factors Relating to Melanoma Metastasis: Potential Roles of Nestin and Fascin in Melanoma. Diagnostics (Basel) 2022; 12:diagnostics12010219. [PMID: 35054386 PMCID: PMC8774891 DOI: 10.3390/diagnostics12010219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/01/2023] Open
Abstract
For melanoma treatment, an early diagnosis and a complete resection of the primary tumor is essential. In addition, detection of factors that may be related to metastasis is indispensable. A total of 30 Japanese patients with Stage I or II melanoma, diagnosed according to the classification of the American Joint Committee on Cancer, are included in this study. Clinical background (sex, onset age, primary tumor area, existence of remaining cancer cells at the resected tissue margin, and treatment after the primary surgery) and immunohistochemical staining (Nestin and Fascin) on the resected tissue were examined to detect factors statistically related to metastasis. The analysis result has shown that older onset age and positive immunohistochemical expressions of Nestin and Fascin are statistically related to metastasis. To facilitate meticulous observation of Nestin and Fascin expression at different timing (e.g., onset and metastasis), double immunofluorescence staining was performed. Nestin is a class VI intermediate filament protein, initially detected in neural stem cells. Fascin is an actin-bundling protein which regulates cell adhesion, migration and invasion. Nestin and Fascin are suggested to relate to melanoma metastasis, however, the potential role of Fascin is controversial. Analysis of variations in Fascin expression detected in this study may contribute to further investigations concerning potential roles of Fascin for progression of melanoma. This is the first study to report double immunofluorescent staining of Nestin and Fascin in melanoma. Nestin and Fascin double-positive melanoma cells were detected.
Collapse
Affiliation(s)
- Yumiko Yamamoto
- Department of Diagnostic Pathology, Kochi University Hospital, Kochi University, 185-1, Kohasu, Oko-cho, Nankoku 783-8505, Japan;
- Correspondence: ; Tel.: +81-88-880-2330
| | - Yoshihiro Hayashi
- Equipment of Support Planning Office, Kochi University, 185-1, Kohasu, Oko-cho, Nankoku 783-8505, Japan;
- Department of Pathology, School of Medicine, Kochi University, 185-1, Kohasu, Oko-cho, Nankoku 783-8505, Japan
| | - Hideyuki Sakaki
- Department of Nutritional Sciences for Well-Being Health, Kansai University of Welfare Sciences, 3-11-1, Asahigaoka, Kahsihara 582-0026, Japan;
| | - Ichiro Murakami
- Department of Diagnostic Pathology, Kochi University Hospital, Kochi University, 185-1, Kohasu, Oko-cho, Nankoku 783-8505, Japan;
- Department of Pathology, School of Medicine, Kochi University, 185-1, Kohasu, Oko-cho, Nankoku 783-8505, Japan
| |
Collapse
|
58
|
Rufino-Ramos D, Lule S, Mahjoum S, Ughetto S, Cristopher Bragg D, Pereira de Almeida L, Breakefield XO, Breyne K. Using genetically modified extracellular vesicles as a non-invasive strategy to evaluate brain-specific cargo. Biomaterials 2022; 281:121366. [PMID: 35033904 PMCID: PMC8886823 DOI: 10.1016/j.biomaterials.2022.121366] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/25/2021] [Accepted: 01/04/2022] [Indexed: 12/18/2022]
Abstract
The lack of techniques to trace brain cell behavior in vivo hampers the ability to monitor status of cells in a living brain. Extracellular vesicles (EVs), nanosized membrane-surrounded vesicles, released by virtually all brain cells might be able to report their status in easily accessible biofluids, such as blood. EVs communicate among tissues using lipids, saccharides, proteins, and nucleic acid cargo that reflect the state and composition of their source cells. Currently, identifying the origin of brain-derived EVs has been challenging, as they consist of a rare population diluted in an overwhelming number of blood and peripheral tissue-derived EVs. Here, we developed a sensitive platform to select out pre-labelled brain-derived EVs in blood as a platform to study the molecular fingerprints of brain cells. This proof-of-principle study used a transducible construct tagging tetraspanin (TSN) CD63, a membrane-spanning hallmark of EVs equipped with affinity, bioluminescent, and fluorescent tags to increase detection sensitivity and robustness in capture of EVs secreted from pre-labelled cells into biofluids. Our platform enables unprecedented efficient isolation of neural EVs from the blood. These EVs derived from pre-labelled mouse brain cells or engrafted human neuronal progenitor cells (hNPCs) were submitted to multiplex analyses, including transcript and protein levels, in compliance with the multibiomolecule EV carriers. Overall, our novel strategy to track brain-derived EVs in a complex biofluid opens up new avenues to study EVs released from pre-labelled cells in near and distal compartments into the biofluid source.
Collapse
Affiliation(s)
- David Rufino-Ramos
- Neurology and Radiology Department, Massachusetts General Hospital, Harvard Medical School, 13(th)Street, Building 149, Charlestown, MA, 02129, USA; CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra, 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal; CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Sevda Lule
- Neurology and Radiology Department, Massachusetts General Hospital, Harvard Medical School, 13(th)Street, Building 149, Charlestown, MA, 02129, USA
| | - Shadi Mahjoum
- Neurology and Radiology Department, Massachusetts General Hospital, Harvard Medical School, 13(th)Street, Building 149, Charlestown, MA, 02129, USA
| | - Stefano Ughetto
- Neurology and Radiology Department, Massachusetts General Hospital, Harvard Medical School, 13(th)Street, Building 149, Charlestown, MA, 02129, USA
| | - D Cristopher Bragg
- Neurology and Radiology Department, Massachusetts General Hospital, Harvard Medical School, 13(th)Street, Building 149, Charlestown, MA, 02129, USA; The Collaborative Center for X-linked Dystonia-Parkinsonism, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - Luís Pereira de Almeida
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Coimbra, 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal; CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Xandra O Breakefield
- Neurology and Radiology Department, Massachusetts General Hospital, Harvard Medical School, 13(th)Street, Building 149, Charlestown, MA, 02129, USA
| | - Koen Breyne
- Neurology and Radiology Department, Massachusetts General Hospital, Harvard Medical School, 13(th)Street, Building 149, Charlestown, MA, 02129, USA.
| |
Collapse
|
59
|
Ping M, Wang S, Guo Y, Jia J. TRIM21 improves apatinib treatment in gastric cancer through suppressing EZH1 stability. Biochem Biophys Res Commun 2022; 586:177-184. [PMID: 34856418 DOI: 10.1016/j.bbrc.2021.07.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/05/2021] [Accepted: 07/11/2021] [Indexed: 12/26/2022]
Abstract
Gastric cancer (GC) is a common tumor with high metastatic rate worldwide. Promoting chemosensitivity is effective for improving therapeutic outcome and survival rate for GC patients. Tripartite motif-containing 21 (TRIM21), a member of TRIM-containing proteins, plays crucial roles in regulating numerous cellular events involved in tumor progression. However, it's regulatory effects on GC growth and drug sensitivity are still unclear. In the present study, we identified that TRIM21 expression was remarkably decreased in human GC tissues compared with the adjacent normal ones, and its down-regulation was closely linked to higher recurrence and lower overall survival rate among GC patients. We then found that apatinib (APA)-reduced GC cell proliferation was significantly abolished by TRIM21 knockdown; however, promoting TRIM21 expression further improved the sensitivity of GC cells to APA treatment, as proved by the remarkably decreased cell viability and colony formation. Furthermore, TRIM21 over-expression dramatically enhanced apoptosis, while its knockdown markedly diminished apoptotic cell death in APA-incubated GC cells. Moreover, stem cell properties of GC cells were also restrained by TRIM21. Our in vivo experiments showed that APA-repressed tumor growth was considerably abolished by TRIM21 knockdown, whereas being further elevated by TRIM21 over-expression. In addition, we showed that TRIM21 markedly decreased enhancer of zeste homolog 1 (EZH1) protein expression levels in GC cells, and importantly, a direct interaction between TRIM21 and EZH1 was verified. Of note, our in vitro studies revealed that EZH1 over-expression remarkably abolished the function of TRIM21 to restrain cell viability and induce apoptosis in APA-incubated GC cells, indicating that EZH1 suppression was necessary for TRIM21 to inhibit GC progression. Together, our findings demonstrated that TRIM21 may be a novel therapeutic target for GC treatment through reducing EZH1 to improve chemosensitivity.
Collapse
Affiliation(s)
- Mei Ping
- Department of Oncology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China
| | - Shumin Wang
- Department of Oncology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China
| | - Yarong Guo
- Department of Oncology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China
| | - Junmei Jia
- Department of Oncology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China.
| |
Collapse
|
60
|
Prognostic Biomarkers in Uveal Melanoma: The Status Quo, Recent Advances and Future Directions. Cancers (Basel) 2021; 14:cancers14010096. [PMID: 35008260 PMCID: PMC8749988 DOI: 10.3390/cancers14010096] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Although rare, uveal melanoma (UM) is the most common cancer that develops inside adult eyes. The prognosis is poor, since 50% of patients will develop lethal metastases in the first decade, especially to the liver. Once metastases are detected, life expectancy is limited, given that the available treatments are mostly unsuccessful. Thus, there is a need to find methods that can accurately predict UM prognosis and also effective therapeutic strategies to treat this cancer. In this manuscript, we initially compile the current knowledge on epidemiological, clinical, pathological and molecular features of UM. Then, we cover the most relevant prognostic factors currently used for the evaluation and follow-up of UM patients. Afterwards, we highlight emerging molecular markers in UM published over the last three years. Finally, we discuss the problems preventing meaningful advances in the treatment and prognostication of UM patients, as well as forecast new roadblocks and paths of UM-related research. Abstract Uveal melanoma (UM) is the most common malignant intraocular tumour in the adult population. It is a rare cancer with an incidence of nearly five cases per million inhabitants per year, which develops from the uncontrolled proliferation of melanocytes in the choroid (≈90%), ciliary body (≈6%) or iris (≈4%). Patients initially present either with symptoms like blurred vision or photopsia, or without symptoms, with the tumour being detected in routine eye exams. Over the course of the disease, metastases, which are initially dormant, develop in nearly 50% of patients, preferentially in the liver. Despite decades of intensive research, the only approach proven to mildly control disease spread are early treatments directed to ablate liver metastases, such as surgical excision or chemoembolization. However, most patients have a limited life expectancy once metastases are detected, since there are limited therapeutic approaches for the metastatic disease, including immunotherapy, which unlike in cutaneous melanoma, has been mostly ineffective for UM patients. Therefore, in order to offer the best care possible to these patients, there is an urgent need to find robust models that can accurately predict the prognosis of UM, as well as therapeutic strategies that effectively block and/or limit the spread of the metastatic disease. Here, we initially summarized the current knowledge about UM by compiling the most relevant epidemiological, clinical, pathological and molecular data. Then, we revisited the most important prognostic factors currently used for the evaluation and follow-up of primary UM cases. Afterwards, we addressed emerging prognostic biomarkers in UM, by comprehensively reviewing gene signatures, immunohistochemistry-based markers and proteomic markers resulting from research studies conducted over the past three years. Finally, we discussed the current hurdles in the field and anticipated the future challenges and novel avenues of research in UM.
Collapse
|
61
|
Nestin-Expressing Cells in the Lung: The Bad and the Good Parts. Cells 2021; 10:cells10123413. [PMID: 34943921 PMCID: PMC8700449 DOI: 10.3390/cells10123413] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 12/27/2022] Open
Abstract
Nestin is a member of the intermediate filament family, which is expressed in a variety of stem or progenitor cells as well as in several types of malignancies. Nestin might be involved in tissue homeostasis or repair, but its expression has also been associated with processes that lead to a poor prognosis in various types of cancer. In this article, we review the literature related to the effect of nestin expression in the lung. According to most of the reports in the literature, nestin expression in lung cancer leads to an aggressive phenotype and resistance to chemotherapy as well as radiation treatments due to the upregulation of phenomena such as cell proliferation, angiogenesis, and metastasis. Furthermore, nestin may be involved in the pathogenesis of some non-cancer-related lung diseases. On the other hand, evidence also indicates that nestin-positive cells may have a role in lung homeostasis and be capable of generating various types of lung tissues. More research is necessary to establish the true value of nestin expression as a prognostic factor and therapeutic target in lung cancer in addition to its usefulness in therapeutic approaches for pulmonary diseases.
Collapse
|
62
|
Liao G, Liao Y, Li D, Fu Z, Wu S, Cheng D, Ouyang Q, Tang Z, Zeng G, Liang X, Xu S, Hu J, Liu M. Human Platelet Lysate Maintains Stemness of Umbilical Cord-Derived Mesenchymal Stromal Cells and Promote Lung Repair in Rat Bronchopulmonary Dysplasia. Front Cell Dev Biol 2021; 9:722953. [PMID: 34858970 PMCID: PMC8631747 DOI: 10.3389/fcell.2021.722953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/05/2021] [Indexed: 11/23/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) show potential for treating preclinical models of newborn bronchopulmonary dysplasia (BPD), but studies of their therapeutic effectiveness have had mixed results, in part due to the use of different media supplements for MSCs expansion in vitro. The current study sought to identify an optimal culture supplement of umbilical cord-derived MSCs (UC-MSCs) for BPD therapy. In this study, we found that UC-MSCs cultured with human platelet lysate (hPL-UCMSCs) were maintained a small size from Passage 1 (P1) to P10, while UC-MSCs cultured with fetal bovine serum (FBS-UCMSCs) became wide and flat. Furthermore, hPL was associated with lower levels of senescence in UC-MSCs during in vitro expansion compared with FBS, as indicated by the results of β-galactosidase staining and measures of senescence-related genes (CDKN2A, CDKN1A, and mTOR). In addition, hPL enhanced the proliferation and cell viability of the UC-MSCs and reduced their doubling time in vitro. Compared with FBS-UCMSCs, hPL-UCMSCs have a greater potential to differentiate into osteocytes and chondrocytes. Moreover, using hPL resulted in greater expression of Nestin and specific paracrine factors (VEGF, TGF-β1, FGF2, IL-8, and IL-6) in UC-MSCs compared to using FBS. Critically, we also found that hPL-UCMSCs are more effective than FBS-UCMSCs for the treatment of BPD in a rat model, with hPL leading to improvements in survival rate, lung architecture and fibrosis, and lung capillary density. Finally, qPCR of rat lung mRNA demonstrated that hPL-UCMSCs had lower expression levels of inflammatory factors (TNF-α and IL-1β) and a key chemokine (MCP-1) at postnatal day 10, and there was significant reduction of CD68+ macrophages in lung tissue after hPL-UCMSCs transplantation. Altogether, our findings suggest that hPL is an optimal culture supplement for UC-MSCs expansion in vitro, and that hPL-UCMSCs promote lung repair in rat BPD disease.
Collapse
Affiliation(s)
- Guilian Liao
- Obstetrics and Gynecology, Maternal and Child Health Hospital of Longgang District, Shenzhen, China
| | - Yan Liao
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Duanduan Li
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Zeqin Fu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Shiduo Wu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Danling Cheng
- Obstetrics and Gynecology, Maternal and Child Health Hospital of Longgang District, Shenzhen, China
| | - Qiuxing Ouyang
- Neurological Rehabilitation for Children, Maternal and Child Health Hospital of Luohu District, Shenzhen, China
| | - Zan Tang
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Guifang Zeng
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Xiao Liang
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Shaokun Xu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Junyuan Hu
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Muyun Liu
- National-Local Associated Engineering Laboratory for Personalized Cell Therapy, Shenzhen, China
| |
Collapse
|
63
|
Guido CB, Sosa LDV, Perez PA, Zlocoswki N, Velazquez FN, Gutierrez S, Petiti JP, Mukdsi JH, Torres AI. Changes of stem cell niche during experimental pituitary tumor development. J Neuroendocrinol 2021; 33:e13051. [PMID: 34708474 DOI: 10.1111/jne.13051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 09/14/2021] [Accepted: 10/07/2021] [Indexed: 12/20/2022]
Abstract
To investigate the putative stem cell/tumor stem cell (SC/TSC) niche contribution to hyperplasic/adenomatous pituitary lesions, we analyzed variation in the pituitary stem cell population during the development of experimental pituitary tumors. Pituitary tumors were induced in female F344 rats with estradiol benzoate for 5, 10, 20 and 30 days. Cells positive for GFRa2, Sox2, Sox9, Nestin, CD133 and CD44 were identified in the marginal zone and in the adenoparenchyma in both control and 30D groups, with predominant adenoparenchyma localization of GRFa2 and SOX9 found in tumoral pituitaries. GFRa2, Nestin, CD133 and CD44 were upregulated at the initial stages of tumor growth, whereas Sox9 significantly decreased at 5D, with Sox2 remaining invariable during the hyperplasic/adenomatous development. In addition, isolated pituispheres from normal and tumoral pituitary glands enriched in SC/TSC were characterized. Pituispheres from the 30D glands were positive for the above-mentioned markers and showed a significant increase in the proliferation. In conclusion, our data revealed pituitary SC pool fluctuations during hyperplastic/adenomatous development, with differential localization of the SC/TSC niche in this process. These findings may help to provide a better understanding of these cell populations, which is crucial for achieving advancements in the field of pituitary tumor biology.
Collapse
Affiliation(s)
- Carolina Beatriz Guido
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud, Córdoba, Argentina
| | - Liliana Del Valle Sosa
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud, Córdoba, Argentina
| | - Pablo Aníbal Perez
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud, Córdoba, Argentina
| | - Natacha Zlocoswki
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud, Córdoba, Argentina
| | - Fabiola Noelia Velazquez
- CIQUIBIC-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Silvina Gutierrez
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud, Córdoba, Argentina
| | - Juan Pablo Petiti
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud, Córdoba, Argentina
| | - Jorge Humberto Mukdsi
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud, Córdoba, Argentina
| | - Alicia Inés Torres
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud, Córdoba, Argentina
| |
Collapse
|
64
|
Basu M, Philipp LM, Baines JF, Sebens S. The Microbiome Tumor Axis: How the Microbiome Could Contribute to Clonal Heterogeneity and Disease Outcome in Pancreatic Cancer. Front Oncol 2021; 11:740606. [PMID: 34631577 PMCID: PMC8495218 DOI: 10.3389/fonc.2021.740606] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant cancers. It is characterized by a poor prognosis with a 5-year survival rate of only around 10% and an ongoing increase in death rate. Due to the lack of early and specific symptoms, most patients are diagnosed at an advanced or even metastasized stage, essentially limiting curative treatment options. However, even curative resection of the primary tumor and adjuvant therapy often fails to provide a long-term survival benefit. One reason for this dismal situation can be seen in the evolution of therapy resistances. Furthermore, PDAC is characterized by high intratumor heterogeneity, pointing towards an abundance of cancer stem cells (CSCs), which are regarded as essential for tumor initiation and drug resistance. Additionally, it was shown that the gut microbiome is altered in PDAC patients, promotes Epithelial-Mesenchymal-Transition (EMT), determines responses towards chemotherapy, and affects survival in PDAC patients. Given the established links between CSCs and EMT as well as drug resistance, and the emerging role of the microbiome in PDAC, we postulate that the composition of the microbiome of PDAC patients is a critical determinant for the abundance and plasticity of CSC populations and thus tumor heterogeneity in PDAC. Unravelling this complex interplay might pave the way for novel treatment strategies.
Collapse
Affiliation(s)
- Meghna Basu
- Max Planck Institute for Evolutionary Biology, Plön, Germany.,Section of Evolutionary Medicine, Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Lisa-Marie Philipp
- Institute for Experimental Cancer Research, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel University, Kiel, Germany
| | - John F Baines
- Max Planck Institute for Evolutionary Biology, Plön, Germany.,Section of Evolutionary Medicine, Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Kiel University, Kiel, Germany
| |
Collapse
|
65
|
Manju CA, Jeena K, Ramachandran R, Manohar M, Ambily AM, Sajesh KM, Gowd GS, Menon K, Pavithran K, Pillai A, Nair SV, Koyakutty M. Intracranially injectable multi-siRNA nanomedicine for the inhibition of glioma stem cells. Neurooncol Adv 2021; 3:vdab104. [PMID: 34604750 PMCID: PMC8482790 DOI: 10.1093/noajnl/vdab104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Nanoparticle siRNA-conjugates are promising clinical therapeutics as indicated by recent US-FDA approval. In glioma stem cells (GSC), multiple stemness associated genes were found aberrant. We report intracranially injectable, multi-gene-targeted siRNA nanoparticle gel (NPG) for the combinatorial silencing of 3 aberrant genes, thus inhibiting the tumorogenic potential of GSCs. Methods NPG loaded with siRNAs targeted against FAK, NOTCH-1, and SOX-2 were prepared by the self-assembly of siRNAs with protamine-hyaluronic acid combination. Electron microscopy, DLS, and agarose gel electrophoresis were used for the physicochemical characterization. Cell transfection and gene-silencing efficiency were studied using human mesenchymal stem cells and rat C6 glioma-derived GSCs. Neurosphere inhibition was tested in vitro using GSCs derived from C6 cell line and glioma patient samples. Patient-derived xenograft model and orthotopic rat glioma model were used to test the effect of NPG on in vivo tumorigenicity. Results The siRNA nanoparticles with an average size ~ 250 nm and ~ 95% loading efficiency showed cellular uptake in ~95.5% GSCs. Simultaneous gene silencing of FAK, NOTCH-1, and SOX-2 led to the inhibition of neurosphere formation by GSCs, whereas normal stem cells remained unaffected and retained neuronal differentiation capability. GBM PDX models manifested significant impairment in the tumorigenic potential of NPG treated GSCs. Intracranial injection of NPG inhibited tumor growth in orthotopic rat brain tumor model. Conclusion Intracranially injectable n-siRNA NPG targeted to multiple stem-cell signaling impairs glioma initiation capabilities of GSCs and inhibited tumor growth in vivo.
Collapse
Affiliation(s)
- Cheripelil Abraham Manju
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Kottarapat Jeena
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Ranjith Ramachandran
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Maneesh Manohar
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Anna Mathew Ambily
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | | | | | - Krishnakumar Menon
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Keechilat Pavithran
- Department of Oncology, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Ashok Pillai
- Department of Neurosurgery, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Shantikumar V Nair
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Manzoor Koyakutty
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| |
Collapse
|
66
|
Larrouquère L, Berthier S, Chovelon B, Garrel C, Vacchina V, Paucot H, Boutonnat J, Faure P, Hazane-Puch F. Preclinical Evaluation of Sodium Selenite in Mice: Toxicological and Tumor Regression Studies after Striatum Implantation of Human Glioblastoma Stem Cells. Int J Mol Sci 2021; 22:ijms221910646. [PMID: 34638987 PMCID: PMC8508933 DOI: 10.3390/ijms221910646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/16/2021] [Accepted: 09/27/2021] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive malignant glioma, with a very poor prognosis; as such, efforts to explore new treatments and GBM’s etiology are a priority. We previously described human GBM cells (R2J-GS) as exhibiting the properties of cancer stem cells (growing in serum-free medium and proliferating into nude mice when orthotopically grafted). Sodium selenite (SS)—an in vitro attractive agent for cancer therapy against GBM—was evaluated in R2J-GS cells. To go further, we launched a preclinical study: SS was given orally, in an escalation-dose study (2.25 to 10.125 mg/kg/day, 5 days on, 2 days off, and 5 days on), to evaluate (1) the absorption of selenium in plasma and organs (brain, kidney, liver, and lung) and (2) the SS toxicity. A 6.75 mg/kg SS dose was chosen to perform a tumor regression assay, followed by MRI, in R2J-GS cells orthotopically implanted in nude mice, as this dose was nontoxic and increased brain selenium concentration. A group receiving TMZ (5 mg/kg) was led in parallel. Although not reaching statistical significance, the group of mice treated with SS showed a slower tumor growth vs. the control group (p = 0.08). No difference was observed between the TMZ and control groups. We provide new insights of the mechanisms of SS and its possible use in chemotherapy.
Collapse
Affiliation(s)
- Louis Larrouquère
- Medical Oncology Department, Centre Léon Bérard, 69000 Lyon, France;
| | - Sylvie Berthier
- Cytometry Platform, Institute of Biology and Pathology, Grenoble Alpes Hospital, 38000 Grenoble, France; (S.B.); (J.B.)
- Unit of Anatomopathology, Institute of Biology and Pathology, Grenoble Alpes Hospital, 380000 Grenoble, France
| | - Benoit Chovelon
- Unit Nutritional and Hormonal Biochemistry, Institute of Biology and Pathology, Grenoble Alpes Hospital, 38000 Grenoble, France; (B.C.); (C.G.); (P.F.)
- Department of Molecular Pharmacochemistry, University Grenoble Alpes, CNRS, UMR 5063, 38000 Grenoble, France
| | - Catherine Garrel
- Unit Nutritional and Hormonal Biochemistry, Institute of Biology and Pathology, Grenoble Alpes Hospital, 38000 Grenoble, France; (B.C.); (C.G.); (P.F.)
| | | | - Hugues Paucot
- University of Pau & des Pays de l’Adour, FORCO, Bâtiment d’Alembert-Rue Jules Ferry, BP 27540-64075 Pau CEDEX, France;
| | - Jean Boutonnat
- Cytometry Platform, Institute of Biology and Pathology, Grenoble Alpes Hospital, 38000 Grenoble, France; (S.B.); (J.B.)
- Unit of Anatomopathology, Institute of Biology and Pathology, Grenoble Alpes Hospital, 380000 Grenoble, France
| | - Patrice Faure
- Unit Nutritional and Hormonal Biochemistry, Institute of Biology and Pathology, Grenoble Alpes Hospital, 38000 Grenoble, France; (B.C.); (C.G.); (P.F.)
- Department of Molecular Pharmacochemistry, University Grenoble Alpes, CNRS, UMR 5063, 38000 Grenoble, France
| | - Florence Hazane-Puch
- Unit Nutritional and Hormonal Biochemistry, Institute of Biology and Pathology, Grenoble Alpes Hospital, 38000 Grenoble, France; (B.C.); (C.G.); (P.F.)
- Correspondence: ; Tel.: +33-476769316
| |
Collapse
|
67
|
Farina AR, Cappabianca LA, Zelli V, Sebastiano M, Mackay AR. Mechanisms involved in selecting and maintaining neuroblastoma cancer stem cell populations, and perspectives for therapeutic targeting. World J Stem Cells 2021; 13:685-736. [PMID: 34367474 PMCID: PMC8316860 DOI: 10.4252/wjsc.v13.i7.685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/09/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric neuroblastomas (NBs) are heterogeneous, aggressive, therapy-resistant embryonal tumours that originate from cells of neural crest (NC) origin and in particular neuroblasts committed to the sympathoadrenal progenitor cell lineage. Therapeutic resistance, post-therapeutic relapse and subsequent metastatic NB progression are driven primarily by cancer stem cell (CSC)-like subpopulations, which through their self-renewing capacity, intermittent and slow cell cycles, drug-resistant and reversibly adaptive plastic phenotypes, represent the most important obstacle to improving therapeutic outcomes in unfavourable NBs. In this review, dedicated to NB CSCs and the prospects for their therapeutic eradication, we initiate with brief descriptions of the unique transient vertebrate embryonic NC structure and salient molecular protagonists involved NC induction, specification, epithelial to mesenchymal transition and migratory behaviour, in order to familiarise the reader with the embryonic cellular and molecular origins and background to NB. We follow this by introducing NB and the potential NC-derived stem/progenitor cell origins of NBs, before providing a comprehensive review of the salient molecules, signalling pathways, mechanisms, tumour microenvironmental and therapeutic conditions involved in promoting, selecting and maintaining NB CSC subpopulations, and that underpin their therapy-resistant, self-renewing metastatic behaviour. Finally, we review potential therapeutic strategies and future prospects for targeting and eradication of these bastions of NB therapeutic resistance, post-therapeutic relapse and metastatic progression.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Lucia Annamaria Cappabianca
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Veronica Zelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Michela Sebastiano
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy.
| |
Collapse
|
68
|
Tang X, Zuo C, Fang P, Liu G, Qiu Y, Huang Y, Tang R. Targeting Glioblastoma Stem Cells: A Review on Biomarkers, Signal Pathways and Targeted Therapy. Front Oncol 2021; 11:701291. [PMID: 34307170 PMCID: PMC8297686 DOI: 10.3389/fonc.2021.701291] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) remains the most lethal and common primary brain tumor, even after treatment with multiple therapies, such as surgical resection, chemotherapy, and radiation. Although great advances in medical development and improvements in therapeutic methods of GBM have led to a certain extension of the median survival time of patients, prognosis remains poor. The primary cause of its dismal outcomes is the high rate of tumor recurrence, which is closely related to its resistance to standard therapies. During the last decade, glioblastoma stem cells (GSCs) have been successfully isolated from GBM, and it has been demonstrated that these cells are likely to play an indispensable role in the formation, maintenance, and recurrence of GBM tumors, indicating that GSCs are a crucial target for treatment. Herein, we summarize the current knowledge regarding GSCs, their related signaling pathways, resistance mechanisms, crosstalk linking mechanisms, and microenvironment or niche. Subsequently, we present a framework of targeted therapy for GSCs based on direct strategies, including blockade of the pathways necessary to overcome resistance or prevent their function, promotion of GSC differentiation, virotherapy, and indirect strategies, including targeting the perivascular, hypoxic, and immune niches of the GSCs. In summary, targeting GSCs provides a tremendous opportunity for revolutionary approaches to improve the prognosis and therapy of GBM, despite a variety of challenges.
Collapse
Affiliation(s)
- Xuejia Tang
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China.,Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Chenghai Zuo
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pengchao Fang
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guojing Liu
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yongyi Qiu
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Huang
- Department of Neurosurgery, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Rongrui Tang
- Department of Neurosurgery, University-Town Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
69
|
Titapun A, Luvira V, Srisuk T, Jareanrat A, Thanasukarn V, Thanee M, Sa-Ngiamwibool P, Padthaisong S, Duangkumpha K, Suksawat M, Loilome W, Sithithaworn P, Techasen A, Thinkhamrop B, Dzienny A, Caglayan A, Park D, Mahmud S, Khuntikeo N. High Levels of Serum IgG for Opisthorchis viverrini and CD44 Expression Predict Worse Prognosis for Cholangiocarcinoma Patients after Curative Resection. Int J Gen Med 2021; 14:2191-2204. [PMID: 34103974 PMCID: PMC8179826 DOI: 10.2147/ijgm.s306339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 04/29/2021] [Indexed: 12/26/2022] Open
Abstract
Background Opisthorchis viverrini (OV)-associated cholangiocarcinoma (CCA) has a high immune response with chronic inflammation and oxidative stress. CD44 and Nestin, two cancer stem cell (CSC) markers, play major roles in cancer cell survival. Effects of immune response and expression CSC markers on survival of patients with CCA remain unclear. Objective To investigate the effects of level of OV IgG together with CSC marker expression and also the combination of these markers on survival of CCA patients after curative resection. Methods All serum specimens from CCA patients who underwent curative surgery from 2005 to 2015 were examined for IgG for OV antigen by ELISA. Tissue specimens were studied for CD44 and Nestin expression. Survival analysis by Cox proportional hazard model was used for estimating hazard ratio (HR) with a 95% confidence interval (CI). Results In this study, 122 (69.3%) of 176 were positive for OV IgG, and 35 (19.9%) were considered to have high-positive OV IgG. CD44s positive expression was found in 54 (40%), CD44v6 high expression in 96 (69.6%), CD44v8-10 high expression in 87 (63.5%) and Nestin high expression in 21 (16.1%). Multivariate survival analysis found that high-positive OV IgG and late stage tumor were independent prognostic factors with the adjusted HR of 2.24 (95% CI 1.27–3.93) and 2.78 (95% CI 1.46–5.29), respectively. Subgroup analysis in early and late stage CCA showed that a combined positive OV IgG and CD44s expression with the high expression of CD44v8-10 had the significantly poorest prognosis with HR of 3.75 (95% CI 1.61–8.72) and HR of 1.76 (95% CI 1.02–3.03), respectively. Conclusion A high level of OV IgG as well as a high level of CSC markers resulted in an aggressive CCA. OV IgG level together with CSC markers can be used as the prognostic markers for CCA patients’ survival. The study of the CD44 pathway is promising for adjuvant treatment.
Collapse
Affiliation(s)
- Attapol Titapun
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand
| | - Vor Luvira
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand
| | - Tharatip Srisuk
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand
| | - Apiwat Jareanrat
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand
| | - Vasin Thanasukarn
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand
| | - Malinee Thanee
- Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand.,Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Prakasit Sa-Ngiamwibool
- Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand.,Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sureerat Padthaisong
- Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kassaporn Duangkumpha
- Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Manida Suksawat
- Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Paiboon Sithithaworn
- Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand.,Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Anchalee Techasen
- Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand.,Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Bandit Thinkhamrop
- Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand.,Department of Epidemiology and Biostatistics, Faculty of Public Health, Khon Kaen University, Khon Kaen, Thailand
| | - Alexa Dzienny
- School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| | - Ayse Caglayan
- Faculty of Medicine, Imperial College London, London, UK
| | - David Park
- Faculty of Medicine, Imperial College London, London, UK
| | - Simran Mahmud
- Faculty of Medicine, Imperial College London, London, UK
| | - Narong Khuntikeo
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute (CARI), Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
70
|
Yoshimura H, Moriya M, Yoshida A, Yamamoto M, Machida Y, Ochiai K, Michishita M, Nakagawa T, Matsuda Y, Takahashi K, Kamiya S, Ishiwata T. Involvement of Nestin in the Progression of Canine Mammary Carcinoma. Vet Pathol 2021; 58:994-1003. [PMID: 34056976 DOI: 10.1177/03009858211018656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Nestin, a class VI intermediate filament protein, is known to be expressed in various types of human neoplasms, including breast cancer, and is associated with their progression. However, its expression and role in canine mammary tumors remain unknown. We analyzed nestin expression in canine mammary tumors using in situ hybridization and immunohistochemistry. We also investigated its role in a canine mammary carcinoma cell line using RNA interference. Nestin expression was not observed in luminal epithelial cells of any of the 62 cases of benign mammary lesions examined, although myoepithelial cells showed its expression in most cases. In 16/50 (32%) primary mammary carcinomas and 6/15 (40%) metastases of mammary carcinomas, cytoplasmic nestin expression was detected in luminal epithelial cells. In luminal cells of primary mammary carcinomas, its expression was positively related to several pathological parameters that indicate high-grade malignancy, including histological grading (P < .01), vascular/lymphatic invasion (P < .01), Ki-67 index (P < .01), and metastasis (P < .05). Immunohistochemistry revealed that nestin expression was related to vimentin expression in mammary carcinomas (P < .01). This relationship was confirmed using reverse transcription-quantitative polymerase chain reaction using 9 cell lines derived from canine mammary carcinoma (P < .01). Finally, nestin knockdown in canine mammary carcinoma cells using small interfering RNA inhibited cell proliferation and migration based on WST-8, Boyden chamber, and cell-tracking assays. These findings suggest that nestin may at least partially mediate these behaviors of canine mammary carcinoma cells.
Collapse
Affiliation(s)
| | - Maiko Moriya
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Ayaka Yoshida
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Masami Yamamoto
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Yukino Machida
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Kazuhiko Ochiai
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | | | | | | | | | - Shinji Kamiya
- 12989Nippon Veterinary and Life Science University, Tokyo, Japan
| | | |
Collapse
|
71
|
Zhang ET, Hannibal RL, Badillo Rivera KM, Song JHT, McGowan K, Zhu X, Meinhardt G, Knöfler M, Pollheimer J, Urban AE, Folkins AK, Lyell DJ, Baker JC. PRG2 and AQPEP are misexpressed in fetal membranes in placenta previa and percreta†. Biol Reprod 2021; 105:244-257. [PMID: 33982062 DOI: 10.1093/biolre/ioab068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 03/03/2021] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
The obstetrical conditions placenta accreta spectrum (PAS) and placenta previa are a significant source of pregnancy-associated morbidity and mortality, yet the specific molecular and cellular underpinnings of these conditions are not known. In this study, we identified misregulated gene expression patterns in tissues from placenta previa and percreta (the most extreme form of PAS) compared with control cases. By comparing this gene set with existing placental single-cell and bulk RNA-Seq datasets, we show that the upregulated genes predominantly mark extravillous trophoblasts. We performed immunofluorescence on several candidate molecules and found that PRG2 and AQPEP protein levels are upregulated in both the fetal membranes and the placental disk in both conditions. While this increased AQPEP expression remains restricted to trophoblasts, PRG2 is mislocalized and is found throughout the fetal membranes. Using a larger patient cohort with a diverse set of gestationally aged-matched controls, we validated PRG2 as a marker for both previa and PAS and AQPEP as a marker for only previa in the fetal membranes. Our findings suggest that the extraembryonic tissues surrounding the conceptus, including both the fetal membranes and the placental disk, harbor a signature of previa and PAS that is characteristic of EVTs and that may reflect increased trophoblast invasiveness.
Collapse
Affiliation(s)
- Elisa T Zhang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberta L Hannibal
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Janet H T Song
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kelly McGowan
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaowei Zhu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Gudrun Meinhardt
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Martin Knöfler
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Jürgen Pollheimer
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Alexander E Urban
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Ann K Folkins
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Deirdre J Lyell
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Julie C Baker
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
72
|
Rasti A, Madjd Z, Saeednejad Zanjani L, Babashah S, Abolhasani M, Asgari M, Mehrazma M. SMAD4 Expression in Renal Cell Carcinomas Correlates With a Stem-Cell Phenotype and Poor Clinical Outcomes. Front Oncol 2021; 11:581172. [PMID: 34012911 PMCID: PMC8127783 DOI: 10.3389/fonc.2021.581172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 03/18/2021] [Indexed: 01/17/2023] Open
Abstract
Renal cell carcinoma (RCC) is the most lethal neoplasm of common urologic cancers with poor prognoses. SMAD4 has a principal role in TGF-β (Transformis growth factorβ)-induced epithelial to mesenchymal transition (EMT) as a key factor in gaining cancer stem cell (CSC) features and tumor aggressiveness. This study aimed to evaluate the expression patterns and clinical significance of SMAD4 in RCC and the impact of its targeting on stem cell/mesenchymal cells and EMT characteristics in renal spheroid derived cells (SDCs) compared to parental cells (PCs) in RCC. The expression pattern and clinical significance of SMAD4 was evaluated in RCC. SDCs were enriched using a sphere culture system. Then SDCs and their PCs were compared with respect to their sphere and colony formation, expression of putative CSC markers, invasiveness as well as expression of genes, including stemness/mesenchymal, SMAD4 and TGFβ1genes. Finally, the effect of SMAD4 knockdown on SDCs was analyzed. We demonstrated that SMAD4 is positively correlated with decreased disease specific survival (DSS) in RCC patients and clear cell RCC (ccRCC) subtype and associates with poor DSS in patients with RCC, especially in ccRCC as the most metastatic RCC subtype. SDCs exhibited higher stem cell/mesenchymal properties. Inhibition of SMAD4 in PCs accelerated the dissociation of SDCs and decreased their clonogenicity, invasiveness, expression of mesenchymal markers and expression of SMAD4 and TGFβ1 genes compared to SDCs before transfection. We suggest that targeting SMAD4 may be useful against renal CSCs and may improve RCC prognosis.
Collapse
Affiliation(s)
- Arezoo Rasti
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Basic Sciences/Medical Surgical Nursing, Faculty of Nursing and Midwifery, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University (TMU), Tehran, Iran
| | - Maryam Abolhasani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Hasheminejad Kidney Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mojgan Asgari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Hasheminejad Kidney Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mitra Mehrazma
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Hasheminejad Kidney Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
73
|
Windmöller BA, Beshay M, Helweg LP, Flottmann C, Beermann M, Förster C, Wilkens L, Greiner JFW, Kaltschmidt C, Kaltschmidt B. Novel Primary Human Cancer Stem-Like Cell Populations from Non-Small Cell Lung Cancer: Inhibition of Cell Survival by Targeting NF-κB and MYC Signaling. Cells 2021; 10:cells10051024. [PMID: 33925297 PMCID: PMC8145874 DOI: 10.3390/cells10051024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
There is growing evidence that cancer stem cells (CSCs), a small subpopulation of self-renewal cancer cells, are responsible for tumor growth, treatment resistance, and cancer relapse and are thus of enormous clinical interest. Here, we aimed to isolate new CSC-like cells derived from human primary non-small cell lung cancer (NSCLC) specimens and to analyze the influence of different inhibitors of NF-κB and MYC signaling on cell survival. CSC-like cells were established from three squamous cell carcinomas (SCC) and three adenocarcinomas (AC) of the lung and were shown to express common CSC markers such as Prominin-1, CD44-antigen, and Nestin. Further, cells gave rise to spherical cancer organoids. Inhibition of MYC and NF-κB signaling using KJ-Pyr-9, dexamethasone, and pyrrolidinedithiocarbamate resulted in significant reductions in cell survival for SCC- and AC-derived cells. However, inhibition of the protein–protein interaction of MYC/NMYC proto-oncogenes with Myc-associated factor X (MAX) using KJ-Pyr-9 revealed the most promising survival-decreasing effects. Next to the establishment of six novel in vitro models for studying NSCLC-derived CSC-like populations, the presented investigations might provide new insights into potential novel therapies targeting NF-κB/MYC to improve clinical outcomes in NSCLC patients. Nevertheless, the full picture of downstream signaling still remains elusive.
Collapse
Affiliation(s)
- Beatrice A. Windmöller
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (L.P.H.); (C.F.); (M.B.); (J.F.W.G.); (C.K.); (B.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e. V., Maraweg 21, 33617 Bielefeld, Germany; (M.B.); (C.F.); (L.W.)
- Correspondence: ; Tel.: +49-0521-106-5629
| | - Morris Beshay
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e. V., Maraweg 21, 33617 Bielefeld, Germany; (M.B.); (C.F.); (L.W.)
- Department of General Thoracic Surgery, Protestant Hospital of Bethel Foundation, Burgsteig 13, 33617 Bielefeld, Germany
| | - Laureen P. Helweg
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (L.P.H.); (C.F.); (M.B.); (J.F.W.G.); (C.K.); (B.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e. V., Maraweg 21, 33617 Bielefeld, Germany; (M.B.); (C.F.); (L.W.)
| | - Clara Flottmann
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (L.P.H.); (C.F.); (M.B.); (J.F.W.G.); (C.K.); (B.K.)
| | - Miriam Beermann
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (L.P.H.); (C.F.); (M.B.); (J.F.W.G.); (C.K.); (B.K.)
| | - Christine Förster
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e. V., Maraweg 21, 33617 Bielefeld, Germany; (M.B.); (C.F.); (L.W.)
- Institute of Pathology, KRH Hospital Nordstadt, Haltenhoffstrasse 41, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany
| | - Ludwig Wilkens
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e. V., Maraweg 21, 33617 Bielefeld, Germany; (M.B.); (C.F.); (L.W.)
- Institute of Pathology, KRH Hospital Nordstadt, Haltenhoffstrasse 41, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany
| | - Johannes F. W. Greiner
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (L.P.H.); (C.F.); (M.B.); (J.F.W.G.); (C.K.); (B.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e. V., Maraweg 21, 33617 Bielefeld, Germany; (M.B.); (C.F.); (L.W.)
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (L.P.H.); (C.F.); (M.B.); (J.F.W.G.); (C.K.); (B.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e. V., Maraweg 21, 33617 Bielefeld, Germany; (M.B.); (C.F.); (L.W.)
| | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (L.P.H.); (C.F.); (M.B.); (J.F.W.G.); (C.K.); (B.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e. V., Maraweg 21, 33617 Bielefeld, Germany; (M.B.); (C.F.); (L.W.)
- Molecular Neurobiology, Bielefeld University, Universitätsstrasse 25, 33615 Bielefeld, Germany
| |
Collapse
|
74
|
Taylor JT, Ellison S, Pandele A, Wood S, Nathan E, Forte G, Parker H, Zindy E, Elvin M, Dickson A, Williams KJ, Karabatsou K, McCabe M, McBain C, Bigger BW. Actinomycin D downregulates Sox2 and improves survival in preclinical models of recurrent glioblastoma. Neuro Oncol 2021; 22:1289-1301. [PMID: 32227096 PMCID: PMC7523458 DOI: 10.1093/neuonc/noaa051] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) has been extensively researched over the last few decades, yet despite aggressive multimodal treatment, recurrence is inevitable and second-line treatment options are limited. Here, we demonstrate how high-throughput screening (HTS) in multicellular spheroids can generate physiologically relevant patient chemosensitivity data using patient-derived cells in a rapid and cost-effective manner. Our HTS system identified actinomycin D (ACTD) to be highly cytotoxic over a panel of 12 patient-derived glioma stemlike cell (GSC) lines. ACTD is an antineoplastic antibiotic used in the treatment of childhood cancers. Here, we validate ACTD as a potential repurposed therapeutic for GBM in 3-dimensional GSC cultures and patient-derived xenograft models of recurrent glioblastoma. METHODS Twelve patient-derived GSC lines were screened at 10 µM, as multicellular spheroids, in a 384-well serum-free assay with 133 FDA-approved compounds. GSCs were then treated in vitro with ACTD at established half-maximal inhibitory concentrations (IC50). Downregulation of sex determining region Y-box 2 (Sox2), a stem cell transcription factor, was investigated via western blot and through immunohistological assessment of murine brain tissue. RESULTS Treatment with ACTD was shown to significantly reduce tumor growth in 2 recurrent GBM patient-derived models and significantly increased survival. ACTD is also shown to specifically downregulate the expression of Sox2 both in vitro and in vivo. CONCLUSION These findings indicate that, as predicted by our HTS, ACTD could deplete the cancer stem cell population within the tumor mass, ultimately leading to a delay in tumor progression. KEY POINTS 1. High-throughput chemosensitivity data demonstrated the broad efficacy of actinomycin D, which was validated in 3 preclinical models of glioblastoma.2. Actinomycin D downregulated Sox2 in vitro and in vivo, indicating that this agent could target the stem cell population of GBM tumors.
Collapse
Affiliation(s)
- Jessica T Taylor
- Brain Tumor Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| | - Stuart Ellison
- Brain Tumor Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| | - Alina Pandele
- Brain Tumor Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| | - Shaun Wood
- Brain Tumor Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| | - Erica Nathan
- CRUK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | - Gabriella Forte
- Brain Tumor Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| | - Helen Parker
- Brain Tumor Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| | - Egor Zindy
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Mark Elvin
- Manchester Institute of Biotechnology, Faculty of Science and Engineering, University of Manchester, Manchester, UK
| | - Alan Dickson
- Manchester Institute of Biotechnology, Faculty of Science and Engineering, University of Manchester, Manchester, UK
| | - Kaye J Williams
- Division of Pharmacy and Optometry, School of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Martin McCabe
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Catherine McBain
- Department of Clinical Oncology, The Christie NHS FT, Manchester, UK
| | - Brian W Bigger
- Brain Tumor Research Group, Stem Cell and Neurotherapies Laboratory, Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| |
Collapse
|
75
|
Jaraíz-Rodríguez M, Talaverón R, García-Vicente L, Pelaz SG, Domínguez-Prieto M, Álvarez-Vázquez A, Flores-Hernández R, Sin WC, Bechberger J, Medina JM, Naus CC, Tabernero A. Connexin43 peptide, TAT-Cx43266-283, selectively targets glioma cells, impairs malignant growth, and enhances survival in mouse models in vivo. Neuro Oncol 2021; 22:493-504. [PMID: 31883012 PMCID: PMC7158688 DOI: 10.1093/neuonc/noz243] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Malignant gliomas are the most frequent primary brain tumors and remain among the most incurable cancers. Although the role of the gap junction protein, connexin43 (Cx43), has been deeply investigated in malignant gliomas, no compounds have been reported with the ability to recapitulate the tumor suppressor properties of this protein in in vivo glioma models. Methods TAT-Cx43266–283 a cell-penetrating peptide which mimics the effect of Cx43 on c-Src inhibition, was studied in orthotopic immunocompetent and immunosuppressed models of glioma. The effects of this peptide in brain cells were also analyzed. Results While glioma stem cell malignant features were strongly affected by TAT-Cx43266–283, these properties were not significantly modified in neurons and astrocytes. Intraperitoneally administered TAT-Cx43266–283 decreased the invasion of intracranial tumors generated by GL261 mouse glioma cells in immunocompetent mice. When human glioma stem cells were intracranially injected with TAT-Cx43266–283 into immunodeficient mice, there was reduced expression of the stemness markers nestin and Sox2 in human glioma cells at 7 days post-implantation. Consistent with the role of Sox2 as a transcription factor required for tumorigenicity, TAT-Cx43266–283 reduced the number and stemness of human glioma cells at 30 days post-implantation. Furthermore, TAT-Cx43266–283 enhanced the survival of immunocompetent mice bearing gliomas derived from murine glioma stem cells. Conclusion TAT-Cx43266–283 reduces the growth, invasion, and progression of malignant gliomas and enhances the survival of glioma-bearing mice without exerting toxicity in endogenous brain cells, which suggests that this peptide could be considered as a new clinical therapy for high-grade gliomas.
Collapse
Affiliation(s)
- Myriam Jaraíz-Rodríguez
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Rocío Talaverón
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Laura García-Vicente
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Sara G Pelaz
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Marta Domínguez-Prieto
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Andrea Álvarez-Vázquez
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Raquel Flores-Hernández
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Wun Chey Sin
- Department of Cellular and Physiological Sciences, The Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - John Bechberger
- Department of Cellular and Physiological Sciences, The Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - José M Medina
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| | - Christian C Naus
- Department of Cellular and Physiological Sciences, The Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Arantxa Tabernero
- Department of Biochemistry and Molecular Biology, Institute of Neurosciences Castilla y León (INCYL), University of Salamanca, Salamanca, Spain
| |
Collapse
|
76
|
Ye H, Pan J, Gong E, Cai X, Xu C, Li Y, Zheng H, Cao Z. Inhibitory Effect of Immunologically Activated Mesenchymal Stem Cells on Lung Cancer Cell Growth and Metastasis. Cancer Biother Radiopharm 2021. [PMID: 33769841 DOI: 10.1089/cbr.2020.3855] [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: 10/21/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) could inhibit the proliferation of lung cancer cells. The authors' study aimed to investigate the effects of immunologically activated human umbilical cord (HUC)-MSCs on A549 lung cancer cells. Materials and Methods: HUC-MSCs were separated from the umbilical cord using the adherence method. Surface markers of HUC-MSCs were detected by flow cytometry for MSC identification. Imiquimod (TLR7 agonist) was incubated with HUC-MSCs for immune activation, and the expression of MSC-specific markers and immune inflammatory molecules was measured by quantitative real-time polymerase chain reaction. HUC A549 cells were cocultured with HUC-MSCs treated with imiquimod, siTLR7 (small interfering RNA for TLR7) or TLR7 overexpression, and then cell viability, proliferation, migration, and invasion, and the expression of phosphatidylinositol-3-kinase (PI3K)/Akt and NF-κB was investigated using MTT assay, clone formation assay, transwell assay, and western blot, respectively. Results: HUC-MSCs were identified as positive for CD73, CD105, CD44, CD29, and CD90. Expression of MSC markers was inhibited, while those of immune inflammatory molecules expression except IL-6 (interleukin-6) was enhanced after MSCs were immunologically activated by imiquimod. After being cocultured with HUC-MSCs treated with imiquimod or overexpressed TLR7, cell viability, proliferation, and metastasis, and the phosphorylation of P65 and AKT in A549 cells were decreased, but apoptosis was increased, while siTLR7 showed the opposite effect HUC. Conclusions: Immunologically activated HUC-MSCs inhibited the growth and metastasis, yet, promoted the apoptosis of A549 lung cancer cells via regulating the PI3K/Akt and NF-κB pathways.
Collapse
Affiliation(s)
- Hong Ye
- Department of Medical Examination Center, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Jiongwei Pan
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Enhui Gong
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Xiaoping Cai
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Cunlai Xu
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Yuling Li
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Hao Zheng
- Department of Respiratory, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Zhuo Cao
- Department of Respiration, The Sixth Affiliated Hospital of Wenzhou Medical University, People's Hospital of Longquan, Lishui, China
| |
Collapse
|
77
|
Su C, Zhang J, Yarden Y, Fu L. The key roles of cancer stem cell-derived extracellular vesicles. Signal Transduct Target Ther 2021; 6:109. [PMID: 33678805 PMCID: PMC7937675 DOI: 10.1038/s41392-021-00499-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs), the subpopulation of cancer cells, have the capability of proliferation, self-renewal, and differentiation. The presence of CSCs is a key factor leading to tumor progression and metastasis. Extracellular vesicles (EVs) are nano-sized particles released by different kinds of cells and have the capacity to deliver certain cargoes, such as nucleic acids, proteins, and lipids, which have been recognized as a vital mediator in cell-to-cell communication. Recently, more and more studies have reported that EVs shed by CSCs make a significant contribution to tumor progression. CSCs-derived EVs are involved in tumor resistance, metastasis, angiogenesis, as well as the maintenance of stemness phenotype and tumor immunosuppression microenvironment. Here, we summarized the molecular mechanism by which CSCs-derived EVs in tumor progression. We believed that the fully understanding of the roles of CSCs-derived EVs in tumor development will definitely provide new ideas for CSCs-based therapeutic strategies.
Collapse
Affiliation(s)
- Chaoyue Su
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China ,grid.410737.60000 0000 8653 1072Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Jianye Zhang
- grid.410737.60000 0000 8653 1072Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Yosef Yarden
- grid.13992.300000 0004 0604 7563Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Liwu Fu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| |
Collapse
|
78
|
Wu H, Du J, Li H, Li Y, Zhang W, Zhou W, Wang W. Aberrant expression of thyroid transcription factor-1 in meningeal solitary fibrous tumor/hemangiopericytoma. Brain Tumor Pathol 2021; 38:122-131. [PMID: 33666786 DOI: 10.1007/s10014-021-00395-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/16/2021] [Indexed: 11/25/2022]
Abstract
Meningeal solitary fibrous tumor (SFT) and hemangiopericytoma (HPC) were categorized as the same entity in the World Health Organization (WHO) 2016 classification of tumors of the central nervous system (CNS). Although NAB2-STAT6 fusion protein can be used to distinguish most of SFT/HPC from the other sarcomas, additional biomarkers were requested to separate meningeal SFT/HPC from meningioma and the molecular pathological difference between meningeal SFT/HPC and extra-CNS SFT/HPC still remains unclear. In this study, we evaluated the expression of TTF-1 in 67 meningeal SFT/HPC, 62 extra-CNS SFT/HPC and 201 meningiomas samples with immunohistochemistry (IHC) assays. The results showed that TTF-1 was detected in 23 of 67 (34.3%) meningeal SFT/HPC, 3 retroperitoneum SFT/HPC and none of meningiomas. Meanwhile, the copy number variation and mRNA expression of TTF-1 were measured by real-time quantitative PCR (qPCR) in meningeal SFT/HPC. These results demonstrated that TTF-1 protein expression level was significantly correlated with its transcription level, but independently related to the gene copy number variant. In conclusion, our study suggested that a large proportion of meningeal SFT/HPC was positive to TTF-1, while very few extra CNS SFT/HPC cases and no meningiomas were stained. So TTF-1 has value as an auxiliary diagnostic marker for meningeal SFT/HPC.
Collapse
Affiliation(s)
- Haibo Wu
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.,Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Jun Du
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.,Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Heng Li
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.,Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Yujie Li
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.,Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Wanqiu Zhang
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.,Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Wenchao Zhou
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China
| | - Wei Wang
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China. .,Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, Anhui, China.
| |
Collapse
|
79
|
Li G, Chen TW, Nickel AC, Muhammad S, Steiger HJ, Tzaridis T, Hänggi D, Zeidler R, Zhang W, Kahlert UD. Carbonic Anhydrase XII is a Clinically Significant, Molecular Tumor-Subtype Specific Therapeutic Target in Glioma with the Potential to Combat Invasion of Brain Tumor Cells. Onco Targets Ther 2021; 14:1707-1718. [PMID: 33692626 PMCID: PMC7939492 DOI: 10.2147/ott.s300623] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/05/2021] [Indexed: 12/15/2022] Open
Abstract
Background The metabolic enzyme carbonic anhydrase 12 (CA12/CAXII) emerges as a promising cancer therapeutic target with drug development projects underway. Previous reports proposed the relevance of CA12 in the context of glioma but are limited in patient data quantity, ignore ethnic diversity of patients or rely on semi-quantitative, thereby out of date, methodology. Moreover, little is known on the association of CA12 to brain tumor stemness or on the effect of anti-CAXII-directed monotherapies on glioma stem cells (GSCs), in particular their response regarding mesenchymal differentiation status. Methods We performed in silico analysis on three independent, large-scale patient datasets interrogating state of the art molecular diagnostics alongside clinical outcomes. We analyzed CAXII abundance on a collection of GSCs and functionally tested their response to exposure to CAXII blocking antibody 6A10. Results CA12 is highly expressed in glial tumors compared with normal tissue and predicts for poor clinical course of tumor patients. CA12 expression in glioblastoma significantly correlates with clinically established, molecular markers of IDH1WT DNA, WHO grade IV or absence of 1p/19q chromosome arm co-deletion. Furthermore, tumors with elevated CA12 cluster into the mesenchymal transcription subclass of the disease. CAXII abundance in different GSCs ranges from almost absent to high levels and does not correlate to stem cell marker CD133/AC133 cell surface expression. Moreover, aiming to pharmacologically block CAXII in our cells with antibody 6A10 caused significant functional response only in one of the tested GSCs models, featuring suppression of cell invasion accompanied by reduction of ZEB1 protein and other stem cell markers. Conclusion CA12 represents a clinically relevant and molecular brain tumor-subtype specific therapeutic target. Our correlative data from experimental and clinical samples does not support CA12/CAXII to be GSC specific. 6A10 possesses promising potential to impede the invasive capacity of glioma cells and supports the emerging concept that CAXII interacts with cancer EMT programs. However, further mechanistic studies are required to comprehensively assess the therapeutic potential of 6A10 and to identify different resistance mechanisms of GSCs.
Collapse
Affiliation(s)
- Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China
| | - Ting-Wei Chen
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Ann-Christin Nickel
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Sajjad Muhammad
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hans-Jakob Steiger
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Theophilos Tzaridis
- Division of Clinical Neurooncology, Department of Neurology and Institute of Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, 53127, Germany.,Tumor Initiation & Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Daniel Hänggi
- Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Reinhard Zeidler
- Department for Otorhinolaryngology, Klinikum der Universität München (LMU), Munich, Germany
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, People's Republic of China
| | - Ulf Dietrich Kahlert
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, People's Republic of China.,Clinic for Neurosurgery, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
80
|
Sánchez-Salazar MG, Álvarez MM, Trujillo-de Santiago G. Advances in 3D bioprinting for the biofabrication of tumor models. ACTA ACUST UNITED AC 2021. [DOI: 10.1016/j.bprint.2020.e00120] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
81
|
Gómez-Oliva R, Domínguez-García S, Carrascal L, Abalos-Martínez J, Pardillo-Díaz R, Verástegui C, Castro C, Nunez-Abades P, Geribaldi-Doldán N. Evolution of Experimental Models in the Study of Glioblastoma: Toward Finding Efficient Treatments. Front Oncol 2021; 10:614295. [PMID: 33585240 PMCID: PMC7878535 DOI: 10.3389/fonc.2020.614295] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is the most common form of brain tumor characterized by its resistance to conventional therapies, including temozolomide, the most widely used chemotherapeutic agent in the treatment of GBM. Within the tumor, the presence of glioma stem cells (GSC) seems to be the reason for drug resistance. The discovery of GSC has boosted the search for new experimental models to study GBM, which allow the development of new GBM treatments targeting these cells. In here, we describe different strategies currently in use to study GBM. Initial GBM investigations were focused in the development of xenograft assays. Thereafter, techniques advanced to dissociate tumor cells into single-cell suspensions, which generate aggregates referred to as neurospheres, thus facilitating their selective expansion. Concomitantly, the finding of genes involved in the initiation and progression of GBM tumors, led to the generation of mice models for the GBM. The latest advances have been the use of GBM organoids or 3D-bioprinted mini-brains. 3D bio-printing mimics tissue cytoarchitecture by combining different types of cells interacting with each other and with extracellular matrix components. These in vivo models faithfully replicate human diseases in which the effect of new drugs can easily be tested. Based on recent data from human glioblastoma, this review critically evaluates the different experimental models used in the study of GB, including cell cultures, mouse models, brain organoids, and 3D bioprinting focusing in the advantages and disadvantages of each approach to understand the mechanisms involved in the progression and treatment response of this devastating disease.
Collapse
Affiliation(s)
- Ricardo Gómez-Oliva
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| | - Samuel Domínguez-García
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| | - Livia Carrascal
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain.,Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | | | - Ricardo Pardillo-Díaz
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| | - Cristina Verástegui
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain.,Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
| | - Carmen Castro
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| | - Pedro Nunez-Abades
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain.,Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Noelia Geribaldi-Doldán
- Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| |
Collapse
|
82
|
Aglan SA, Elsammak M, Elsammak O, El-Bakoury EA, Elsheredy HG, Ahmed YS, Sultan MH, Awad AM. Evaluation of serum Nestin and HOTAIR rs12826786 C>T polymorphism as screening tools for breast cancer in Egyptian women. J Med Biochem 2021; 40:17-25. [PMID: 33584136 PMCID: PMC7857851 DOI: 10.5937/jomb0-25295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Nestin is a neural stem cell protein that plays an important role in cancer stem cells (CSC) development and proliferation. It has been identified as a marker for newly formed endothelial cells and was shown to be preferentially expressed in basal and myoepithelial cells of the mammary gland. HOTAIR is long intergenic non-coding (linRNA) associated with tumorigenesis through promotion of epithelial-mesenchymal transition (EMT) and stemness as well. HOTAIR gene contains a functioning single nucleotide polymorphic site rs12826786 C>T that has been associated with several cancer types. METHODS We evaluated serum Nestin and the HOTAIR rs12826786 C>T polymorphism in healthy Egyptian women and those with breast cancer as a possible screening tool to identify patients with breast cancer. Also, we tested the possible association of the two markers with each other and the aggressiveness of the disease. RESULTS Patients with breast cancer had a median (Min-Max) of serum Nestin 31.3 (6.7-167.3 pg/mL), while control subjects had a median (Min-Max) of serum Nestin 42.3 (25.7-315.95) pg/mL. The best cut-off value for serum Nestin to differentiate normal subjects and patients with breast cancer was 39.9 pg/mL. This cut-off value had a diagnostic sensitivity of 84.8% and specificity of 65.1%. There was a significant difference in the distribution of different alleles in patients with breast cancer than normal subjects (P=0.039 Exact Fisher test). The breast cancer patients group had 23.9% CC, 52.1% CT, and 23.9% TT genotypes, respectively, while the control group had 46.9% CC, 42.8% CT, and 10.2% TT, respectively. CONCLUSIONS A significantly low serum Nestin below 39.9 pg/mL and a higher percentage of the T/T homozygous variant allele of HOTAIR rs12826786 C>T were found in Egyptian patients with breast cancer. We suggest that the reported cut-off value of serum Nestin and the presence of C/T polymorphism can be used to assess the risk of females for developing breast cancer and might be of potential benefit in screening the disease. Larger studies in different ethnic groups are needed to confirm our findings.
Collapse
Affiliation(s)
- Sarah A. Aglan
- Alexandria University, Medical Research Institute, Department of Chemical Pathology, Egypt
| | - Mohamed Elsammak
- Alexandria University, Medical Research Institute, Department of Chemical Pathology, Egypt
| | | | - Eman A. El-Bakoury
- Alexandria University, Medical Research Institute, Department of Radio-diagnosis, Egypt
| | - Heba G. Elsheredy
- Alexandria University, Medical Research Institute, Department of Cancer Management and Research, Alexandria, Egypt
| | - Yasser S. Ahmed
- Alexandria University, Medical Research Institute, Department of Experimental and Clinical Surgery, Alexandria, Egypt
| | - Mohamed H. Sultan
- Alexandria University, Medical Research Institute, Department of Experimental and Clinical Surgery, Alexandria, Egypt
| | - Ahmed M. Awad
- Alexandria University, Medical Research Institute, Department of Chemical Pathology, Egypt
| |
Collapse
|
83
|
Poulin JF, Luppi MP, Hofer C, Caronia G, Hsu PK, Chan CS, Awatramani R. PRISM: A Progenitor-Restricted Intersectional Fate Mapping Approach Redefines Forebrain Lineages. Dev Cell 2021; 53:740-753.e3. [PMID: 32574593 DOI: 10.1016/j.devcel.2020.05.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/24/2020] [Accepted: 05/15/2020] [Indexed: 01/08/2023]
Abstract
Lineage tracing aims to identify the progeny of a defined population of dividing progenitor cells, a daunting task in the developing central nervous system where thousands of cell types are generated. In mice, lineage analysis has been accomplished using Cre recombinase to indelibly label a defined progenitor population and its progeny. However, the interpretation of historical recombination events is hampered by the fact that driver genes are often expressed in both progenitors and postmitotic cells. Genetically inducible approaches provide temporal specificity but are afflicted by mosaicism and toxicity. Here, we present PRISM, a progenitor-restricted intersectional fate mapping approach in which Flp recombinase expression is both dependent on Cre and restricted to neural progenitors, thus circumventing the aforementioned confounds. This tool can be used in conjunction with existing Cre lines making it broadly applicable. We applied PRISM to resolve two developmentally important, but contentious, lineages-Shh and Cux2.
Collapse
Affiliation(s)
- Jean-François Poulin
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University Montreal, Quebec H3A 0G4, Canada
| | - Milagros Pereira Luppi
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Caitlyn Hofer
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Giuliana Caronia
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Pei-Ken Hsu
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - C Savio Chan
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rajeshwar Awatramani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
84
|
Zhong C, Tao B, Tang F, Yang X, Peng T, You J, Xia K, Xia X, Chen L, Peng L. Remodeling cancer stemness by collagen/fibronectin via the AKT and CDC42 signaling pathway crosstalk in glioma. Am J Cancer Res 2021; 11:1991-2005. [PMID: 33408794 PMCID: PMC7778591 DOI: 10.7150/thno.50613] [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: 07/13/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer development is a complex set of proliferative progression, which arises in most cases via multistep pathways associated with various factors, including the tumor microenvironment and extracellular matrix. However, the underlying mechanisms of cancer development remain unclear and this study aimed to explore the role of extracellular matrix in glioma progression. Methods: The expression of type I collagen and fibronectin in tumor tissues from glioma patients was examined by immunofluorescence staining. The correlations between collagen/fibronectin and glioma progression were then analyzed. A 3D collagen/fibronectin cultured system was established for tumor cells culture in vitro. Quantitative, real-time PCR and western blot were used to detect PI3K/ATK and CDC42 signals associated proteins expression in glioma. We used in vitro Cell Counting Kit-8, colony formation, and tumorigenesis assays to investigate the function of PI3K/AKT and CDC42 signals associated proteins. A xenograft glioma mice model was also used to study the anticancer effects of integrin inhibitor in vivo. Results: Our study demonstrated that type I collagen and fibronectin collaborate to regulate glioma cell stemness and tumor growth. In a 3D collagen/fibronectin culture model, glioma cells acquired tumorigenic potential and revealed strengthened proliferative characteristics. More significantly, collagen/fibronectin could facilitate the activation of PI3K/AKT/SOX2 and CDC42/YAP-1/NUPR1/Nestin signaling pathways via integrin αvβ3, eliciting sustained tumor growth and cancer relapse. Combination of the integrin signaling pathway inhibitor and the chemotherapeutic agent efficiently suppressed glioma cell proliferation and tumorigenic ability. Conclusion: We demonstrated that type I collagen and fibronectin could collaborate to promote glioma progression through PI3K/AKT/SOX2 and CDC42/YAP-1/NUPR1/Nestin signaling pathways. Blockade of the upstream molecular integrin αvβ3 revealed improved outcome in glioma therapy, which provide new insights for eradicating tumors and reducing glioma cancer relapse.
Collapse
|
85
|
Li M, Li G, Kiyokawa J, Tirmizi Z, Richardson LG, Ning J, Das S, Martuza RL, Stemmer-Rachamimov A, Rabkin SD, Wakimoto H. Characterization and oncolytic virus targeting of FAP-expressing tumor-associated pericytes in glioblastoma. Acta Neuropathol Commun 2020; 8:221. [PMID: 33308315 PMCID: PMC7730751 DOI: 10.1186/s40478-020-01096-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are activated fibroblasts constituting the major stromal components in many types of cancer. CAFs contribute to hallmarks of cancer such as proliferation, invasion and immunosuppressive tumor microenvironment, and are associated with poor prognosis of patients with cancer. However, in glioblastoma (GBM), the most common and aggressive primary malignant brain tumor, our knowledge about CAFs or CAF-like stromal cells is limited. Here, using commonly accepted CAF markers, we characterized CAF-like cell populations in clinical glioma specimens and datasets along with mouse models of GBM. We found that tumor-associated pericytes marked by co-expression of fibroblast activation protein α (FAP) and PDGFRβ represent major stromal cell subsets in both human GBM and mouse GBM models, while a fraction of mesenchymal neoplastic cells also express FAP in patient tumors. Since oncolytic viruses can kill cancer cells and simultaneously modulate the tumor microenvironment by impacting non-neoplastic populations such as immune cells and tumor vasculature, we further investigated the ability of oncolytic viruses to target GBM-associated stromal cells. An oncolytic adenovirus, ICOVIR15, carrying ∆24-E1A and an RGD-fiber, infects and depletes FAP+ pericytes as well as GBM cells in murine GBM. Our study thus identifies FAP+/PDGFRβ+ pericytes as a major CAF-like stromal cell population in GBM, and highlights the unique property of this oncolytic adenovirus to target both GBM cells and GBM-associated stromal FAP+ cells.
Collapse
|
86
|
Matsumoto T, Chino H, Akiya M, Hashimura M, Yokoi A, Tochimoto M, Nakagawa M, Jiang Z, Saegusa M. Requirements of LEFTY and Nodal overexpression for tumor cell survival under hypoxia in glioblastoma. Mol Carcinog 2020; 59:1409-1419. [PMID: 33111989 DOI: 10.1002/mc.23265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 01/06/2023]
Abstract
Glioblastomas (GBM) contain numerous hypoxic foci associated with a rare fraction of glioma stem cells (GSCs). Left-right determination factor (LEFTY) and Nodal, members of the transforming growth factor β (TGF-β) superfamily, have glycogen synthase kinase 3β (GSK-3β) phosphorylation motifs and are linked with stemness in human malignancies. Herein, we investigated the roles of LEFTY and Nodal in GBM hypoxic foci. In clinical samples, significantly higher expression of LEFTY, Nodal, phospho (p) GSK-3β, pSmad2, and Nestin, as well as higher apoptotic and lower proliferation rates, were observed in nonpseudopalisading (non-Ps) perinecrotic lesions as compared to Ps and non-necrotic tumor lesions, with a positive correlation between LEFTY, Nodal, pGSK-3β, or pSmad2 scores. In KS-1, a GBM cell line that lacks endogenous Nodal expression, treatment with the hypoxic mimetic CoCl2 increased LEFTY, pGSK-3β, and pSmad2 levels, but decreased pAkt levels. Moreover, the promoter for LEFTY, but not Nodal, was activated by Smad2 or TGF-β1, suggesting that overexpression of LEFTY and Nodal may be due to Akt-independent GSK-3β inactivation, with or without cooperation of the TGF-β1/Smad2 axis. LEFTY and Nodal overexpression increased proliferation rates and reduced susceptibility to CoCl2 -induced apoptosis, and increased the expression of epithelial-mesenchymal transition (EMT)/GSC-related markers. An increased ALDH1high population and more efficient spheroid formation was also observed in LEFTY-overexpressing cells. These findings suggest that LEFTY and Nodal may contribute to cell survival in non-Ps GBM perinecrotic lesions, leading to alterations in apoptosis, proliferation, or EMT/GCS features.
Collapse
Affiliation(s)
- Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hiromi Chino
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masashi Akiya
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Miki Hashimura
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Ako Yokoi
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Masataka Tochimoto
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Mayu Nakagawa
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Zesong Jiang
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
87
|
Marayati R, Bownes LV, Stafman LL, Williams AP, Quinn CH, Atigadda V, Aye JM, Stewart JE, Yoon KJ, Beierle EA. 9-cis-UAB30, a novel rexinoid agonist, decreases tumorigenicity and cancer cell stemness of human neuroblastoma patient-derived xenografts. Transl Oncol 2020; 14:100893. [PMID: 33010553 PMCID: PMC7530346 DOI: 10.1016/j.tranon.2020.100893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/28/2022] Open
Abstract
Retinoic acid (RA) therapy has been utilized as maintenance therapy for high-risk neuroblastoma, but over half of patients treated with RA relapse. Neuroblastoma stem cell-like cancer cells (SCLCCs) are a subpopulation of cells characterized by the expression of the cell surface marker CD133 and are hypothesized to contribute to drug resistance and disease relapse. A novel rexinoid compound, 9-cis-UAB30 (UAB30), was developed having the same anti-tumor effects as RA but a more favorable toxicity profile. In the current study, we investigated the efficacy of UAB30 in neuroblastoma patient-derived xenografts (PDX). Two PDXs, COA3 and COA6, were utilized and alterations in the malignant phenotype were assessed following treatment with RA or UAB30. UAB30 significantly decreased proliferation, viability, and motility of both PDXs. UAB30 induced cell-cycle arrest as demonstrated by the significant increase in percentage of cells in G1 (COA6: 33.7 ± 0.7 vs. 43.3 ± 0.7%, control vs. UAB30) and decrease in percentage of cells in S phase (COA6: 44.7 ± 1.2 vs. 38.6 ± 1%, control vs. UAB30). UAB30 led to differentiation of PDX cells, as evidenced by the increase in neurite outgrowth and mRNA abundance of differentiation markers. CD133 expression was decreased by 40% in COA6 cells after UAB30. The ability to form tumorspheres and mRNA abundance of known stemness markers were also significantly decreased following treatment with UAB30, further indicating decreased cancer cell stemness. These results provide evidence that UAB30 decreased tumorigenicity and cancer cell stemness in neuroblastoma PDXs, warranting further exploration as therapy for high-risk neuroblastoma.
Collapse
Affiliation(s)
- Raoud Marayati
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Laura V Bownes
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Laura L Stafman
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Adele P Williams
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Colin H Quinn
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Venkatram Atigadda
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jamie M Aye
- Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jerry E Stewart
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Karina J Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Elizabeth A Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
88
|
Chen RH, Xiao L, Zhang RZ, Wang SY, Li Y. Dedifferentiation of human epidermal melanocytes in vitro by long-term trypsinization. Cell Tissue Bank 2020; 22:67-75. [PMID: 32978700 DOI: 10.1007/s10561-020-09866-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/26/2020] [Accepted: 09/18/2020] [Indexed: 11/26/2022]
Abstract
Human epidermal melanocytes can be induced to form melanocyte spheroids and revert to immature characteristics by long-term trypsinization (LTT). To further explore the biological characteristics of melanocytes after LTT and to study the underlying mechanism. Melanocytes were subjected to long-term (2 h) trypsinization in this study, after which their viability, proliferation and autophagy were characterized. The expression of melanocyte markers [human melanoma black45 (HMB45), tyrosinase (TYR) and Nestin] was detected and relative expression levels of mRNAs encoding TYR, Nestin, c-Kit and microphthalmia-associated transcription factor (MITF) were determined. After LTT, more short spindle-shaped melanocytes appeared and viability assays demonstrated that most melanocytes survived that treatment but had decreased proliferation rates compared to the untreated controls. There was a significant increase in autophagy of melanocytes after LTT and the expression of TYR was decreased compared with untreated control melanocytes. There were no significant differences in the expression of HMB45 or Nestin between the two groups. Compared with untreated melanocytes, levels of message ribonucleic acid (mRNAs) encoding TYR, c-Kit and MITF were decreased after LTT, while Nestin mRNA levels were increased. These results clarified that Long-term treatment with trypsin causes the dedifferentiation of mature epidermal melanocytes in vitro.
Collapse
Affiliation(s)
- Ren-He Chen
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, 185 Juqian Road, Changzhou, 213003, China
| | - Li Xiao
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, 185 Juqian Road, Changzhou, 213003, China
| | - Ru-Zhi Zhang
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, 185 Juqian Road, Changzhou, 213003, China.
| | - Sheng-Yi Wang
- Department of Dermatology, Xuzhou Central Hospital, Xuzhou, 221000, China
| | - Yue Li
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, 185 Juqian Road, Changzhou, 213003, China
| |
Collapse
|
89
|
Schulte am Esch J, Windmöller BA, Hanewinkel J, Storm J, Förster C, Wilkens L, Krüger M, Kaltschmidt B, Kaltschmidt C. Isolation and Characterization of Two Novel Colorectal Cancer Cell Lines, Containing a Subpopulation with Potential Stem-Like Properties: Treatment Options by MYC/NMYC Inhibition. Cancers (Basel) 2020; 12:cancers12092582. [PMID: 32927768 PMCID: PMC7564713 DOI: 10.3390/cancers12092582] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary The aim of this study was to gain a better understanding of cancer stem cells, which are a small subpopulation of tumor cells with high plasticity driving tumor growth and metastasis. Here we isolated two novel colorectal cancer cell lines originating from a rectal neuroendocrine carcinoma and a colorectal adenocarcinoma, depicting stem-like properties. These in vitro models offer the possibility to evaluate pathophysiological mechanisms in order to develop tailored therapeutic strategies for distinct colorectal malignancies. Investigations revealed gene copy number gain of the N-myc proto-oncogene for both. Accordingly, inhibition of the protein–protein interaction of myc and N-myc proto-oncogenes with the myc-associated factor X utilizing small molecule KJ-Pyr-9, exhibited a significant reduction in survival of both cell lines by the induction of apoptosis. Consequently, the blockage of these interactions may serve as a possible treatment strategy for colorectal cancer cell lines with gene copy number gain of the N-myc proto-oncogene. Abstract Cancer stem cells (CSC) are crucial mediators of cancer relapse. Here, we isolated two primary human colorectal cancer cell lines derived from a rectal neuroendocrine carcinoma (BKZ-2) and a colorectal adenocarcinoma (BKZ-3), both containing subpopulations with potential stem-like properties. Protein expression of CSC-markers prominin-1 and CD44 antigen was significantly higher for BKZ-2 and BKZ-3 in comparison to well-established colon carcinoma cell lines. High sphere-formation capacity further confirmed the existence of a subpopulation with potential stem-like phenotype. Epithelial–mesenchymal transition markers as well as immune checkpoint ligands were expressed more pronounced in BKZ-2. Both cell populations demonstrated N-myc proto-oncogene (NMYC) copy number gain. Myc proto-oncogene (MYC)/NMYC activity inhibitor all-trans retinoic acid (ATRA) significantly reduced the number of tumor spheres for both and the volume of BKZ-2 spheres. In contrast, the sphere volume of ATRA-treated BKZ-3 was increased, and only BKZ-2 cell proliferation was reduced in monolayer culture. Treatment with KJ-Pyr-9, a specific inhibitor of MYC/NMYC-myc-associated factor X interaction, decreased survival by the induction of apoptosis of both. In summary, here, we present the novel colorectal cancer cell lines BKZ-2 and BKZ-3 as promising cellular in vitro models for colorectal carcinomas and identify the MYC/NMYC molecular pathway involved in CSC-induced carcinogenesis with relevant therapeutic potential.
Collapse
Affiliation(s)
- Jan Schulte am Esch
- Department of General and Visceral Surgery, Protestant Hospital of Bethel Foundation, 33611 Bielefeld, Germany;
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
| | - Beatrice Ariane Windmöller
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
- Correspondence: ; Tel.: +49-0521-106-5629
| | - Johannes Hanewinkel
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
| | - Jonathan Storm
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
| | - Christine Förster
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Institute of Pathology, KRH Hospital Nordstadt, affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany
| | - Ludwig Wilkens
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Institute of Pathology, KRH Hospital Nordstadt, affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany
| | - Martin Krüger
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Internal Medicine and Gastroenterology, Protestant Hospital of Bethel Foundation, 33611 Bielefeld, Germany
| | - Barbara Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
- Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany
| | - Christian Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
| |
Collapse
|
90
|
Ye J, Gong P. NGF-CS/HA-coating composite titanium facilitates the differentiation of bone marrow mesenchymal stem cells into osteoblast and neural cells. Biochem Biophys Res Commun 2020; 531:290-296. [PMID: 32800542 DOI: 10.1016/j.bbrc.2020.06.158] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022]
Abstract
Dental implant is the most effective way to repair the defect or absence of dentition. Currently, the modification in titanium surface properties has become a hot topic in the study of oral implantology. However, more suitable titanium surface coating still needs to be further explored. We prepared the nerve growth factor-chondroitin sulfate/hydroxyapatite (NGF-CS/HA)-coating composite titanium by modified biomimetic method. We also observed the surface morphology, thickness, surface adhesion and component analysis of NGF-CS/HA-coating composite titanium by scanning electron microscope, and the release of NGF was also identified via ELISA assay. Besides, the identification of bone marrow mesenchymal stem cells (BMSCs) was conducted through alizarin red staining, oil red O staining and fluorescence detection. and the osteogenesis differentiation and neuronal differentiation-related genes were determined by RT-qPCR assay. The surface of NGF-CS/HA coating with the 65.4 ± 6.4 μm thickness presented a porous network, and the main components of NGF-CS/HA coating were Ti and HA, and maintained the activity and release of NGF. Besides, we successfully obtained and identified BMSCs, and proved that NGF-CS/HA-coating composite titanium could notably upregulated the expression levels of the osteogenesis differentiation and neuronal differentiation-related genes and proteins in BMSCs. In conclusion, NGF-CS/HA-coating composite titanium has significant promoting effects on the differentiation of BMSCs into osteoblast and neural cells.
Collapse
Affiliation(s)
- Jun Ye
- Department of Prosthodontics, School and Hospital of Stomatology, Tongji University and Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, PR China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, Department of Oral Implant, West China School of Stomatology, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
91
|
Sone K, Maeno K, Masaki A, Kunii E, Takakuwa O, Kagawa Y, Takeuchi A, Fukuda S, Uemura T, Fukumitsu K, Kanemitsu Y, Ohkubo H, Takemura M, Ito Y, Oguri T, Inagaki H, Niimi A. Nestin Expression Affects Resistance to Chemotherapy and Clinical Outcome in Small Cell Lung Cancer. Front Oncol 2020; 10:1367. [PMID: 32903755 PMCID: PMC7438916 DOI: 10.3389/fonc.2020.01367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 06/29/2020] [Indexed: 12/18/2022] Open
Abstract
Objectives: Small cell lung cancer (SCLC) is an aggressive and highly metastatic lung cancer subtype. Nestin is a member of the intermediate filament family and serves as a potential proliferative and multipotency marker in neural progenitor and stem cells. Aberrant expression of nestin is linked to poor prognosis in different cancers, including non-small cell lung cancer. However, the association between nestin expression and clinicopathological feature or prognosis has remained unclear for SCLC. This study examined whether nestin expression was associated with malignant features and clinical outcomes in SCLC. Materials and Methods: Using previously established Nestin knock-down cells and a newly established Nestin-overexpressing cell line, we examined the relationship between nestin expression and cell proliferation in vitro and in vivo and chemosensitivity. We also analyzed nestin expression in three drug-resistant lung cancer cell lines. Furthermore, we examined samples from 84 SCLC patients (16 patients with surgical resection, and 68 patients with biopsy), and immunohistochemically analyzed nestin expression. Results: Nestin expression correlated positively with cell proliferation, but negatively with chemosensitivity. Nestin expression in drug-resistant cell lines was upregulated compared to their parental cells. Among the 84 SCLC patients, 24 patients (28.6%) showed nestin-positive tumor. Nestin-positive ratio tended to be higher in operated patients than in biopsied patients. Nestin-positive and -negative patients showed no significant differences in response rate (RR) or progression-free survival (PFS) following first-line chemotherapy. However, positive expression of nestin was associated with shorter PFS following second-line chemotherapy (median PFS: nestin-positive, 81 days vs. nestin-negative, 117 days; P = 0.029). Conclusions: Nestin expression may be associated with malignant phenotype and worse outcome in SCLC patients.
Collapse
Affiliation(s)
- Kazuki Sone
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ken Maeno
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ayako Masaki
- Department of Pathology and Molecular Diagnosis, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Eiji Kunii
- Department of Respiratory Medicine, Nagoya City West Medical Center, Nagoya, Japan
| | - Osamu Takakuwa
- Department of Respiratory Medicine, Nagoya City West Medical Center, Nagoya, Japan
| | - Yusuke Kagawa
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akira Takeuchi
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoshi Fukuda
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takehiro Uemura
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kensuke Fukumitsu
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshihiro Kanemitsu
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirotsugu Ohkubo
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masaya Takemura
- Department of Education and Research Center for Community Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yutaka Ito
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tetsuya Oguri
- Department of Education and Research Center for Community Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroshi Inagaki
- Department of Pathology and Molecular Diagnosis, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akio Niimi
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
92
|
Teh DBL, Bansal A, Chai C, Toh TB, Tucker RAJ, Gammad GGL, Yeo Y, Lei Z, Zheng X, Yang F, Ho JS, Bolem N, Wu BC, Gnanasammandhan MK, Hooi L, Dawe GS, Libedinsky C, Ong WY, Halliwell B, Chow EKH, Lim KL, Zhang Y, Kennedy BK. A Flexi-PEGDA Upconversion Implant for Wireless Brain Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001459. [PMID: 32484308 DOI: 10.1002/adma.202001459] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 05/12/2023]
Abstract
Near-infrared (NIR) activatable upconversion nanoparticles (UCNPs) enable wireless-based phototherapies by converting deep-tissue-penetrating NIR to visible light. UCNPs are therefore ideal as wireless transducers for photodynamic therapy (PDT) of deep-sited tumors. However, the retention of unsequestered UCNPs in tissue with minimal options for removal limits their clinical translation. To address this shortcoming, biocompatible UCNPs implants are developed to deliver upconversion photonic properties in a flexible, optical guide design. To enhance its translatability, the UCNPs implant is constructed with an FDA-approved poly(ethylene glycol) diacrylate (PEGDA) core clad with fluorinated ethylene propylene (FEP). The emission spectrum of the UCNPs implant can be tuned to overlap with the absorption spectra of the clinically relevant photosensitizer, 5-aminolevulinic acid (5-ALA). The UCNPs implant can wirelessly transmit upconverted visible light till 8 cm in length and in a bendable manner even when implanted underneath the skin or scalp. With this system, it is demonstrated that NIR-based chronic PDT is achievable in an untethered and noninvasive manner in a mouse xenograft glioblastoma multiforme (GBM) model. It is postulated that such encapsulated UCNPs implants represent a translational shift for wireless deep-tissue phototherapy by enabling sequestration of UCNPs without compromising wireless deep-tissue light delivery.
Collapse
Affiliation(s)
- Daniel Boon Loong Teh
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Akshaya Bansal
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Chou Chai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health, National University of Singapore, Singapore, 117599, Singapore
| | - Robert Alan Jappy Tucker
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Gil Gerald Lasam Gammad
- The N.1 Institute for Health, National University of Singapore, Singapore, 117599, Singapore
| | - Yanzhuang Yeo
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Zhendong Lei
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, 117456, Singapore
| | - Xiang Zheng
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, 117456, Singapore
| | - Fengyuan Yang
- Department of Electrical & Computer Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - John S Ho
- The N.1 Institute for Health, National University of Singapore, Singapore, 117599, Singapore
- Department of Electrical & Computer Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Nagarjun Bolem
- Division of Neurosurgery, National University Hospital, Singapore, 119228, Singapore
| | - Bing Cheng Wu
- Department of Pathology, National University Hospital, Singapore, 119228, Singapore
| | - Muthu Kumar Gnanasammandhan
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Lissa Hooi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gavin Stewart Dawe
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Camilo Libedinsky
- The N.1 Institute for Health, National University of Singapore, Singapore, 117599, Singapore
- Department of Psychology, Faculty of Arts and Social Sciences, National University of Singapore, Singapore, 117570, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Edward Kai-Hua Chow
- The N.1 Institute for Health, National University of Singapore, Singapore, 117599, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Kah-Leong Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Yong Zhang
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Brian K Kennedy
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- Center for Healthy Ageing, National University Health System, Singapore, 119228, Singapore
| |
Collapse
|
93
|
Cancer Stem Cells in Soft-Tissue Sarcomas. Cells 2020; 9:cells9061449. [PMID: 32532153 PMCID: PMC7349510 DOI: 10.3390/cells9061449] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Soft tissue sarcomas (STS) are a rare group of mesenchymal solid tumors with heterogeneous genetic profiles and clinical features. Systemic chemotherapy is the backbone treatment for advanced STS; however, STS frequently acquire resistance to standard therapies, which highlights the need to improve treatments and identify novel therapeutic targets. Increases in the knowledge of the molecular pathways that drive sarcomas have brought to light different molecular alterations that cause tumor initiation and progression. These findings have triggered a breakthrough of targeted therapies that are being assessed in clinical trials. Cancer stem cells (CSCs) exhibit mesenchymal stem cell (MSC) features and represent a subpopulation of tumor cells that play an important role in tumor progression, chemotherapy resistance, recurrence and metastasis. In fact, CSCs phenotypes have been identified in sarcomas, allied to drug resistance and tumorigenesis. Herein, we will review the published evidence of CSCs in STS, discussing the molecular characteristic of CSCs, the commonly used isolation techniques and the new possibilities of targeting CSCs as a way to improve STS treatment and consequently patient outcome.
Collapse
|
94
|
Hassn Mesrati M, Behrooz AB, Y. Abuhamad A, Syahir A. Understanding Glioblastoma Biomarkers: Knocking a Mountain with a Hammer. Cells 2020; 9:E1236. [PMID: 32429463 PMCID: PMC7291262 DOI: 10.3390/cells9051236] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/18/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
Gliomas are the most frequent and deadly form of human primary brain tumors. Among them, the most common and aggressive type is the high-grade glioblastoma multiforme (GBM), which rapidly grows and renders patients a very poor prognosis. Meanwhile, cancer stem cells (CSCs) have been determined in gliomas and play vital roles in driving tumor growth due to their competency in self-renewal and proliferation. Studies of gliomas have recognized CSCs via specific markers. This review comprehensively examines the current knowledge of the most significant CSCs markers in gliomas in general and in glioblastoma in particular and specifically focuses on their outlook and importance in gliomas CSCs research. We suggest that CSCs should be the superior therapeutic approach by directly targeting the markers. In addition, we highlight the association of these markers with each other in relation to their cascading pathways, and interactions with functional miRNAs, providing the role of the networks axes in glioblastoma signaling pathways.
Collapse
Affiliation(s)
| | | | | | - Amir Syahir
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia; (M.H.M.); (A.B.B.); (A.Y.A.)
| |
Collapse
|
95
|
Clinicopathological and prognostic significance of nestin expression in patients with breast cancer: a systematic review and meta-analysis. Cancer Cell Int 2020; 20:169. [PMID: 32467665 PMCID: PMC7227264 DOI: 10.1186/s12935-020-01252-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/10/2020] [Indexed: 12/21/2022] Open
Abstract
Background Nestin has been revealed to promote tumorigenesis, progression, metastasis, and angiogenesis of breast cancer. Although the prognostic and clinicopathological impact of nestin expression on breast cancer patients has been assessed in several independent studies, their results remained conflicting. Therefore, we performed this meta-analysis to elucidate the prognostic and clinicopathological association of nestin expression with breast cancer. Methods A comprehensive literature search was performed in the electronic databases PubMed, EMBASE, Web of Science, the Cochrane Library, China National Knowledge Infrastructure (CNKI), and the Wangfang Data. The statistical analysis was conducted using Stata 15.0 and Review Manager 5.3. Results A total of 15 studies with 6066 breast cancer patients were included in this meta-analysis. Pooled results indicated that positive expression of nestin was significantly associated with reduced breast cancer-specific survival (BCSS, univariate analysis, HR = 2.11, 95% CI [1.79, 2.49], P < 0.00001; multivariate analysis, HR = 1.30, 95% CI [1.06, 1.60], P = 0.01), worse overall survival (OS, univariate analysis, HR = 1.88, 95% CI [1.31, 2.71], P = 0.0007; multivariate analysis, HR = 1.89, 95% CI [1.34, 2.67], P = 0.0003) and poorer recurrence-free survival (univariate analysis, HR = 2.60, 95% CI [1.52, 4.46], P = 0.0005), but not with distant metastasis-free survival in univariate analysis (P > 0.05). In addition, increased nestin expression was correlated with younger age, higher tumor grade, larger tumor size, positive blood vessel invasion and high vascular proliferation index, but not with lymph node metastasis or lymph vessel invasion. Nestin was preferentially expressed in invasive ductal carcinoma, triple-negative breast cancer and basal-like subtypes. Nestin expression was inversely associated with the expression of ER and PR, but not with HER-2. Conversely, nestin expression was positively correlated with the expression of basal-like markers CK5, P-cadherin and EGFR. Moreover, nestin expression was strongly associated with the presence of five basal-like profiles (BLP1-5). Conclusions This meta-analysis revealed the prognostic value and clinicopathological significance of nestin expression in breast cancer. Nestin is an independent prognostic factor for worse BCSS and OS of breast cancer patients. Nestin is also a valuable biomarker for unfavorable clinicopathological features and tumor angiogenesis of breast cancer. Therefore, nestin is a promising therapeutic target for malignant breast cancer, especially for TNBC and basal-like phenotype.
Collapse
|
96
|
Farrokhfar S, Tiraihi T, Movahedin M, Azizi H. Differential gene expression by lithium chloride induction of adipose-derived stem cells into neural phenotype cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:544-550. [PMID: 32489570 PMCID: PMC7239415 DOI: 10.22038/ijbms.2020.41582.9820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/12/2019] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Adipose-derived stem cells (ADSCs), with suitable and easy access, are multipotential cells that have the ability for differentiation into other mesodermal and transdifferentiate into neural phenotype cells. In this study, Lithium chloride (LiCl) was used for in vitro transdifferentiation of rat ADSCs into neuron-like cells (NLCs). MATERIALS AND METHODS ADSCs were isolated from the rats' perinephric region using Dulbecco΄s Modified Eagle΄s Medium (DMEM) with Fetal Bovine Serum (FBS), cultured for 3 passages, characterized by flowcytometry and differentiation into adipogenic and osteogenic phenotypes. The ADSCs were exposed to 0.1, 0.5, 1, 1.5, 2, 5, and 10 millimolar (mM) LiCl without serum for 24 hr. The optimum dose of LiCl was selected according the maximum viability of cells. The expression of neurofilament light chain (NfL), neurofilament high chain (NfH), and nestin was evaluated by immunocytochemistry. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to evaluate the amount of synaptophysin, neurogenin-1, neuroD1, NfL, NfH, and nestin genes' expression in ADSCs and NLCs. RESULTS The optimum dose of LiCl was 1 mM in 24 hr. The transdifferentiated ADSCs showed cytoplasmic extension with synapse-like formation. Synaptophysin, neurogenin-1, neuroD1, NfL, NfH, and nestin genes were significantly expressed more in NLCs than in ADSCs. CONCLUSION LiCl can induce ADSCs into neural phenotype cells with higher expression of neural and neuronal genes.
Collapse
Affiliation(s)
- Samaneh Farrokhfar
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Taki Tiraihi
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, P.O.BOX.14115-331 Tehran, Iran
| | - Mansoureh Movahedin
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, P.O.BOX.14115-331 Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, P.O.BOX.14115-331 Tehran, Iran
| |
Collapse
|
97
|
Bott CJ, Winckler B. Intermediate filaments in developing neurons: Beyond structure. Cytoskeleton (Hoboken) 2020; 77:110-128. [PMID: 31970897 DOI: 10.1002/cm.21597] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/20/2022]
Abstract
Neuronal development relies on a highly choreographed progression of dynamic cellular processes by which newborn neurons migrate, extend axons and dendrites, innervate their targets, and make functional synapses. Many of these dynamic processes require coordinated changes in morphology, powered by the cell's cytoskeleton. Intermediate filaments (IFs) are the third major cytoskeletal elements in vertebrate cells, but are rarely considered when it comes to understanding axon and dendrite growth, pathfinding and synapse formation. In this review, we first introduce the many new and exciting concepts of IF function, discovered mostly in non-neuronal cells. These roles include dynamic rearrangements, crosstalk with microtubules and actin filaments, mechano-sensing and -transduction, and regulation of signaling cascades. We then discuss the understudied roles of neuronally expressed IFs, with a particular focus on IFs expressed during development, such as nestin, vimentin and α-internexin. Lastly, we illustrate how signaling modulation by the unconventional IF nestin shapes neuronal morphogenesis in unexpected and novel ways. Even though the first IF knockout mice were made over 20 years ago, the study of the cell biological functions of IFs in the brain still has much room for exciting new discoveries.
Collapse
Affiliation(s)
- Christopher J Bott
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
98
|
Zajac J, Novohradsky V, Markova L, Brabec V, Kasparkova J. Platinum (IV) Derivatives with Cinnamate Axial Ligands as Potent Agents Against Both Differentiated and Tumorigenic Cancer Stem Rhabdomyosarcoma Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201913996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Juraj Zajac
- Czech Academy of Sciences Institute of Biophysics Kralovoposlka 135 CZ-61265 Brno Czech Republic
| | - Vojtech Novohradsky
- Czech Academy of Sciences Institute of Biophysics Kralovoposlka 135 CZ-61265 Brno Czech Republic
| | - Lenka Markova
- Czech Academy of Sciences Institute of Biophysics Kralovoposlka 135 CZ-61265 Brno Czech Republic
| | - Viktor Brabec
- Czech Academy of Sciences Institute of Biophysics Kralovoposlka 135 CZ-61265 Brno Czech Republic
| | - Jana Kasparkova
- Czech Academy of Sciences Institute of Biophysics Kralovoposlka 135 CZ-61265 Brno Czech Republic
| |
Collapse
|
99
|
Zajac J, Novohradsky V, Markova L, Brabec V, Kasparkova J. Platinum (IV) Derivatives with Cinnamate Axial Ligands as Potent Agents Against Both Differentiated and Tumorigenic Cancer Stem Rhabdomyosarcoma Cells. Angew Chem Int Ed Engl 2020; 59:3329-3335. [PMID: 31802607 DOI: 10.1002/anie.201913996] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Indexed: 12/15/2022]
Abstract
To design an anticancer drug capable of inhibiting not only the proliferation of the differentiated tumor cells but also reducing the tumorigenic capability of cancer stem cells (CSCs), the new PtIV prodrugs with axial cinnamate ligands were synthesized. We demonstrate their superior antiproliferative activity in monolayer and 3D spheroid antiproliferative activity tests using panel of cancer cell lines. An outstanding activity was found against rhabdomyosarcoma cells, one of the most problematic and poorly treatable pediatric tumors. The results also suggest that the released PtII compound inhibits antiproliferative activity of cancer cells by DNA-damage mediated mechanism; the released cinnamic acid can trigger processes leading to differentiation, making the CSCs more sensitive to killing by the platinum part of the complex. PtIV complex with axial cinnamate ligands is the first PtIV prodrug capable of overcoming CSCs resistance and induce death in both CSCs and bulk cancer.
Collapse
Affiliation(s)
- Juraj Zajac
- Czech Academy of Sciences, Institute of Biophysics, Kralovoposlka 135, CZ-61265, Brno, Czech Republic
| | - Vojtech Novohradsky
- Czech Academy of Sciences, Institute of Biophysics, Kralovoposlka 135, CZ-61265, Brno, Czech Republic
| | - Lenka Markova
- Czech Academy of Sciences, Institute of Biophysics, Kralovoposlka 135, CZ-61265, Brno, Czech Republic
| | - Viktor Brabec
- Czech Academy of Sciences, Institute of Biophysics, Kralovoposlka 135, CZ-61265, Brno, Czech Republic
| | - Jana Kasparkova
- Czech Academy of Sciences, Institute of Biophysics, Kralovoposlka 135, CZ-61265, Brno, Czech Republic
| |
Collapse
|
100
|
Tamura R, Morimoto Y, Sato M, Kuranari Y, Oishi Y, Kosugi K, Yoshida K, Toda M. Difference in the hypoxic immunosuppressive microenvironment of patients with neurofibromatosis type 2 schwannomas and sporadic schwannomas. J Neurooncol 2020; 146:265-273. [PMID: 31897926 DOI: 10.1007/s11060-019-03388-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 12/27/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Neurofibromatosis type 2 (NF2) patients uniformly develop multiple schwannomas. The tumor-microenvironment (TME) is associated with hypoxia and consists of immunosuppressive cells, including regulatory T cells (Tregs) and tumor-associated macrophages (TAMs). The hypoxic TME of NF2 schwannomas remains unclear. In addition, no comparative study has investigated immunosuppressive cells in NF2 and sporadic schwannomas. METHODS In 22 NF2 and 21 sporadic schwannomas, we analyzed the immunohistochemistry for Ki-67, hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor receptor 1 (VEGFR1) and VEGFR2, platelet derived growth factor receptor-beta (PDGFR-β), programmed cell death-1 (PD-1)/ programmed cell death ligand-1 (PD-L1), Foxp3, CD163, CD3, and CD8 to assess the immunosuppressive TME. RESULTS Most vessels in sporadic schwannomas exhibited slight or negative VEGFR1 and 2 expressions with pericytes coverage. In contrast, large vessels in NF2 schwannomas exhibited strong VEGFR1 and 2 expressions without pericytes. The number of CD3+, CD8+, and CD163+ cells was significantly higher in NF2 schwannomas than in sporadic ones. The expression of PD-L1 and nestin positive cell ratio was higher in NF2 schwannomas than that in sporadic ones. The number of CD163+ cells, nestin positive cell ratio, and HIF-1α expression were significantly associated with shorter progression-free survival in NF2 schwannomas. CONCLUSIONS This study presents the clinicopathological features of the differences in immunosuppressive cells and the expression of immune checkpoint molecules between NF2 and sporadic schwannomas. Hypoxic TME was first detected in NF2-schwannomas, which was associated with the tumor progression.
Collapse
Affiliation(s)
- Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yukina Morimoto
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Mizuto Sato
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuki Kuranari
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yumiko Oishi
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kenzo Kosugi
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|