1
|
Lv W, Wang Y. Neural Influences on Tumor Progression Within the Central Nervous System. CNS Neurosci Ther 2024; 30:e70097. [PMID: 39469896 PMCID: PMC11519750 DOI: 10.1111/cns.70097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/21/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024] Open
Abstract
For decades, researchers have studied how brain tumors, the immune system, and drugs interact. With the advances in cancer neuroscience, which centers on defining and therapeutically targeting nervous system-cancer interactions, both within the local tumor microenvironment (TME) and on a systemic level, the subtle relationship between neurons and tumors in the central nervous system (CNS) has been deeply studied. Neurons, as the executors of brain functional activities, have been shown to significantly influence the emergence and development of brain tumors, including both primary and metastatic tumors. They engage with tumor cells via chemical or electrical synapses, directly regulating tumors or via intricate coupling networks, and also contribute to the TME through paracrine signaling, secreting proteins that exert regulatory effects. For instance, in a study involving a mouse model of glioblastoma, the authors observed a 42% increase in tumor volume when neuronal activity was stimulated, compared to controls (p < 0.01), indicating a direct correlation between neural activity and tumor growth. These thought-provoking results offer promising new strategies for brain tumor therapies, highlighting the potential of neuronal modulation to curb tumor progression. Future strategies may focus on developing drugs to inhibit or neutralize proteins and other bioactive substances secreted by neurons, break synaptic connections and interactions between infiltrating cells and tumor cells, as well as disrupt electrical coupling within glioma cell networks. By harnessing the insights gained from this research, we aspire to usher in a new era of brain tumor therapies that are both more potent and precise.
Collapse
Affiliation(s)
- Wenhao Lv
- Affiliated Hospital of Hangzhou Normal UniversityHangzhou Normal UniversityHangzhouZhejiangChina
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityHangzhouZhejiangChina
| |
Collapse
|
2
|
Pan Y, Hysinger JD, Yalçın B, Lennon JJ, Byun YG, Raghavan P, Schindler NF, Anastasaki C, Chatterjee J, Ni L, Xu H, Malacon K, Jahan SM, Ivec AE, Aghoghovwia BE, Mount CW, Nagaraja S, Scheaffer S, Attardi LD, Gutmann DH, Monje M. Nf1 mutation disrupts activity-dependent oligodendroglial plasticity and motor learning in mice. Nat Neurosci 2024; 27:1555-1564. [PMID: 38816530 PMCID: PMC11303248 DOI: 10.1038/s41593-024-01654-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 04/18/2024] [Indexed: 06/01/2024]
Abstract
Neurogenetic disorders, such as neurofibromatosis type 1 (NF1), can cause cognitive and motor impairments, traditionally attributed to intrinsic neuronal defects such as disruption of synaptic function. Activity-regulated oligodendroglial plasticity also contributes to cognitive and motor functions by tuning neural circuit dynamics. However, the relevance of oligodendroglial plasticity to neurological dysfunction in NF1 is unclear. Here we explore the contribution of oligodendrocyte progenitor cells (OPCs) to pathological features of the NF1 syndrome in mice. Both male and female littermates (4-24 weeks of age) were used equally in this study. We demonstrate that mice with global or OPC-specific Nf1 heterozygosity exhibit defects in activity-dependent oligodendrogenesis and harbor focal OPC hyperdensities with disrupted homeostatic OPC territorial boundaries. These OPC hyperdensities develop in a cell-intrinsic Nf1 mutation-specific manner due to differential PI3K/AKT activation. OPC-specific Nf1 loss impairs oligodendroglial differentiation and abrogates the normal oligodendroglial response to neuronal activity, leading to impaired motor learning performance. Collectively, these findings show that Nf1 mutation delays oligodendroglial development and disrupts activity-dependent OPC function essential for normal motor learning in mice.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jared D Hysinger
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Belgin Yalçın
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - James J Lennon
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Youkyeong Gloria Byun
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Preethi Raghavan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Nicole F Schindler
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jit Chatterjee
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lijun Ni
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Haojun Xu
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Karen Malacon
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Samin M Jahan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Alexis E Ivec
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Benjamin E Aghoghovwia
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher W Mount
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Surya Nagaraja
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Suzanne Scheaffer
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Laura D Attardi
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
3
|
He Z, Peng Y, Wang D, Yang C, Zhou C, Gong B, Song S, Wang Y. Single-cell transcriptomic analysis identifies downregulated phosphodiesterase 8B as a novel oncogene in IDH-mutant glioma. Front Immunol 2024; 15:1427200. [PMID: 38989284 PMCID: PMC11233524 DOI: 10.3389/fimmu.2024.1427200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/04/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction Glioma, a prevalent and deadly brain tumor, is marked by significant cellular heterogeneity and metabolic alterations. However, the comprehensive cell-of-origin and metabolic landscape in high-grade (Glioblastoma Multiforme, WHO grade IV) and low-grade (Oligoastrocytoma, WHO grade II) gliomas remains elusive. Methods In this study, we undertook single-cell transcriptome sequencing of these glioma grades to elucidate their cellular and metabolic distinctions. Following the identification of cell types, we compared metabolic pathway activities and gene expressions between high-grade and low-grade gliomas. Results Notably, astrocytes and oligodendrocyte progenitor cells (OPCs) exhibited the most substantial differences in both metabolic pathways and gene expression, indicative of their distinct origins. The comprehensive analysis identified the most altered metabolic pathways (MCPs) and genes across all cell types, which were further validated against TCGA and CGGA datasets for clinical relevance. Discussion Crucially, the metabolic enzyme phosphodiesterase 8B (PDE8B) was found to be exclusively expressed and progressively downregulated in astrocytes and OPCs in higher-grade gliomas. This decreased expression identifies PDE8B as a metabolism-related oncogene in IDH-mutant glioma, marking its dual role as both a protective marker for glioma grading and prognosis and as a facilitator in glioma progression.
Collapse
Affiliation(s)
- Zongze He
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Peng
- Department of Academic Journal, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Duo Wang
- Department of Critical Care Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chen Yang
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chengzhi Zhou
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo Gong
- Department of Health Management, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and Institute of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, China
| |
Collapse
|
4
|
Saratsis AM, Knowles T, Petrovic A, Nazarian J. H3K27M mutant glioma: Disease definition and biological underpinnings. Neuro Oncol 2024; 26:S92-S100. [PMID: 37818718 PMCID: PMC11066930 DOI: 10.1093/neuonc/noad164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Indexed: 10/12/2023] Open
Abstract
High-grade glioma (HGG) is the most common cause of cancer death in children and the most common primary central nervous system tumor in adults. While pediatric HGG was once thought to be biologically similar to the adult form of disease, research has shown these malignancies to be significantly molecularly distinct, necessitating distinct approaches to their clinical management. However, emerging data have shown shared molecular events in pediatric and adult HGG including the histone H3K27M mutation. This somatic missense mutation occurs in genes encoding one of two isoforms of the Histone H3 protein, H3F3A (H3.3), or HIST1H3B (H3.1), and is detected in up to 80% of pediatric diffuse midline gliomas and in up to 60% of adult diffuse gliomas. Importantly, the H3K27M mutation is associated with poorer overall survival and response to therapy compared to patients with H3 wild-type tumors. Here, we review the clinical features and biological underpinnings of pediatric and adult H3K27M mutant glioma, offering a groundwork for understanding current research and clinical approaches for the care of patients suffering with this challenging disease.
Collapse
Affiliation(s)
| | | | - Antonela Petrovic
- DMG Research Center, Department of Oncology, University Children’s Hospital, University of Zürich, Zürich, Switzerland
| | - Javad Nazarian
- Research Center for Genetic Medicine, Children’s National Health System, Washington, District of Columbia, USA
- Brain Tumor Institute, Children’s National Health System, Washington, District of Columbia, USA
- DMG Research Center, Department of Pediatrics, University Children’s Hospital, University of Zurich, Zürich, Switzerland
| |
Collapse
|
5
|
de Blank P, Nishiyama A, López-Juárez A. A new era for myelin research in Neurofibromatosis type 1. Glia 2023; 71:2701-2719. [PMID: 37382486 PMCID: PMC10592420 DOI: 10.1002/glia.24432] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 06/30/2023]
Abstract
Evidence for myelin regulating higher-order brain function and disease is rapidly accumulating; however, defining cellular/molecular mechanisms remains challenging partially due to the dynamic brain physiology involving deep changes during development, aging, and in response to learning and disease. Furthermore, as the etiology of most neurological conditions remains obscure, most research models focus on mimicking symptoms, which limits understanding of their molecular onset and progression. Studying diseases caused by single gene mutations represents an opportunity to understand brain dys/function, including those regulated by myelin. Here, we discuss known and potential repercussions of abnormal central myelin on the neuropathophysiology of Neurofibromatosis Type 1 (NF1). Most patients with this monogenic disease present with neurological symptoms diverse in kind, severity, and onset/decline, including learning disabilities, autism spectrum disorders, attention deficit and hyperactivity disorder, motor coordination issues, and increased risk for depression and dementia. Coincidentally, most NF1 patients show diverse white matter/myelin abnormalities. Although myelin-behavior links were proposed decades ago, no solid data can prove or refute this idea yet. A recent upsurge in myelin biology understanding and research/therapeutic tools provides opportunities to address this debate. As precision medicine moves forward, an integrative understanding of all cell types disrupted in neurological conditions becomes a priority. Hence, this review aims to serve as a bridge between fundamental cellular/molecular myelin biology and clinical research in NF1.
Collapse
Affiliation(s)
- Peter de Blank
- Department of Pediatrics, The Cure Starts Now Brain Tumor Center, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | - Alejandro López-Juárez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| |
Collapse
|
6
|
Zhang Y, Zeng J, Xu B. Phenotypic analysis with trans-recombination-based genetic mosaic models. J Biol Chem 2023; 299:105265. [PMID: 37734556 PMCID: PMC10587715 DOI: 10.1016/j.jbc.2023.105265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/01/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
Mosaicism refers to the presence of genetically distinct cell populations in an individual derived from a single zygote, which occurs during the process of development, aging, and genetic diseases. To date, a variety of genetically engineered mosaic analysis models have been established and widely used in studying gene function at exceptional cellular and spatiotemporal resolution, leading to many ground-breaking discoveries. Mosaic analysis with a repressible cellular marker and mosaic analysis with double markers are genetic mosaic analysis models based on trans-recombination. These models can generate sibling cells of distinct genotypes in the same animal and simultaneously label them with different colors. As a result, they offer a powerful approach for lineage tracing and studying the behavior of individual mutant cells in a wildtype environment, which is particularly useful for determining whether gene function is cell autonomous or nonautonomous. Here, we present a comprehensive review on the establishment and applications of mosaic analysis with a repressible cellular marker and mosaic analysis with double marker systems. Leveraging the capabilities of these mosaic models for phenotypic analysis will facilitate new discoveries on the cellular and molecular mechanisms of development and disease.
Collapse
Affiliation(s)
- Yu Zhang
- School of Life Sciences, Nantong University, Nantong, Jiangsu, China
| | - Jianhao Zeng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Bing Xu
- School of Life Sciences, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
7
|
Shah AH, Rivas SR, Doucet-O’Hare TT, Govindarajan V, DeMarino C, Wang T, Ampie L, Zhang Y, Banasavadi-Siddegowda YK, Walbridge S, Maric D, Garcia-Montojo M, Suter RK, Lee MH, Zaghloul KA, Steiner J, Elkahloun AG, Chandar J, Seetharam D, Desgraves J, Li W, Johnson K, Ivan ME, Komotar RJ, Gilbert MR, Heiss JD, Nath A. Human endogenous retrovirus K contributes to a stem cell niche in glioblastoma. J Clin Invest 2023; 133:e167929. [PMID: 37395282 PMCID: PMC10313366 DOI: 10.1172/jci167929] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/19/2023] [Indexed: 07/04/2023] Open
Abstract
Human endogenous retroviruses (HERVs) are ancestral viral relics that constitute nearly 8% of the human genome. Although normally silenced, the most recently integrated provirus HERV-K (HML-2) can be reactivated in certain cancers. Here, we report pathological expression of HML-2 in malignant gliomas in both cerebrospinal fluid and tumor tissue that was associated with a cancer stem cell phenotype and poor outcomes. Using single-cell RNA-Seq, we identified glioblastoma cellular populations with elevated HML-2 transcripts in neural progenitor-like cells (NPC-like) that drive cellular plasticity. Using CRISPR interference, we demonstrate that HML-2 critically maintained glioblastoma stemness and tumorigenesis in both glioblastoma neurospheres and intracranial orthotopic murine models. Additionally, we demonstrate that HML-2 critically regulated embryonic stem cell programs in NPC-derived astroglia and altered their 3D cellular morphology by activating the nuclear transcription factor OCT4, which binds to an HML-2-specific long-terminal repeat (LTR5Hs). Moreover, we discovered that some glioblastoma cells formed immature retroviral virions, and inhibiting HML-2 expression with antiretroviral drugs reduced reverse transcriptase activity in the extracellular compartment, tumor viability, and pluripotency. Our results suggest that HML-2 fundamentally contributes to the glioblastoma stem cell niche. Because persistence of glioblastoma stem cells is considered responsible for treatment resistance and recurrence, HML-2 may serve as a unique therapeutic target.
Collapse
Affiliation(s)
- Ashish H. Shah
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, USA
| | - Sarah R. Rivas
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Tara T. Doucet-O’Hare
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Vaidya Govindarajan
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, USA
| | - Catherine DeMarino
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Tongguang Wang
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Leonel Ampie
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Yong Zhang
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | | | - Stuart Walbridge
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Marta Garcia-Montojo
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Robert K. Suter
- Georgetown University, Bioinformatics Section, Washington, DC, USA
| | - Myoung-Hwa Lee
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Kareem A. Zaghloul
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Joseph Steiner
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Abdel G. Elkahloun
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Jay Chandar
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, USA
| | - Deepa Seetharam
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, USA
| | - Jelisah Desgraves
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, USA
| | - Wenxue Li
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Kory Johnson
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Michael E. Ivan
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, USA
| | - Ricardo J. Komotar
- University of Miami School of Medicine, Department of Neurosurgery, Miami, Florida, USA
| | - Mark R. Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - John D. Heiss
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| | - Avindra Nath
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, Maryland, USA
| |
Collapse
|
8
|
Li B, Li H, Zhang L, Ren T, Meng J. Expression analysis of human glioma susceptibility gene and P53 in human glioma and its clinical significance based on bioinformatics. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:53. [PMID: 36819578 PMCID: PMC9929792 DOI: 10.21037/atm-22-5646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023]
Abstract
Background The exact mechanism of glioblastoma multiforme (GBM) remains unclear. This study was to clarify the expression of P53 in glioma and its molecular mechanism, and to explore the possibility of P53 as a potential therapeutic target of glioma and its clinical application value, so as to provide a new theoretical basis for the treatment of glioma. Methods Firstly, a dataset was established to analyze the expression of P53 in different stages of glioma and its relationship with prognosis by using The Cancer Genome Atlas (TCGA) database, RNA-seq data, and survival data of glioma and normal control samples in gene expression profiling and interactive analysis (GEPIA). The genes co-expressed with P53 were screened out, their differential expression between glioma and normal control group was analyzed, and their functions were analyzed by enrichment analysis. The TGGA database was used for data verification and analysis. The correlation between P53 expression and clinicopathological parameters was analyzed. Kaplan-Meier survival analysis was used to analyze the relationship between P53 expression and overall survival (OS) and progression-free survival (PFS) of glioma patients, and Cox regression analysis was used to analyze the independent factors affecting OS and PFS of glioma patients. Results The results of TCGA data analysis were as follows: The expression level of P53 was different from that of different stages of glioma, namely, the expression level of P53 between grade II and grade III, grade III and grade IV, and grade II and grade IV were significantly different (P<0.05). The results of P53 gene-related survival analysis showed that KNL1 high expression and low expression were significantly different in OS, and the high expression group was associated with poor prognosis (P<0.05). Conclusions The P53 expression can be an effective biological indicator of poor prognosis of glioma.
Collapse
Affiliation(s)
- Baiyu Li
- Department of Neurology Care Ward, Gansu Provincial Hospital, Lanzhou, China
| | - Hang Li
- Department of Geriatrics, Chengdu Eighth People's Hospital (Geriatric Hospital of Chengdu Medical College), Chengdu, China
| | - Linghui Zhang
- Department of Internal Medicine, Department of Clinical Medicine, Shijiazhuang Medical College, Shijiazhuang, China
| | - Taowen Ren
- Department of Neurology Care Ward, Gansu Provincial Hospital, Lanzhou, China
| | - Jie Meng
- Department of Psychiatry, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
9
|
Yamauchi Y, Matsukura H, Motone K, Ueda M, Aoki W. Evaluation of a library of loxP variants with a wide range of recombination efficiencies by Cre. PLoS One 2022; 17:e0276657. [PMID: 36269789 PMCID: PMC9586403 DOI: 10.1371/journal.pone.0276657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/11/2022] [Indexed: 11/18/2022] Open
Abstract
Sparse labeling of individual cells is an important approach in neuroscience and many other fields of research. Various methods have been developed to sparsely label only a small population of cells; however, there is no simple and reproducible strategy for managing the probability of sparse labeling at desired levels. Here, we aimed to develop a novel methodology based on the Cre-lox system to regulate sparseness at desired levels, and we purely analyzed cleavage efficiencies of loxP mutants by Cre. We hypothesized that mutations in the loxP sequence reduce the recognition efficiency by Cre, which enables the regulation of the sparseness level of gene expression. In this research, we mutagenized the loxP sequence and analyzed a library of loxP variants. We evaluated more than 1000 mutant loxP sequences, including mutants with reduced excision efficiencies by Cre ranging from 0.51% to 59%. This result suggests that these mutant loxP sequences can be useful in regulating the sparseness of genetic labeling at desired levels.
Collapse
Affiliation(s)
- Yuji Yamauchi
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, Japan
- Japan Society for the Promotion of Science, Sakyo-ku, Kyoto, Japan
| | - Hidenori Matsukura
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Keisuke Motone
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, Washington, United States of America
| | - Mitsuyoshi Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Wataru Aoki
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo-ku, Kyoto, Japan
- * E-mail:
| |
Collapse
|
10
|
Chalcone 9X Contributed to Repressing Glioma Cell Growth and Migration and Inducing Cell Apoptosis by Reducing FOXM1 Expression In Vitro and Repressing Tumor Growth In Vivo. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8638085. [PMID: 35978634 PMCID: PMC9377910 DOI: 10.1155/2022/8638085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/30/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022]
Abstract
Objective. Natural and synthetic chalcones played roles in inflammation and cancers. Chalcone 9X was an aromatic ketone that was found to inhibit cell growth of hepatic cancer and lung cancer cells. In this study, we wanted to investigate the functions of Chalcone 9X in glioma. Materials and Methods. Chemical Chalcone 9X was added in human glioma cell lines (U87 and T98G cells) and normal astrocyte cell lines (HA1800) with various concentrations (0 μmol/L, 20 μmol/L, 50 μmol/L, and 100 μmol/L). CCK-8 assay was used to measure cell viability. Flow cytometric assay was used to measure cell apoptotic rates. Wound healing assay and transwell assay were used to measure cell invasion. RT-PCR was used to detect relative mRNA expressions, and the protein expressions were detected by western blot (WB) and immunohistochemical staining (IHC). Finally, nude mouse xenograft assay was performed to prove the effects of Chalcone 9X in vivo. Results. Results revealed that Chalcone 9X treatment suppressed cell viability and cell migration capacity; it could also induce cell apoptosis in U87 and T98G cells with dose dependence. However, it had little cytotoxicity to normal astrocyte HA1800 cells. Moreover, Chalcone 9X treatment could repress the mRNA and protein expressions of FOXM1 in human glioma cell lines, which was an oncogene that could promote the progression and malignancy of glioma. In addition, FOXM1 overexpression dismissed the Chalcone 9X effects on cell proliferation, apoptosis, and migration in human glioma cell lines. Finally, in vivo assay showed that Chalcone 9X treatment repressed the expression of FOXM1, which inhibited the tumor growth of a xenograft model injected with U87 in nude mice. Conclusions. In all, we found that Chalcone 9X could suppress cell proliferation and migration and induce cell apoptosis in human glioma cells, while it has little cytotoxicity to normal astrocyte cells. Therefore, we uncovered a novel way that Chalcone 9X could inhibit FOXM1 expression and repress the progression and biofunctions of glioma cells, which might be a potential therapeutic drug for treating human glioma.
Collapse
|
11
|
Liu R, Jia Y, Guo P, Jiang W, Bai R, Liu C. In Vivo Clonal Analysis Reveals Development Heterogeneity of Oligodendrocyte Precursor Cells Derived from Distinct Germinal Zones. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102274. [PMID: 34396711 PMCID: PMC8529438 DOI: 10.1002/advs.202102274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Indexed: 05/14/2023]
Abstract
Mounting evidence supports that oligodendrocyte precursor cells (OPCs) play important roles in maintaining the integrity of normal brains, and that their dysfunction is the etiology of numerous severe neurological diseases. OPCs exhibit diverse heterogeneity in the adult brain, and distinct germinal zones of the embryonic brain contribute to OPC genesis. However, it remains obscure whether developmental origins shape OPC heterogeneity in the adult brain. Here, an in vivo clonal analysis approach is developed to address this. By combining OPC-specific transgenes, in utero electroporation, and the PiggyBac transposon system, the lineages of individual neonatal OPCs derived from either dorsal or ventral embryonic germinal zones are traced, and the landscape of their trajectories is comprehensively described throughout development. Surprisingly, despite behaving indistinguishably in the brain before weaning, dorsally derived OPCs continuously expand throughout life, but ventrally derived OPCs eventually diminish. Importantly, clonal analysis supports the existence of an intrinsic cellular "clock" to control OPC expansion. Moreover, knockout of NF1 could circumvent the distinction of ventrally derived OPCs in the adult brain. Together, this work shows the importance of in vivo clonal analysis in studying stem/progenitor cell heterogeneity, and reveals that developmental origins play a role in determining OPC fate.
Collapse
Affiliation(s)
- Rui Liu
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiang310058P.R. China
- Department of Pathology and PathophysiologyZhejiang University School of MedicineHangzhouZhejiang310058P.R. China
- NHC and CAMS Key Laboratory of Medical NeurobiologyMOE Frontier Science Center for Brain Science and Brain‐machine IntegrationSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouZhejiang310058P.R. China
- School of MedicineZhejiang University City CollegeHangzhouZhejiang310015P.R. China
| | - Yinhang Jia
- Department of Physical Medicine and Rehabilitation of The Affiliated Sir Run Shumen Shaw HospitalInterdisciplinary Institute of Neuroscience and TechnologyZhejiang University School of MedicineHangzhouZhejiang310029P.R. China
| | - Peng Guo
- Department of Pathology and PathophysiologyZhejiang University School of MedicineHangzhouZhejiang310058P.R. China
| | - Wenhong Jiang
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiang310058P.R. China
- Department of Pathology and PathophysiologyZhejiang University School of MedicineHangzhouZhejiang310058P.R. China
- NHC and CAMS Key Laboratory of Medical NeurobiologyMOE Frontier Science Center for Brain Science and Brain‐machine IntegrationSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouZhejiang310058P.R. China
- School of MedicineZhejiang University City CollegeHangzhouZhejiang310015P.R. China
| | - Ruiliang Bai
- Department of Physical Medicine and Rehabilitation of The Affiliated Sir Run Shumen Shaw HospitalInterdisciplinary Institute of Neuroscience and TechnologyZhejiang University School of MedicineHangzhouZhejiang310029P.R. China
| | - Chong Liu
- Department of Neurobiology and Department of Neurosurgery of Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiang310058P.R. China
- Department of Pathology and PathophysiologyZhejiang University School of MedicineHangzhouZhejiang310058P.R. China
- NHC and CAMS Key Laboratory of Medical NeurobiologyMOE Frontier Science Center for Brain Science and Brain‐machine IntegrationSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouZhejiang310058P.R. China
- School of MedicineZhejiang University City CollegeHangzhouZhejiang310015P.R. China
| |
Collapse
|
12
|
Contreras X, Amberg N, Davaatseren A, Hansen AH, Sonntag J, Andersen L, Bernthaler T, Streicher C, Heger A, Johnson RL, Schwarz LA, Luo L, Rülicke T, Hippenmeyer S. A genome-wide library of MADM mice for single-cell genetic mosaic analysis. Cell Rep 2021; 35:109274. [PMID: 34161767 PMCID: PMC8317686 DOI: 10.1016/j.celrep.2021.109274] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 04/14/2021] [Accepted: 05/28/2021] [Indexed: 10/21/2022] Open
Abstract
Mosaic analysis with double markers (MADM) offers one approach to visualize and concomitantly manipulate genetically defined cells in mice with single-cell resolution. MADM applications include the analysis of lineage, single-cell morphology and physiology, genomic imprinting phenotypes, and dissection of cell-autonomous gene functions in vivo in health and disease. Yet, MADM can only be applied to <25% of all mouse genes on select chromosomes to date. To overcome this limitation, we generate transgenic mice with knocked-in MADM cassettes near the centromeres of all 19 autosomes and validate their use across organs. With this resource, >96% of the entire mouse genome can now be subjected to single-cell genetic mosaic analysis. Beyond a proof of principle, we apply our MADM library to systematically trace sister chromatid segregation in distinct mitotic cell lineages. We find striking chromosome-specific biases in segregation patterns, reflecting a putative mechanism for the asymmetric segregation of genetic determinants in somatic stem cell division.
Collapse
Affiliation(s)
- Ximena Contreras
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Nicole Amberg
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | | | - Andi H Hansen
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Johanna Sonntag
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Lill Andersen
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Tina Bernthaler
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Carmen Streicher
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Anna Heger
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Randy L Johnson
- Department of Biochemistry and Molecular Biology, University of Texas, Houston, TX 77030, USA
| | - Lindsay A Schwarz
- HHMI and Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Liqun Luo
- HHMI and Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
13
|
Guo X, Wang T, Huang G, Li R, Da Costa C, Li H, Lv S, Li N. Rediscovering potential molecular targets for glioma therapy through the analysis of the cell of origin, microenvironment, and metabolism. Curr Cancer Drug Targets 2021; 21:558-574. [PMID: 33949933 DOI: 10.2174/1568009621666210504091722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 11/22/2022]
Abstract
Gliomas are the most common type of malignant brain tumors. Despite significant medical advances, gliomas remain incurable and are associated with high mortality. Although numerous biomarkers of diagnostic value have been identified and significant progress in the prognosis of the outcome has been made, the treatment has not been parallelly improved during the last three decades. This review summarizes and discusses three aspects of recent discoveries related to glioma, with the objective to highlight the advantages of glioma-specific drugs targeting the cell of origin, microenvironment, and metabolism. Given the heterogeneous nature of gliomas, various cell populations have been implicated as likely sources of the tumor. Depending on the mutation(s) acquired by the cells, it is believed that neuronal stem/progenitor cells, oligodendrocyte progenitor cells, mature neurons, and glial cells can initiate cell transformation into a malignant phenotype. The level of tumorigenicity appears to be inversely correlated with the maturation of a given cell population. The microenvironment of gliomas includes non-cancer cells such as immune cells, fibroblasts, and cells of blood vessels, as well as secreted molecules and the extracellular matrix, and all these components play a vital role during tumor initiation and progression. We will discuss in detail how the tumor microenvironment can stimulate and drive the transformation of non-tumor cell populations into tumor-supporting cells or glioma cells. Metabolic reprogramming is a key feature of gliomas and is thought to reflect the adaptation to the increased nutritional requirements of tumor cell proliferation, growth, and survival. Mutations in the IDH gene can shape metabolic reprogramming and may generate some vulnerabilities in glioma cells, such as abnormal lipid metabolism and sensitivity to endoplasmic reticulum stress (ERS). We will analyze the prominent metabolic features of malignant gliomas and the key pathways regulating glioma metabolism. This review is intended to provide a conceptual background for the development of glioma therapies based on the properties of tumor cell populations, microenvironment, and metabolism.
Collapse
Affiliation(s)
- Xiaoran Guo
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| | - Tao Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| | - Guohao Huang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, No. 183 Xinqiao Street, Shapingba District, Chongqing City 400037. China
| | - Ruohan Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| | - Clive Da Costa
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT. United Kingdom
| | - Huafu Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| | - Shengqing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, No. 183 Xinqiao Street, Shapingba District, Chongqing City 400037. China
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University (SYSU), No.628, Zhenyuan Rd, Guangming Dist., Shenzhen 518107. China
| |
Collapse
|
14
|
Harder A. MEK inhibitors - novel targeted therapies of neurofibromatosis associated benign and malignant lesions. Biomark Res 2021; 9:26. [PMID: 33863389 PMCID: PMC8052700 DOI: 10.1186/s40364-021-00281-0] [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: 10/29/2020] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
MAP/ERK kinase 1 and 2 (MEK 1/2) inhibitors (MEKi) are investigated in several trials to treat lesions that arise from pathogenic variants of the Neurofibromatosis type 1 and type 2 genes (NF1, NF2). These trials showed that MEKi are capable to shrink volume of low grade gliomas and plexiform neurofibromas in NF1. Targeting other lesions being associated with a high morbidity in NF1 seems to be promising. Due to involvement of multiple pathways in NF2 associated lesions as well as in malignant tumors, MEKi are also used in combination therapies. This review outlines the current state of MEKi application in neurofibromatosis and associated benign and malignant lesions.
Collapse
Affiliation(s)
- Anja Harder
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 14, 06120, Halle (Saale), Germany. .,Institute of Neuropathology, University Hospital Münster, Münster, Germany. .,Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Potsdam, Germany.
| |
Collapse
|
15
|
Sutcliffe MD, Galvao RP, Wang L, Kim J, Rosenfeld LK, Singh S, Zong H, Janes KA. Premalignant Oligodendrocyte Precursor Cells Stall in a Heterogeneous State of Replication Stress Prior to Gliomagenesis. Cancer Res 2021; 81:1868-1882. [PMID: 33531372 PMCID: PMC8137536 DOI: 10.1158/0008-5472.can-20-1037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 12/02/2020] [Accepted: 01/28/2021] [Indexed: 11/16/2022]
Abstract
Cancer evolves from premalignant clones that adopt unusual cell states to achieve transformation. We previously pinpointed the oligodendrocyte precursor cell (OPC) as a cell of origin for glioma, but the early changes of mutant OPCs during premalignancy remained unknown. Using mice engineered for inducible Nf1-Trp53 loss in OPCs, we acutely isolated labeled mutant OPCs by laser-capture microdissection, determined global gene-expression changes by bulk RNA sequencing, and compared with cell-state fluctuations at the single-cell level by stochastic profiling, which uses RNA-sequencing measurements from random pools of 10 mutant cells. At 12 days after Nf1-Trp53 deletion, bulk differences were mostly limited to mitotic hallmarks and genes for ribosome biosynthesis, and stochastic profiling revealed a spectrum of stem-progenitor (Axl, Aldh1a1), proneural, and mesenchymal states as potential starting points for gliomagenesis. At 90 days, bulk sequencing detected few differentially expressed transcripts, whereas stochastic profiling revealed cell states for neurons and mural cells that do not give rise to glial tumors, suggesting cellular dead-ends for gliomagenesis. Importantly, mutant OPCs that strongly expressed key effectors of nonsense-mediated decay (Upf3b) and homology-dependent DNA repair (Rad51c, Slx1b, Ercc4) were identified along with DNA-damage markers, suggesting transcription-associated replication stress. Analysis of 10-cell transcriptomes at 90 days identified a locus of elevated gene expression containing an additional repair endonuclease (Mus81) and Rin1, a Ras-Raf antagonist and possible counterbalance to Nf1 loss, which was microdeleted or downregulated in gliomas at 150 days. These hidden cell-state variations uncover replication stress as a potential bottleneck that must be resolved for glioma initiation. SIGNIFICANCE: Profiling premalignant cell states in a mouse model of glioma uncovers regulatory heterogeneity in glioma cells-of-origin and defines a state of replication stress that precedes tumor initiation.See related articles by Singh and colleagues, p. 1840 and Schaff and colleagues, p. 1853.
Collapse
Affiliation(s)
- Matthew D Sutcliffe
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Rui P Galvao
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Jungeun Kim
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Lauren K Rosenfeld
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Shambhavi Singh
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
| | - Hui Zong
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, Virginia.
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia.
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
16
|
Aggarwal N, Yadav J, Thakur K, Bibban R, Chhokar A, Tripathi T, Bhat A, Singh T, Jadli M, Singh U, Kashyap MK, Bharti AC. Human Papillomavirus Infection in Head and Neck Squamous Cell Carcinomas: Transcriptional Triggers and Changed Disease Patterns. Front Cell Infect Microbiol 2020. [PMID: 33344262 DOI: 10.3389/fcimb.2020.537650,] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a heterogeneous group of cancers. Collectively, HNSCC ranks sixth in incidence rate worldwide. Apart from classical risk factors like tobacco and alcohol, infection of human papillomavirus (HPV) is emerging as a discrete risk factor for HNSCC. HPV-positive HNSCC represent a distinct group of diseases that differ in their clinical presentation. These lesions are well-differentiated, occur at an early age, and have better prognosis. Epidemiological studies have demonstrated a specific increase in the proportions of the HPV-positive HNSCC. HPV-positive and HPV-negative HNSCC lesions display different disease progression and clinical response. For tumorigenic-transformation, HPV essentially requires a permissive cellular environment and host cell factors for induction of viral transcription. As the spectrum of host factors is independent of HPV infection at the time of viral entry, presumably entry of HPV only selects host cells that are permissive to establishment of HPV infection. Growing evidence suggest that HPV plays a more active role in a subset of HNSCC, where they are transcriptionally-active. A variety of factors provide a favorable environment for HPV to become transcriptionally-active. The most notable are the set of transcription factors that have direct binding sites on the viral genome. As HPV does not have its own transcription machinery, it is fully dependent on host transcription factors to complete the life cycle. Here, we review and evaluate the current evidence on level of a subset of host transcription factors that influence viral genome, directly or indirectly, in HNSCC. Since many of these transcription factors can independently promote carcinogenesis, the composition of HPV permissive transcription factors in a tumor can serve as a surrogate marker of a separate molecularly-distinct class of HNSCC lesions including those cases, where HPV could not get a chance to infect but may manifest better prognosis.
Collapse
Affiliation(s)
- Nikita Aggarwal
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Joni Yadav
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Kulbhushan Thakur
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Rakhi Bibban
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Arun Chhokar
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Tanya Tripathi
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Anjali Bhat
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Tejveer Singh
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Mohit Jadli
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Ujala Singh
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Manoj K Kashyap
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India.,Amity Medical School, Stem Cell Institute, Amity University Haryana, Amity Education Valley Panchgaon, Gurugram, India
| | - Alok C Bharti
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| |
Collapse
|
17
|
Terry TT, Cheng T, Mahjoub M, Zong H. Mosaic Analysis with Double Markers reveals IGF1R function in granule cell progenitors during cerebellar development. Dev Biol 2020; 465:130-143. [PMID: 32697974 DOI: 10.1016/j.ydbio.2020.07.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
During cerebellar development, granule cell progenitors (GCPs) proliferate exponentially for a fixed period, promoted by paracrine mitogenic factor Sonic Hedgehog (Shh) secreted from Purkinje cells (PCs). Dysregulation of Shh signaling leads to uncontrolled GCP proliferation and medulloblastoma. Serendipitously our previous work discovered insulin-like growth factor 1 (IGF1) as another key driver for medulloblastoma, which led to the current investigation into the role of IGF1 in GCPs during normal development. While the IGF1R conditional knockout model revealed GCP defects in anterior cerebellum, the posterior cerebellum was mostly intact, likely owing to incomplete excision of floxed alleles. To circumvent this hurdle, we enlisted a mouse genetic system called Mosaic Analysis of Double Markers (MADM), which sporadically generates homozygous null cells unequivocally labeled with GFP and their wildtype sibling cells labeled with RFP, enabling phenotypic analysis at single-cell resolution. Using MADM, we found that loss of IGF1R resulted in a 10-fold reduction of GCs in both anterior and posterior cerebellum; and that hindered S phase entry and increased cell cycle exit collectively led to this phenotype. Genetic interaction studies showed that IGF1 signaling prevents GCP cell cycle exit at least partially through suppressing the level of p27kip1, a negative regulator of cell cycle. Finally, we found that IGF1 is produced by PCs in a temporally regulated fashion: it is highly expressed early in development when GCPs proliferate exponentially, then gradually decline as GCPs commit to cell cycle exit. Taken together, our studies reveal IGF1 as a paracrine factor that positively regulates GCP cell cycle in cooperation with Shh, through dampening the level of p27 to prevent precocious cell cycle exit. Our work not only showcases the power of phenotypic analysis by the MADM system but also provides an excellent example of multi-factorial regulation of robust developmental programs.
Collapse
Affiliation(s)
- Tiffany T Terry
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Tao Cheng
- Department of Medicine, Division of Nephrology, Washington University in St. Louis, St. Louis, MO, USA
| | - Moe Mahjoub
- Department of Medicine, Division of Nephrology, Washington University in St. Louis, St. Louis, MO, USA
| | - Hui Zong
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
18
|
Yao M, Ventura PB, Jiang Y, Rodriguez FJ, Wang L, Perry JSA, Yang Y, Wahl K, Crittenden RB, Bennett ML, Qi L, Gong CC, Li XN, Barres BA, Bender TP, Ravichandran KS, Janes KA, Eberhart CG, Zong H. Astrocytic trans-Differentiation Completes a Multicellular Paracrine Feedback Loop Required for Medulloblastoma Tumor Growth. Cell 2020; 180:502-520.e19. [PMID: 31983537 PMCID: PMC7259679 DOI: 10.1016/j.cell.2019.12.024] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/16/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment (TME) is critical for tumor progression. However, the establishment and function of the TME remain obscure because of its complex cellular composition. Using a mouse genetic system called mosaic analysis with double markers (MADMs), we delineated TME evolution at single-cell resolution in sonic hedgehog (SHH)-activated medulloblastomas that originate from unipotent granule neuron progenitors in the brain. First, we found that astrocytes within the TME (TuAstrocytes) were trans-differentiated from tumor granule neuron precursors (GNPs), which normally never differentiate into astrocytes. Second, we identified that TME-derived IGF1 promotes tumor progression. Third, we uncovered that insulin-like growth factor 1 (IGF1) is produced by tumor-associated microglia in response to interleukin-4 (IL-4) stimulation. Finally, we found that IL-4 is secreted by TuAstrocytes. Collectively, our studies reveal an evolutionary process that produces a multi-lateral network within the TME of medulloblastoma: a fraction of tumor cells trans-differentiate into TuAstrocytes, which, in turn, produce IL-4 that stimulates microglia to produce IGF1 to promote tumor progression.
Collapse
Affiliation(s)
- Maojin Yao
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - P Britten Ventura
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ying Jiang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Fausto J Rodriguez
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Justin S A Perry
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yibo Yang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kelsey Wahl
- Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Rowena B Crittenden
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
| | - Mariko L Bennett
- Department of Neurobiology, Stanford University, Palo Alto, CA 94305, USA
| | - Lin Qi
- Brain Tumor Program, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cong-Cong Gong
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Xiao-Nan Li
- Brain Tumor Program, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University, Palo Alto, CA 94305, USA
| | - Timothy P Bender
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA; VIB-UGent Center for Inflammation Research and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Charles G Eberhart
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hui Zong
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
19
|
Aggarwal N, Yadav J, Thakur K, Bibban R, Chhokar A, Tripathi T, Bhat A, Singh T, Jadli M, Singh U, Kashyap MK, Bharti AC. Human Papillomavirus Infection in Head and Neck Squamous Cell Carcinomas: Transcriptional Triggers and Changed Disease Patterns. Front Cell Infect Microbiol 2020; 10:537650. [PMID: 33344262 PMCID: PMC7738612 DOI: 10.3389/fcimb.2020.537650] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 11/02/2020] [Indexed: 02/05/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a heterogeneous group of cancers. Collectively, HNSCC ranks sixth in incidence rate worldwide. Apart from classical risk factors like tobacco and alcohol, infection of human papillomavirus (HPV) is emerging as a discrete risk factor for HNSCC. HPV-positive HNSCC represent a distinct group of diseases that differ in their clinical presentation. These lesions are well-differentiated, occur at an early age, and have better prognosis. Epidemiological studies have demonstrated a specific increase in the proportions of the HPV-positive HNSCC. HPV-positive and HPV-negative HNSCC lesions display different disease progression and clinical response. For tumorigenic-transformation, HPV essentially requires a permissive cellular environment and host cell factors for induction of viral transcription. As the spectrum of host factors is independent of HPV infection at the time of viral entry, presumably entry of HPV only selects host cells that are permissive to establishment of HPV infection. Growing evidence suggest that HPV plays a more active role in a subset of HNSCC, where they are transcriptionally-active. A variety of factors provide a favorable environment for HPV to become transcriptionally-active. The most notable are the set of transcription factors that have direct binding sites on the viral genome. As HPV does not have its own transcription machinery, it is fully dependent on host transcription factors to complete the life cycle. Here, we review and evaluate the current evidence on level of a subset of host transcription factors that influence viral genome, directly or indirectly, in HNSCC. Since many of these transcription factors can independently promote carcinogenesis, the composition of HPV permissive transcription factors in a tumor can serve as a surrogate marker of a separate molecularly-distinct class of HNSCC lesions including those cases, where HPV could not get a chance to infect but may manifest better prognosis.
Collapse
Affiliation(s)
- Nikita Aggarwal
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Joni Yadav
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Kulbhushan Thakur
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Rakhi Bibban
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Arun Chhokar
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Tanya Tripathi
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Anjali Bhat
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Tejveer Singh
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Mohit Jadli
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Ujala Singh
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Manoj K. Kashyap
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Amity Medical School, Stem Cell Institute, Amity University Haryana, Amity Education Valley Panchgaon, Gurugram, India
| | - Alok C. Bharti
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- *Correspondence: Alok C. Bharti,
| |
Collapse
|
20
|
Ohka F, Shinjo K, Deguchi S, Matsui Y, Okuno Y, Katsushima K, Suzuki M, Kato A, Ogiso N, Yamamichi A, Aoki K, Suzuki H, Sato S, Arul Rayan N, Prabhakar S, Göke J, Shimamura T, Maruyama R, Takahashi S, Suzumura A, Kimura H, Wakabayashi T, Zong H, Natsume A, Kondo Y. Pathogenic Epigenetic Consequences of Genetic Alterations in IDH-Wild-Type Diffuse Astrocytic Gliomas. Cancer Res 2019; 79:4814-4827. [DOI: 10.1158/0008-5472.can-19-1272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/19/2019] [Accepted: 07/24/2019] [Indexed: 11/16/2022]
|