1
|
Li K, Zheng Y, Cai S, Fan Z, Yang J, Liu Y, Liang S, Song M, Du S, Qi L. The subventricular zone structure, function and implications for neurological disease. Genes Dis 2025; 12:101398. [PMID: 39935607 PMCID: PMC11810716 DOI: 10.1016/j.gendis.2024.101398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 05/28/2024] [Accepted: 07/28/2024] [Indexed: 02/13/2025] Open
Abstract
The subventricular zone (SVZ) is a region surrounding the lateral ventricles that contains neural stem cells and neural progenitor cells, which can proliferate and differentiate into various neural and glial cells. SVZ cells play important roles in neurological diseases like neurodegeneration, neural injury, and glioblastoma multiforme. Investigating the anatomy, structure, composition, physiology, disease associations, and related mechanisms of SVZ is significant for neural stem cell therapy and treatment/prevention of neurological disorders. However, challenges remain regarding the mechanisms regulating SVZ cell proliferation, differentiation, and migration, delivering cells to damaged areas, and immune responses. In-depth studies of SVZ functions and related therapeutic developments may provide new insights and approaches for treating brain injuries and degenerative diseases, as well as a scientific basis for neural stem cell therapy. This review summarizes research findings on SVZ and neurological diseases to provide references for relevant therapies.
Collapse
Affiliation(s)
- Kaishu Li
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Yin Zheng
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Shubing Cai
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Zhiming Fan
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Junyi Yang
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Yuanrun Liu
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Shengqi Liang
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Meihui Song
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Siyuan Du
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Ling Qi
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| |
Collapse
|
2
|
Gecici NN, Habib A, Mallela AN, Rich JN, Drappatz J, Mantica M, Abdullah KG, Zinn PO. Ventricular Entry During Glioblastoma Resection is Associated With Reduced Survival and Increased Risk of Distant Recurrence. Neurosurgery 2025:00006123-990000000-01554. [PMID: 40178259 DOI: 10.1227/neu.0000000000003431] [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: 09/18/2024] [Accepted: 12/10/2024] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Although subventricular zone (SVZ) involvement is known to correlate with more aggressive tumor behavior and reduced survival in glioblastoma (GBM), the role of ventricular entry (VE) on outcomes is less clear and remains debated. This study aims to investigate the impact of VE on outcomes and overall survival (OS) in GBM. METHODS A retrospective analysis of patients with newly diagnosed supratentorial GBMtreated between 2013 and 2023 at the University of Pittsburgh Medical Center was performed. SVZ involvement, size, and extent of resection were identified through preoperative and postoperative imaging. VE was identified through operative notes and postoperative imaging review. RESULTS A total of 282 patients met inclusion criteria. VE occurred in 38.3% (n = 108) of patients and was more common in those with SVZ-contacting tumors (P < .001). Patients who had VE had significantly lower median OS compared with non-VE (12 months vs 18 months, P < .001). VE was identified as an independent risk factor for decreased OS in patients with GBM, after adjusting for well-known prognostic factors and SVZ contact (hazard ratios: 1.62 [1.12-2.34], P = .001). Only patients who had VE developed postoperative hydrocephalus (n = 4, 1.4%, P = .021) and had external ventricular drain placed (n = 6, 2.1%, P = .003). Distant parenchymal recurrence and leptomeningeal dissemination (LMD) rates were significantly higher in the VE group compared with the non-VE group (63.9% vs 39.7%, P < .001, and 23.1% vs 13.2%, P = .035), and VE emerged as an independent predictor of distant recurrences/LMDs in multivariable logistic regression (odds ratio: 4.7 [2.11-10.4], P < .001). CONCLUSION Our data suggest that VE during GBM resection is a significant independent risk factor for decreased survival and increased distant recurrence/LMD. While maximizing tumor resection remains critical, neurosurgeons must consider the potential adverse outcomes associated with VE because it may diminish the survival benefits of gross-total resection. Prospective studies are warranted to better understand the risks and benefits of VE in GBM surgery.
Collapse
Affiliation(s)
- Neslihan Nisa Gecici
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Rodríguez-Mendoza B, Figueroa-González A, Cano-Herrera G, Gutiérrez-Rosas LE, Romero-Torres CI, Victoria-García LO, González-Castillo P, Guerrero-Cázares H, Ibarra A. [Glioblastoma and its interaction with neurogenesis]. Rev Neurol 2024; 79:279-287. [PMID: 39540380 PMCID: PMC11605900 DOI: 10.33588/rn.7910.2024226] [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: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GBM) is the most frequent and aggressive malignant primary tumor of the central nervous system in adults, with an incidence of 3.23 per 100,000 people. Despite the existence of various therapeutic approaches, the absence of a cure and the unfavorable prognosis persist for this neoplasm, with a median survival of approximately 8-15 months and a 5-year survival rate of 6.9%. In this review, we address the epidemiology, histopathology, molecular characteristics, and treatment of GBM. We highlight the relationship of GBM with the microenvironment in the lateral ventricle wall and the cerebrospinal fluid. The location of GBM in this region results in more aggressive tumors and shorter life expectancy for patients. Understanding the malignancy mechanisms that hinder remission, treatment, and positive prognosis opens the possibility of improving diagnostic and therapeutic interventions against GBM.
Collapse
Affiliation(s)
- Brenda Rodríguez-Mendoza
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Aura Figueroa-González
- Neurosurgery Department. Mayo Clinic Jacksonville. Jacksonville, Estados UnidosMayo Clinic JacksonvilleMayo Clinic JacksonvilleJacksonvilleEstados Unidos
| | - Gabriela Cano-Herrera
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Luis E. Gutiérrez-Rosas
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Carlos I. Romero-Torres
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Luis O. Victoria-García
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Paola González-Castillo
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
| | - Hugo Guerrero-Cázares
- Neurosurgery Department. Mayo Clinic Jacksonville. Jacksonville, Estados UnidosMayo Clinic JacksonvilleMayo Clinic JacksonvilleJacksonvilleEstados Unidos
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA). FCS. Universidad Anáhuac México Campus Norte. Huixquilucan. Ciudad de México, MéxicoUniversidad Anáhuac México Campus NorteUniversidad Anáhuac México Campus NorteCiudad de MéxicoMéxico
- Secretaría de la Defensa Nacional. Escuela Militar de Graduados en Sanidad. Ciudad de México, MéxicoEscuela Militar de Graduados en SanidadEscuela Militar de Graduados en SanidadCiudad de MéxicoMéxico
| |
Collapse
|
4
|
Hu L, Liu M, Tang B, Li X, Xu H, Wang H, Wang D, Liu S, Xu C. PARD6A promotes lung adenocarcinoma cell proliferation and invasion through Serpina3. Cancer Gene Ther 2024; 31:1696-1707. [PMID: 39300216 DOI: 10.1038/s41417-024-00829-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024]
Abstract
Par6α encoded by PARD6A is a member of the PAR6 family and is reported to promote cancer initiation and progression. PARD6A is frequently upregulated in different types of cancers, but its regulatory role in lung cancer progression is yet to be established. In this study, we analyzed the PARD6A expression in biopsies from lung adenocarcinoma (LUAD) patients, and the survival probability using LUAD tissue microarray (TMA) and online datasets from TCGA and GEO. We conducted in vitro and in vivo assays to assess the role of PARD6A in regulating lung cancer progression, including proliferation, wound healing, transwell, RNA-seq, and subcutaneous tumor mice models. Our findings revealed that PARD6A is highly expressed in cancer tissues from LUAD patients and is associated with poor prognosis in LUAD patients. In vitro assays showed that PARD6A promoted cell proliferation, migration, and invasion. The transcriptome sequencing identified Serpina3 as one of the key downstream molecules of PARD6A. Ectopic expression of Serpina3 rescued impaired proliferation, migration, and invasion in PARD6A-knocking down H1299 cells, whereas silencing Serpina3 impeded enhanced proliferation, migration, and invasion in PARD6A-overexpressing H1975 cells. Our findings suggest that PARD6A promotes lung cancer progression by inducing Serpina3, which may be a promising therapeutic target.
Collapse
Affiliation(s)
- Lanlin Hu
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Mingxin Liu
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610042, China
| | - Bo Tang
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Xurui Li
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, 400716, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Huasheng Xu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Huani Wang
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Dandan Wang
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Guangxi Engineering Center in Biomedical Material for Tissue and Organ Regeneration & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, 530021, China.
| | - Chuan Xu
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
5
|
Norton ES, Whaley LA, Jones VK, Brooks MM, Russo MN, Morderer D, Jessen E, Schiapparelli P, Ramos-Fresnedo A, Zarco N, Carrano A, Rossoll W, Asmann YW, Lam TT, Chaichana KL, Anastasiadis PZ, Quiñones-Hinojosa A, Guerrero-Cázares H. Cell-specific cross-talk proteomics reveals cathepsin B signaling as a driver of glioblastoma malignancy near the subventricular zone. SCIENCE ADVANCES 2024; 10:eadn1607. [PMID: 39110807 PMCID: PMC11305394 DOI: 10.1126/sciadv.adn1607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Glioblastoma (GBM) is the most prevalent and aggressive malignant primary brain tumor. GBM proximal to the lateral ventricles (LVs) is more aggressive, potentially because of subventricular zone contact. Despite this, cross-talk between GBM and neural stem/progenitor cells (NSC/NPCs) is not well understood. Using cell-specific proteomics, we show that LV-proximal GBM prevents neuronal maturation of NSCs through induction of senescence. In addition, GBM brain tumor-initiating cells (BTICs) increase expression of cathepsin B (CTSB) upon interaction with NPCs. Lentiviral knockdown and recombinant protein experiments reveal that both cell-intrinsic and soluble CTSB promote malignancy-associated phenotypes in BTICs. Soluble CTSB stalls neuronal maturation in NPCs while promoting senescence, providing a link between LV-tumor proximity and neurogenesis disruption. Last, we show LV-proximal CTSB up-regulation in patients, showing the relevance of this cross-talk in human GBM biology. These results demonstrate the value of proteomic analysis in tumor microenvironment research and provide direction for new therapeutic strategies in GBM.
Collapse
Affiliation(s)
- Emily S. Norton
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
- Regenerative Sciences Training Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Lauren A. Whaley
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Vanessa K. Jones
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Mieu M. Brooks
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Marissa N. Russo
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Dmytro Morderer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Erik Jessen
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | | | - Natanael Zarco
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Anna Carrano
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Wilfried Rossoll
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yan W. Asmann
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA
| | - TuKiet T. Lam
- Keck MS and Proteomics Resource, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | |
Collapse
|
6
|
Zhu M, Lan Z, Park J, Gong S, Wang Y, Guo F. Regulation of CNS pathology by Serpina3n/SERPINA3: The knowns and the puzzles. Neuropathol Appl Neurobiol 2024; 50:e12980. [PMID: 38647003 PMCID: PMC11131959 DOI: 10.1111/nan.12980] [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: 01/09/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024]
Abstract
Neuroinflammation, blood-brain barrier (BBB) dysfunction, neuron and glia injury/death and myelin damage are common central nervous system (CNS) pathologies observed in various neurological diseases and injuries. Serine protease inhibitor (Serpin) clade A member 3n (Serpina3n), and its human orthologue SERPINA3, is an acute-phase inflammatory glycoprotein secreted primarily by the liver into the bloodstream in response to systemic inflammation. Clinically, SERPINA3 is dysregulated in brain cells, cerebrospinal fluid and plasma in various neurological conditions. Although it has been widely accepted that Serpina3n/SERPINA3 is a reliable biomarker of reactive astrocytes in diseased CNS, recent data have challenged this well-cited concept, suggesting instead that oligodendrocytes and neurons are the primary sources of Serpina3n/SERPINA3. The debate continues regarding whether Serpina3n/SERPINA3 induction represents a pathogenic or a protective mechanism. Here, we propose possible interpretations for previously controversial data and present perspectives regarding the potential role of Serpina3n/SERPINA3 in CNS pathologies, including demyelinating disorders where oligodendrocytes are the primary targets. We hypothesise that the 'good' or 'bad' aspects of Serpina3n/SERPINA3 depend on its cellular sources, its subcellular distribution (or mis-localisation) and/or disease/injury types. Furthermore, circulating Serpina3n/SERPINA3 may cross the BBB to impact CNS pathologies. Cell-specific genetic tools are critically important to tease out the potential roles of cell type-dependent Serpina3n in CNS diseases/injuries.
Collapse
Affiliation(s)
- Meina Zhu
- Department of Neurology, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, California, USA
| | - Zhaohui Lan
- Center for Brain Health and Brain Technology, Global Institute of Future Technology, Shanghai Jiao Tong University, Shanghai, China
| | - Joohyun Park
- Department of Neurology, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, California, USA
| | | | - Yan Wang
- Department of Neurology, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, California, USA
| | - Fuzheng Guo
- Department of Neurology, UC Davis School of Medicine, Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals for Children, Sacramento, California, USA
| |
Collapse
|
7
|
Alberti G, Sánchez-López CM, Marcilla A, Barone R, Caruso Bavisotto C, Graziano F, Conway de Macario E, Macario AJL, Bucchieri F, Cappello F, Campanella C, Rappa F. Hsp70 and Calcitonin Receptor Protein in Extracellular Vesicles from Glioblastoma Multiforme: Biomarkers with Putative Roles in Carcinogenesis and Potential for Differentiating Tumor Types. Int J Mol Sci 2024; 25:3415. [PMID: 38542389 PMCID: PMC10969952 DOI: 10.3390/ijms25063415] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 11/11/2024] Open
Abstract
Glioblastoma multiforme (GBM) is a malignancy of bad prognosis, and advances in early detection and treatment are needed. GBM is heterogenous, with varieties differing in malignancy within a tumor of a patient and between patients. Means are needed to distinguish these GMB forms, so that specific strategies can be deployed for patient management. We study the participation of the chaperone system (CS) in carcinogenesis. The CS is dynamic, with its members moving around the body in extracellular vesicles (EVs) and interacting with components of other physiological systems in health and disease, including GBM. Here, we describe the finding of high amounts of Hsp70 (HSPA1A) and the calcitonin receptor protein (CTR) in EVs in patients with GBM. We present a standardized protocol for collecting, purifying, and characterizing EVs carrying Hsp70 and CTR in plasma-derived EVs from patients with GBM. EVs from GBM patients were obtained just before tumor ablative surgery (T0) and 7 days afterwards (T1); Hsp70 was highly elevated at T0 and less so at T1, and CTR was greatly increased at T0 and reduced to below normal values at T1. Our results encourage further research to assess Hsp70 and CTR as biomarkers for differentiating tumor forms and to determine their roles in GBM carcinogenesis.
Collapse
Affiliation(s)
- Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (R.B.); (C.C.B.); (F.B.); (F.C.); (C.C.)
| | - Christian M. Sánchez-López
- Área de Parasitología, Departamento Farmacia y Tecnología Farmacéutica y Parasitología, Universitat de València, 46100 Burjassot, Spain; (C.M.S.-L.); (A.M.)
- Joint Unit of Endocrinology, Nutrition and Clinical Dietetics, Instituto de Investigación Sanitaria-La Fe, 46026 Valencia, Spain
| | - Antonio Marcilla
- Área de Parasitología, Departamento Farmacia y Tecnología Farmacéutica y Parasitología, Universitat de València, 46100 Burjassot, Spain; (C.M.S.-L.); (A.M.)
- Joint Unit of Endocrinology, Nutrition and Clinical Dietetics, Instituto de Investigación Sanitaria-La Fe, 46026 Valencia, Spain
| | - Rosario Barone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (R.B.); (C.C.B.); (F.B.); (F.C.); (C.C.)
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (R.B.); (C.C.B.); (F.B.); (F.C.); (C.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
| | - Francesca Graziano
- Department of Neurosurgery, Highly Specialized Hospital of National Importance “Garibaldi”, 95122 Catania, Italy;
| | - Everly Conway de Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (R.B.); (C.C.B.); (F.B.); (F.C.); (C.C.)
| | - Francesco Cappello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (R.B.); (C.C.B.); (F.B.); (F.C.); (C.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy; (E.C.d.M.); (A.J.L.M.)
| | - Claudia Campanella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (R.B.); (C.C.B.); (F.B.); (F.C.); (C.C.)
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (R.B.); (C.C.B.); (F.B.); (F.C.); (C.C.)
- The Institute of Translational Pharmacology, National Research Council of Italy (CNR), 90146 Palermo, Italy
| |
Collapse
|
8
|
de Mezer M, Markowska A, Markowska J, Krzyżaniak M, Grabarek BO, Pokusa F, Żurawski J. Immunohistochemical Expression of the SERPINA3 Protein in Uterine Fibroids. Curr Pharm Biotechnol 2024; 25:1758-1765. [PMID: 38204235 DOI: 10.2174/0113892010264673231111082438] [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: 06/22/2023] [Revised: 09/01/2023] [Accepted: 09/21/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND SERPINA3 (α-1-antichymotrypsin, AACT, ACT) is produced by the liver and released into plasma in an anti-inflammatory response and plays a role as a modulator of extracellular matrix (ECM) by inhibiting serine proteases. Numerous studies proved an increased level of SERPINA3 in many types of cancer, which could be linked to SERPINA3's anti-apoptotic function. AIM In the context of progressive ECM fibrosis during the development of uterine fibroids, which are one of the most common hypertrophic changes within the uterus, it is interesting to describe the level of SERPINA3 protein in this type of lesion and the surrounding tissues. METHODS We used immunohistochemical staining of the SERPINA3 protein and compared the intensity of the signal between the myoma tissue and the surrounding normal tissue. RESULTS We showed a surprising reduction in the amount of the SERPINA3 protein within uterine fibroids compared to surrounding tissues. CONCLUSION This observation sheds new light on the role of this protein in the formation of proliferative changes and suggests that understanding the mechanism of its action may become the basis for the development of new diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Mateusz de Mezer
- Department of Immunobiology, Chair of Medical Biology, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Markowska
- Department of Perinatology and Women's Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Monika Krzyżaniak
- Department of Oncological Pathology, Lord's Transfiguration Clinical Hospital, Partner of Poznan University of Medical Sciences, Poznan, Poland
| | - Beniamin Oskar Grabarek
- Collegium Medicum, WSB University, 41-300 Dabrowa Gornicza, Poland
- Department of Histology, Cytophysiology and Embryology, Katowice School of Technology, Katowice, Poland
| | - Filip Pokusa
- Faculty of Economics and Pedagogy, Higher School of Management and Administration in Opole, 46-020 Opole, Poland
| | - Jakub Żurawski
- Department of Immunobiology, Chair of Medical Biology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
9
|
Singh S, Joshi V, Upadhyay A. Amyloids and brain cancer: molecular linkages and crossovers. Biosci Rep 2023; 43:BSR20230489. [PMID: 37335084 PMCID: PMC10548166 DOI: 10.1042/bsr20230489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/31/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023] Open
Abstract
Amyloids are high-order proteinaceous formations deposited in both intra- and extracellular spaces. These aggregates have tendencies to deregulate cellular physiology in multiple ways; for example, altered metabolism, mitochondrial dysfunctions, immune modulation, etc. When amyloids are formed in brain tissues, the endpoint often is death of neurons. However, interesting but least understood is a close connection of amyloids with another set of conditions in which brain cells proliferate at an extraordinary rate and form tumor inside brain. Glioblastoma is one such condition. Increasing number of evidence indicate a possible link between amyloid formation and depositions in brain tumors. Several proteins associated with cell cycle regulation and apoptotic pathways themselves have shown to possess high tendencies to form amyloids. Tumor suppressor protein p53 is one prominent example that mutate, oligomerize and form amyloids leading to loss- or gain-of-functions and cause increased cell proliferation and malignancies. In this review article, we present available examples, genetic links and common pathways that indicate that possibly the two distantly placed pathways: amyloid formation and developing cancers in the brain have similarities and are mechanistically intertwined together.
Collapse
Affiliation(s)
- Shalini Singh
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, U.S.A
| | - Vibhuti Joshi
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh 201310, India
| | - Arun Upadhyay
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, U.S.A
| |
Collapse
|
10
|
Norton ES, Whaley LA, Jones VK, Brooks MM, Russo MN, Morderer D, Jessen E, Schiapparelli P, Ramos-Fresnedo A, Zarco N, Carrano A, Rossoll W, Asmann YW, Lam TT, Chaichana KL, Anastasiadis PZ, Quiñones-Hinojosa A, Guerrero-Cázares H. Cell-specific crosstalk proteomics reveals cathepsin B signaling as a driver of glioblastoma malignancy near the subventricular zone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.19.553966. [PMID: 37662251 PMCID: PMC10473635 DOI: 10.1101/2023.08.19.553966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Glioblastoma (GBM) is the most prevalent and aggressive malignant primary brain tumor. GBM proximal to the lateral ventricles (LVs) is more aggressive, potentially due to subventricular zone (SVZ) contact. Despite this, crosstalk between GBM and neural stem/progenitor cells (NSC/NPCs) is not well understood. Using cell-specific proteomics, we show that LV-proximal GBM prevents neuronal maturation of NSCs through induction of senescence. Additionally, GBM brain tumor initiating cells (BTICs) increase expression of CTSB upon interaction with NPCs. Lentiviral knockdown and recombinant protein experiments reveal both cell-intrinsic and soluble CTSB promote malignancy-associated phenotypes in BTICs. Soluble CTSB stalls neuronal maturation in NPCs while promoting senescence, providing a link between LV-tumor proximity and neurogenesis disruption. Finally, we show LV-proximal CTSB upregulation in patients, showing the relevance of this crosstalk in human GBM biology. These results demonstrate the value of proteomic analysis in tumor microenvironment research and provide direction for new therapeutic strategies in GBM. Highlights Periventricular GBM is more malignant and disrupts neurogenesis in a rodent model.Cell-specific proteomics elucidates tumor-promoting crosstalk between GBM and NPCs.NPCs induce upregulated CTSB expression in GBM, promoting tumor progression.GBM stalls neurogenesis and promotes NPC senescence via CTSB.
Collapse
|
11
|
Jin Y, Zhang Y, Huang A, Chen Y, Wang J, Liu N, Wang X, Gong Y, Wang W, Pan J. Overexpression of SERPINA3 suppresses tumor progression by modulating SPOP/NF‑κB in lung cancer. Int J Oncol 2023; 63:96. [PMID: 37417362 PMCID: PMC10552721 DOI: 10.3892/ijo.2023.5544] [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: 02/02/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
The pathogenesis mechanism of lung cancer is very complex, with high incidence and mortality. Serpin family A member 3 (SERPINA3) expression levels were reduced in the sera of patients with lung cancer and may be a candidate diagnostic and prognostic survival biomarker in lung cancer, as previously reported. However, the detailed biological functions of SERPINA3 in the pathogenesis of lung cancer remain unknown. In the present study, it was aimed to explore the effects of SERPINA3 on the occurrence of lung cancer. SERPINA3 expression was assessed using bioinformatics database analysis and experimental detection. Then, the biological effects of SERPINA3 were investigated in a cell culture system and a xenograft model of human lung cancer. The potential regulatory mechanism of SERPINA3 in lung cancer was explored by data‑independent acquisition mass spectrometry (DIA‑MS) detection and further validated by western blotting (WB). The results indicated that SERPINA3 expression levels were significantly downregulated in lung cancer tissues and cell lines. At the cellular level, it was revealed that overexpressed SERPINA3 inhibited cell growth, proliferation, migration and invasion and promoted the apoptosis of lung cancer cells. Moreover, overexpressed SERPINA3 enhanced the sensitivity of lung cancer cells to osimertinib. In vivo, a xenograft model of human lung cancer was established with BALB/c nude mice. After the injection of A549 cells, the tumor growth of the tumor‑bearing mice in the SERPINA3‑overexpressing group increased more slowly, and the tumor volume was smaller than that in the empty‑vector group. Mechanistically, a total of 65 differentially expressed proteins were identified. It was found that the speckle‑type POZ protein (SPOP) was significantly upregulated in SERPINA3‑overexpressing H157 cells using DIA‑MS detection and analysis. WB validation showed that SPOP expression increased, and NF‑kappaB (NF‑κB) p65 was inhibited in cell lines and tumor tissues of mice when SERPINA3 was overexpressed. The present findings suggest that SERPINA3 is involved in the development of lung cancer and has an antineoplastic role in lung cancer.
Collapse
Affiliation(s)
- Yanxia Jin
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, Huangshi, Hubei 435002
| | - Yueyang Zhang
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, Huangshi, Hubei 435002
| | - Ankang Huang
- Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Ying Chen
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, Huangshi, Hubei 435002
| | - Jinsong Wang
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, Huangshi, Hubei 435002
| | - Na Liu
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, Huangshi, Hubei 435002
| | - Xianping Wang
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, Huangshi, Hubei 435002
| | - Yongsheng Gong
- Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Weidong Wang
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, Huangshi, Hubei 435002
| | - Jicheng Pan
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, Huangshi, Hubei 435002
| |
Collapse
|
12
|
Saeui CT, Shah SR, Fernandez-Gil BI, Zhang C, Agatemor C, Dammen-Brower K, Mathew MP, Buettner M, Gowda P, Khare P, Otamendi-Lopez A, Yang S, Zhang H, Le A, Quinoñes-Hinojosa A, Yarema KJ. Anticancer Properties of Hexosamine Analogs Designed to Attenuate Metabolic Flux through the Hexosamine Biosynthetic Pathway. ACS Chem Biol 2023; 18:151-165. [PMID: 36626752 DOI: 10.1021/acschembio.2c00784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Altered cellular metabolism is a hallmark of cancer pathogenesis and progression; for example, a near-universal feature of cancer is increased metabolic flux through the hexosamine biosynthetic pathway (HBP). This pathway produces uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), a potent oncometabolite that drives multiple facets of cancer progression. In this study, we synthesized and evaluated peracetylated hexosamine analogs designed to reduce flux through the HBP. By screening a panel of analogs in pancreatic cancer and glioblastoma multiform (GBM) cells, we identified Ac4Glc2Bz─a benzyl-modified GlcNAc mimetic─as an antiproliferative cancer drug candidate that down-regulated oncogenic metabolites and reduced GBM cell motility at concentrations non-toxic to non-neoplastic cells. More specifically, the growth inhibitory effects of Ac4Glc2Bz were linked to reduced levels of UDP-GlcNAc and concomitant decreases in protein O-GlcNAc modification in both pancreatic cancer and GBM cells. Targeted metabolomics analysis in GBM cells showed that Ac4Glc2Bz disturbed glucose metabolism, amino acid pools, and nucleotide precursor biosynthesis, consistent with reduced proliferation and other anti-oncogenic properties of this analog. Furthermore, Ac4Glc2Bz reduced the invasion, migration, and stemness of GBM cells. Importantly, normal metabolic functions mediated by UDP-GlcNAc were not disrupted in non-neoplastic cells, including maintenance of endogenous levels of O-GlcNAcylation with no global disruption of N-glycan production. Finally, a pilot in vivo study showed that a potential therapeutic window exists where animals tolerated 5- to 10-fold higher levels of Ac4Glc2Bz than projected for in vivo efficacy. Together, these results establish GlcNAc analogs targeting the HBP through salvage mechanisms as a new therapeutic approach to safely normalize an important facet of aberrant glucose metabolism associated with cancer.
Collapse
Affiliation(s)
- Christopher T Saeui
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Sagar R Shah
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | | | - Cissy Zhang
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Christian Agatemor
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Kris Dammen-Brower
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Mohit P Mathew
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Matthew Buettner
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Prateek Gowda
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| | - Pratik Khare
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21205, United States
| | | | - Shuang Yang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Hui Zhang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
| | - Anne Le
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States.,Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21205, United States
| | | | - Kevin J Yarema
- Department of Biomedical Engineering and The Translational Tissue Engineering Center, The Johns Hopkins University and Johns Hopkins School of Medicine, Baltimore, Maryland 21231, United States
| |
Collapse
|
13
|
SERPINA3: Stimulator or Inhibitor of Pathological Changes. Biomedicines 2023; 11:biomedicines11010156. [PMID: 36672665 PMCID: PMC9856089 DOI: 10.3390/biomedicines11010156] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
SERPINA3, also called α-1-antichymotrypsin (AACT, ACT), is one of the inhibitors of serine proteases, one of which is cathepsin G. As an acute-phase protein secreted into the plasma by liver cells, it plays an important role in the anti-inflammatory response and antiviral response. Elevated levels of SERPINA3 have been observed in heart failure and neurological diseases such as Alzheimer's disease or Creutzfeldt-Jakob disease. Many studies have shown increased expression levels of the SERPINA3 gene in various types of cancer, such as glioblastoma, colorectal cancer, endometrial cancer, breast cancer, or melanoma. In this case, the SERPINA3 protein is associated with an antiapoptotic function implemented by adjusting the PI3K/AKT or MAPK/ERK 1/2 signal pathways. However, the functions of the SERPINA3 protein are still only partially understood, mainly in the context of cancerogenesis, so it seems necessary to summarize the available information and describe its mechanism of action. In particular, we sought to amass the existing body of research focusing on the description of the underlying mechanisms of various diseases not related to cancer. Our goal was to present an overview of the correct function of SERPINA3 as part of the defense system, which unfortunately easily becomes the "Fifth Column" and begins to support processes of destruction.
Collapse
|
14
|
Suarez-Meade P, Watanabe F, Ruiz-Garcia H, Rafferty SB, Moniz-Garcia D, Schiapparelli PV, Jentoft ME, Imitola J, Quinones-Hinojosa A. SARS-CoV2 entry factors are expressed in primary human glioblastoma and recapitulated in cerebral organoid models. J Neurooncol 2023; 161:67-76. [PMID: 36595192 PMCID: PMC9808689 DOI: 10.1007/s11060-022-04205-2] [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: 10/17/2022] [Accepted: 12/01/2022] [Indexed: 01/04/2023]
Abstract
PURPOSE Glioblastoma (GBM) is the most common and malignant primary brain tumor in adults with a median overall survival of only 14.6 months despite aggressive treatment. While immunotherapy has been successful in other cancers, its benefit has been proven elusive in GBM, mainly due to a markedly immunosuppressive tumor microenvironment. SARS-CoV-2 has been associated with the development of a pronounced central nervous system (CNS) inflammatory response when infecting different cells including astrocytes, endothelial cells, and microglia. While SARS-CoV2 entry factors have been described in different tissues, their presence and implication on GBM aggressiveness or microenvironment has not been studied on appropriate preclinical models. METHODS We evaluated the presence of crucial SARS-CoV-2 entry factors: ACE2, TMPRSS2, and NRP1 in matched surgically-derived GBM tissue, cells lines, and organoids; as well as in human brain derived specimens using immunohistochemistry, confocal pixel line intensity quantification, and transcriptome analysis. RESULTS We show that patient derived-GBM tissue and cell cultures express SARS-CoV2 entry factors, being NRP1 the most crucial facilitator of SARS-CoV-2 infection in GBM. Moreover, we demonstrate that, receptor expression remains present in our GBM organoids, making them an adequate model to study the effect of this virus in GBM for the potential development of viral therapies in the future. CONCLUSION Our findings suggest that the SARS-CoV-2 virus entry factors are expressed in primary tissues and organoid models and could be potentially utilized to study the susceptibility of GBM to this virus to target or modulate the tumor microenviroment.
Collapse
Affiliation(s)
- Paola Suarez-Meade
- Brain Tumor Stem Cell Laboratory, Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Fumihiro Watanabe
- Laboratory of Neural Stem Cells and Functional Neurogenetics, Departments of Neuroscience, Neurology, Genetics and Genome Sciences, UConn Health, Farmington, CT, 06030, USA
| | - Henry Ruiz-Garcia
- Brain Tumor Stem Cell Laboratory, Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Seamus B Rafferty
- Laboratory of Neural Stem Cells and Functional Neurogenetics, Departments of Neuroscience, Neurology, Genetics and Genome Sciences, UConn Health, Farmington, CT, 06030, USA
| | - Diogo Moniz-Garcia
- Brain Tumor Stem Cell Laboratory, Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Paula V Schiapparelli
- Brain Tumor Stem Cell Laboratory, Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, USA
| | - Mark E Jentoft
- Division of Anatomic Pathology, Mayo Clinic, Jacksonville, USA
| | - Jaime Imitola
- Laboratory of Neural Stem Cells and Functional Neurogenetics, Departments of Neuroscience, Neurology, Genetics and Genome Sciences, UConn Health, Farmington, CT, 06030, USA.
- Laboratory for Neural Stem Cells and Functional Neurogenetics, Division of Multiple Sclerosis and Neuroimmunology, Department of Neurology, UConn Health Comprehensive Multiple Sclerosis Center, UConn School of Medicine, 263 Farmington Avenue, Farmington, 06030, USA.
| | - Alfredo Quinones-Hinojosa
- Brain Tumor Stem Cell Laboratory, Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, USA.
- Neurologic Surgery, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, 32224, USA.
| |
Collapse
|
15
|
Melatonin Treatment Triggers Metabolic and Intracellular pH Imbalance in Glioblastoma. Cells 2022; 11:cells11213467. [DOI: 10.3390/cells11213467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Metabolic rewiring in glioblastoma (GBM) is linked to intra- and extracellular pH regulation. In this study, we sought to characterize the role of melatonin on intracellular pH modulation and metabolic consequences to identify the mechanisms of action underlying melatonin oncostatic effects on GBM tumor initiating cells. GBM tumor initiating cells were treated at different times with melatonin (1.5 and 3.0 mM). We analyzed melatonin’s functional effects on GBM proliferation, cell cycle, viability, stemness, and chemo-radiosensitivity. We then assessed the effects of melatonin on GBM metabolism by analyzing the mitochondrial and glycolytic parameters. We also measured the intracellular and extracellular pH. Finally, we tested the effects of melatonin on a mouse subcutaneous xenograft model. We found that melatonin downregulated LDHA and MCT4, decreasing lactate production and inducing a decrease in intracellular pH that was associated with an increase in ROS and ATP depletion. These changes blocked cell cycle progression and induced cellular death and we observed similar results in vivo. Melatonin’s cytotoxic effects on GBM were due, at least in part, to intracellular pH modulation, which has emerged as a newly identified mechanism, providing new insights into the oncostatic effect of melatonin on GBM.
Collapse
|
16
|
Zhou S, Wang H, Huang Y, Wu Y, Lin Z. The global change of gene expression pattern caused by PTEN mutation affects the prognosis of glioblastoma. Front Oncol 2022; 12:952521. [PMID: 36016609 PMCID: PMC9396408 DOI: 10.3389/fonc.2022.952521] [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: 05/25/2022] [Accepted: 06/30/2022] [Indexed: 12/01/2022] Open
Abstract
Glioblastoma (GBM), an aggressive primary tumor, is common in humans, accounting for 12–15% of all intracranial tumors, and has median survival of fewer than 15 months. Since a growing body of evidence suggests that conventional drugs are ineffective against GBM, our goal is to find emerging therapies that play a role in its treatment. This research constructs a risk model to predict the prognosis of GBM patients. A set of genes associated with GBM was taken from a GBM gene data bank, and clinical information on patients with GBM was retrieved from the Cancer Genome Atlas (TCGA) data bank. One-way Cox and Kaplan–Meier analyses were performed to identify genes in relation to prognosis. Groups were classified into high and low expression level of PTEN expression. Prognosis-related genes were further identified, and multi-factor Cox regression analysis was used to build risk score equations for the prognostic model to construct a survival prognostic model. The area under the ROC curve suggested that the pattern had high accuracy. When combined with nomogram analysis, GJB2 was considered an independent predictor of GBM prognosis. This study provides a potential prognostic predictive biological marker for GBM patients and confirms that GJB2 is a key gene for GBM progression.
Collapse
|
17
|
Fatima S, Gupta S, Khan AB, Rehman SU, Jairajpuri MA. Identification and validation of two alternatively spliced novel isoforms of human alpha-1-antichymotrypsin. Biochem Biophys Res Commun 2022; 628:25-31. [DOI: 10.1016/j.bbrc.2022.08.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 11/02/2022]
|
18
|
Norton ES, Whaley LA, Ulloa-Navas MJ, García-Tárraga P, Meneses KM, Lara-Velazquez M, Zarco N, Carrano A, Quiñones-Hinojosa A, García-Verdugo JM, Guerrero-Cázares H. Glioblastoma disrupts the ependymal wall and extracellular matrix structures of the subventricular zone. Fluids Barriers CNS 2022; 19:58. [PMID: 35821139 PMCID: PMC9277938 DOI: 10.1186/s12987-022-00354-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/27/2022] [Indexed: 12/04/2022] Open
Abstract
Background Glioblastoma (GBM) is the most aggressive and common type of primary brain tumor in adults. Tumor location plays a role in patient prognosis, with tumors proximal to the lateral ventricles (LVs) presenting with worse overall survival, increased expression of stem cell genes, and increased incidence of distal tumor recurrence. This may be due in part to interaction of GBM with factors of the subventricular zone (SVZ), including those contained within the cerebrospinal fluid (CSF). However, direct interaction of GBM tumors with CSF has not been proved and would be hindered in the presence of an intact ependymal cell layer. Methods Here, we investigate the ependymal cell barrier and its derived extracellular matrix (ECM) fractones in the vicinity of a GBM tumor. Patient-derived GBM cells were orthotopically implanted into immunosuppressed athymic mice in locations distal and proximal to the LV. A PBS vehicle injection in the proximal location was included as a control. At four weeks post-xenograft, brain tissue was examined for alterations in ependymal cell health via immunohistochemistry, scanning electron microscopy, and transmission electron microscopy. Results We identified local invading GBM cells within the LV wall and increased influx of CSF into the LV-proximal GBM tumor bulk compared to controls. In addition to the physical disruption of the ependymal cell barrier, we also identified increased signs of compromised ependymal cell health in LV-proximal tumor-bearing mice. These signs include increased accumulation of lipid droplets, decreased cilia length and number, and decreased expression of cell channel proteins. We additionally identified elevated numbers of small fractones in the SVZ within this group, suggesting increased indirect CSF-contained molecule signaling to tumor cells. Conclusions Our data is the first to show that LV-proximal GBMs physically disrupt the ependymal cell barrier in animal models, resulting in disruptions in ependymal cell biology and increased CSF interaction with the tumor bulk. These findings point to ependymal cell health and CSF-contained molecules as potential axes for therapeutic targeting in the treatment of GBM. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00354-8.
Collapse
Affiliation(s)
- Emily S Norton
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Lauren A Whaley
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.,Department of Biology, University of North Florida, Jacksonville, FL, USA
| | - María José Ulloa-Navas
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain.,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Patricia García-Tárraga
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain
| | - Kayleah M Meneses
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.,Biochemistry and Molecular Biology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | | | - Natanael Zarco
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Anna Carrano
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | | | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, Paterna, Spain
| | - Hugo Guerrero-Cázares
- Department of Neurosurgery, Mayo Clinic Florida, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|
19
|
Keep RF, Jones HC, Drewes LR. Advances in brain barriers and brain fluids research in 2021: great progress in a time of adversity. Fluids Barriers CNS 2022; 19:48. [PMID: 35681151 PMCID: PMC9178944 DOI: 10.1186/s12987-022-00343-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022] Open
Abstract
This editorial highlights advances in brain barrier and brain fluid research in 2021. It covers research on components of the blood–brain barrier, neurovascular unit and brain fluid systems; how brain barriers and brain fluid systems are impacted by neurological disorders and their role in disease progression; and advances in strategies for treating such disorders.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| | | | - Lester R Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN, 55812, USA
| |
Collapse
|
20
|
Jin Y, Wang W, Wang Q, Zhang Y, Zahid KR, Raza U, Gong Y. Alpha-1-antichymotrypsin as a novel biomarker for diagnosis, prognosis, and therapy prediction in human diseases. Cancer Cell Int 2022; 22:156. [PMID: 35439996 PMCID: PMC9019971 DOI: 10.1186/s12935-022-02572-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 04/06/2022] [Indexed: 12/15/2022] Open
Abstract
The glycoprotein alpha-1-antichymotrypsin (AACT), a serine protease inhibitor, is mainly synthesized in the liver and then secreted into the blood and is involved in the acute phase response, inflammation, and proteolysis. The dysregulation of AACT and its glycosylation levels are associated with tumor progression and recurrence, and could be used as a biomarker for tumor monitoring. In this review, we summarized the expression level, glycosylation modification, and biological characteristics of AACT during inflammation, neurodegenerative or other elderly diseases, and tumorigenesis, as well as, focused on the biological roles of AACT in cancer. The aberrant expression of AACT in cancer might be due to genetic alterations and/or immune by bioinformatics analysis. Moreover, AACT may serve as a diagnostic or prognostic biomarker or therapeutic target in tumors. Furthermore, we found that the expression of AACT was associated with the overall survival of patients with human cancers. Decreased AACT expression was associated with poor survival in patients with liver cancer, increased AACT expression was associated with shorter survival in patients with pancreatic cancer, and decreased AACT expression was associated with shorter survival in patients with early lung cancer. The review confirmed the key roles of AACT in tumorigenesis, suggesting that the glycoprotein AACT may serve as a biomarker for tumor diagnosis and prognosis, and could be a potential therapeutic target for human diseases.
Collapse
Affiliation(s)
- Yanxia Jin
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, No. 11 Cihu Road, Huangshi District, Huangshi, 435002, China
| | - Weidong Wang
- College of Life Sciences, Hubei Normal University, No. 11 Cihu Road, Huangshi District, Huangshi, 435002, China.
| | - Qiyun Wang
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, No. 11 Cihu Road, Huangshi District, Huangshi, 435002, China
| | - Yueyang Zhang
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, College of Life Sciences, Hubei Normal University, No. 11 Cihu Road, Huangshi District, Huangshi, 435002, China
| | - Kashif Rafiq Zahid
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Science and Oceanography, Carson International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Umar Raza
- Department of Biological Sciences, National University of Medical Sciences (NUMS), PWD Campus, Rawalpindi, Pakistan
| | - Yongsheng Gong
- Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, No.26 Daoqian Street, Suzhou, 215002, China.
| |
Collapse
|
21
|
Sun J, Wang L. HOXA-AS2 enhances GBM cell malignancy by suppressing miR-2116-3p thereby upregulating SERPINA3. BMC Cancer 2022; 22:366. [PMID: 35387643 PMCID: PMC8985346 DOI: 10.1186/s12885-022-09462-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
Background Glioblastoma (GBM) is malignant, demanding more attention to the improvement of the diagnosis and therapy. LncRNAs have been implicated in the malignancy of GBM cells. Methods HOXA-AS2, miR-2116-3p and SERPINA3 expression levels in GBM tissues and cell lines were detected by qRT-PCR. Western blotting was performed to detect the protein levels of Bax and Bcl-2. Dual-luciferase reporter assay was for detection of relationship among these factors, together with RIP and RNA pull-down. CCK-8, EdU, wound healing and transwell assays were for detection of the role of HOXA-AS2, miR-2116-3p and SERPINA3 in cell viability, proliferation, migration and invasion in GBM, respectively. Results HOXA-AS2 and SERPINA3 showed higher level in GBM tissues and cell lines. Low level of HOXA-AS2 attenuated GBM cell growth in vitro. Moreover, the anti-tumor impact of silenced HOXA-AS2 was restored by miR-2116-3p inhibitor, but its tumor-promotional effect could be reversed by silenced SERPINA3. Conclusion HOXA-AS2 enhanced GBM cell malignancy through sponging miR-2116-3p and releasing SERPINA3, which might shed light on the diagnosis and therapy for GBM in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09462-y.
Collapse
Affiliation(s)
- Jianrui Sun
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450052, Henan, China.
| | - Lin Wang
- Information Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| |
Collapse
|
22
|
Lara-Velazquez M, Mehkri Y, Panther E, Hernandez J, Rao D, Fiester P, Makary R, Rutenberg M, Tavanaiepour D, Rahmathulla G. Current Advances in the Management of Adult Craniopharyngiomas. Curr Oncol 2022; 29:1645-1671. [PMID: 35323338 PMCID: PMC8946973 DOI: 10.3390/curroncol29030138] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 12/23/2022] Open
Abstract
Craniopharyngiomas (CPs) are slow growing, histologically benign intracranial tumors located in the sellar–suprasellar region. Although known to have low mortality, their location and relationship to the adjacent neural structures results in patients having significant neurologic, endocrine, and visual comorbidities. The invasive nature of this tumor makes complete resection a challenge and contributes to its recurrence. Additionally, these tumors are bimodally distributed, being treated with surgery, and are followed by other adjuncts, such as focused radiation therapy, e.g., Gamma knife. Advances in surgical techniques, imaging tools, and instrumentations have resulted in the evolution of surgery using endoscopic techniques, with residual components being treated by radiotherapy to target the residual tumor. Advances in molecular biology have elucidated the main pathways involved in tumor development and recurrence, but presently, no other treatments are offered to patients, besides surgery, radiation, and endocrine management, as the disease and tumor evolve. We review the contemporary management of these tumors, from the evolution of surgical treatments, utilizing standard open microscopic approaches to the more recent endoscopic surgery, and discuss the current recommendations for care of these patients. We discuss the developments in radiation therapy, such as radiosurgery, being used as treatment strategies for craniopharyngioma, highlighting their beneficial effects on tumor resections while decreasing the rates of adverse outcomes. We also outline the recent chemotherapy modalities, which help control tumor growth, and the immune landscape on craniopharyngiomas that allow the development of novel immunotherapies.
Collapse
Affiliation(s)
- Montserrat Lara-Velazquez
- Department of Neurosurgery, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA; (M.L.-V.); (Y.M.); (E.P.); (J.H.); (D.T.)
| | - Yusuf Mehkri
- Department of Neurosurgery, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA; (M.L.-V.); (Y.M.); (E.P.); (J.H.); (D.T.)
| | - Eric Panther
- Department of Neurosurgery, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA; (M.L.-V.); (Y.M.); (E.P.); (J.H.); (D.T.)
| | - Jairo Hernandez
- Department of Neurosurgery, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA; (M.L.-V.); (Y.M.); (E.P.); (J.H.); (D.T.)
| | - Dinesh Rao
- Department of Neuroradiology, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA; (D.R.); (P.F.)
| | - Peter Fiester
- Department of Neuroradiology, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA; (D.R.); (P.F.)
| | - Raafat Makary
- Department of Pathology, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA;
| | - Michael Rutenberg
- Department of Radiation Oncology, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA;
| | - Daryoush Tavanaiepour
- Department of Neurosurgery, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA; (M.L.-V.); (Y.M.); (E.P.); (J.H.); (D.T.)
| | - Gazanfar Rahmathulla
- Department of Neurosurgery, College of Medicine, University of Florida, 653 8th St W., Jacksonville, FL 32209, USA; (M.L.-V.); (Y.M.); (E.P.); (J.H.); (D.T.)
- Correspondence: ; Tel.: +1-904-244-1418; Fax: +1-888-939-4093
| |
Collapse
|
23
|
Bhargav AG, Domino JS, Chamoun R, Thomas SM. Mechanical Properties in the Glioma Microenvironment: Emerging Insights and Theranostic Opportunities. Front Oncol 2022; 11:805628. [PMID: 35127517 PMCID: PMC8813748 DOI: 10.3389/fonc.2021.805628] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/29/2021] [Indexed: 12/30/2022] Open
Abstract
Gliomas represent the most common malignant primary brain tumors, and a high-grade subset of these tumors including glioblastoma are particularly refractory to current standard-of-care therapies including maximal surgical resection and chemoradiation. The prognosis of patients with these tumors continues to be poor with existing treatments and understanding treatment failure is required. The dynamic interplay between the tumor and its microenvironment has been increasingly recognized as a key mechanism by which cellular adaptation, tumor heterogeneity, and treatment resistance develops. Beyond ongoing lines of investigation into the peritumoral cellular milieu and microenvironmental architecture, recent studies have identified the growing role of mechanical properties of the microenvironment. Elucidating the impact of these biophysical factors on disease heterogeneity is crucial for designing durable therapies and may offer novel approaches for intervention and disease monitoring. Specifically, pharmacologic targeting of mechanical signal transduction substrates such as specific ion channels that have been implicated in glioma progression or the development of agents that alter the mechanical properties of the microenvironment to halt disease progression have the potential to be promising treatment strategies based on early studies. Similarly, the development of technology to measure mechanical properties of the microenvironment in vitro and in vivo and simulate these properties in bioengineered models may facilitate the use of mechanical properties as diagnostic or prognostic biomarkers that can guide treatment. Here, we review current perspectives on the influence of mechanical properties in glioma with a focus on biophysical features of tumor-adjacent tissue, the role of fluid mechanics, and mechanisms of mechanical signal transduction. We highlight the implications of recent discoveries for novel diagnostics, therapeutic targets, and accurate preclinical modeling of glioma.
Collapse
Affiliation(s)
- Adip G. Bhargav
- Department of Neurological Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| | - Joseph S. Domino
- Department of Neurological Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| | - Roukoz Chamoun
- Department of Neurological Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| | - Sufi M. Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
24
|
Zhang M, Zhou Z, Liu Z, Liu F, Zhao C. Exploring the potential biomarkers for prognosis of glioblastoma via weighted gene co-expression network analysis. PeerJ 2022; 10:e12768. [PMID: 35111402 PMCID: PMC8781321 DOI: 10.7717/peerj.12768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 12/17/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common malignant tumor in the central system with a poor prognosis. Due to the complexity of its molecular mechanism, the recurrence rate and mortality rate of GBM patients are still high. Therefore, there is an urgent need to screen GBM biomarkers to prove the therapeutic effect and improve the prognosis. RESULTS We extracted data from GBM patients from the Gene Expression Integration Database (GEO), analyzed differentially expressed genes in GEO and identified key modules by weighted gene co-expression network analysis (WGCNA). GSE145128 data was obtained from the GEO database, and the darkturquoise module was determined to be the most relevant to the GBM prognosis by WGCNA (r = - 0.62, p = 0.01). We performed enrichment analysis of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) to reveal the interaction activity in the selected modules. Then Kaplan-Meier survival curve analysis was used to extract genes closely related to GBM prognosis. We used Kaplan-Meier survival curves to analyze the 139 genes in the darkturquoise module, identified four genes (DARS/GDI2/P4HA2/TRUB1) associated with prognostic GBM. Low expression of DARS/GDI2/TRUB1 and high expression of P4HA2 had a poor prognosis. Finally, we used tumor genome map (TCGA) data, verified the characteristics of hub genes through Co-expression analysis, Drug sensitivity analysis, TIMER database analysis and GSVA analysis. We downloaded the data of GBM from the TCGA database, the results of co-expression analysis showed that DARS/GDI2/P4HA2/TRUB1 could regulate the development of GBM by affecting genes such as CDC73/CDC123/B4GALT1/CUL2. Drug sensitivity analysis showed that genes are involved in many classic Cancer-related pathways including TSC/mTOR, RAS/MAPK.TIMER database analysis showed DARS expression is positively correlated with tumor purity (cor = 0.125, p = 1.07e-02)), P4HA2 expression is negatively correlated with tumor purity (cor =-0.279, p = 6.06e-09). Finally, GSVA analysis found that DARS/GDI2/P4HA2/TRUB1 gene sets are closely related to the occurrence of cancer. CONCLUSION We used two public databases to identify four valuable biomarkers for GBM prognosis, namely DARS/GDI2/P4HA2/TRUB1, which have potential clinical application value and can be used as prognostic markers for GBM.
Collapse
Affiliation(s)
- Mengyuan Zhang
- Department of Neurology and Stroke Center, The First Hospital of China Medical University, Shenyang, China
| | - Zhike Zhou
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, China
| | - Zhouyang Liu
- Department of Neurology and Stroke Center, The First Hospital of China Medical University, Shenyang, China
| | - Fangxi Liu
- Department of Neurology and Stroke Center, The First Hospital of China Medical University, Shenyang, China
| | - Chuansheng Zhao
- Department of Neurology and Stroke Center, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
25
|
Lou D, Zhu D, Wang Z, Zhang R, Yu Z, Gong F, Peng Y, Zeng S, Liu Y, Li A, Fan Q. Effect of GADD45G on the radioresistance of nasopharyngeal carcinoma cells. Anticancer Drugs 2022; 33:e84-e93. [PMID: 34282742 DOI: 10.1097/cad.0000000000001145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The development of radioresistance by nasopharyngeal carcinoma (NPC) cells almost always results in tumor recurrence and metastasis, making clinical treatment of the disease difficult. In this study, the mechanism of radioresistance in NPC cells was investigated. First, a gene array and quantitative reverse-transcription-PCR assays were used to screen for genes exhibiting significantly altered expression in the DNA damage signaling pathway. Based on those results, GADD45G was further studied in the context of radioresistance. A GADD45G-knockout NPC cell line (CNE-2R-KO) was constructed using CRISPR-Cas9 technology and used for a comparison of differences in radioresistance with other radiosensitive and radioresistant NPC cells, as evaluated using colony formation assays. Cell cycle changes were observed using flow cytometry. Cell proliferation and migration were measured using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide and wound healing assays, respectively. The sequencing results revealed the successful construction of the CNE-2R-KO cell line, the radiosensitivity of which was higher than that of its parent radioresistant cell line owing to the GADD45G knockout. This was likely related to the increase in the number of cells in the G1 phase and decrease in those in the S1 phase as well as the increased cell proliferation rate and decreased migratory ability. GADD45G is associated with radioresistance in NPC cells and likely has a role in the occurrence and metastasis of NPC.
Collapse
Affiliation(s)
- Dandan Lou
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Daoqi Zhu
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Zetai Wang
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Ruhua Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center
| | - Zhijian Yu
- Department of Traditional Chinese Medicine Formulae, School of Traditional Chinese Medicine, Southern Medical University
| | - Fengying Gong
- Department of Traditional Chinese Medicine, NanFang Hospital, Guangdong, Guangzhou, China
| | - Yan Peng
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Siying Zeng
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| | - Ying Liu
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou
| | - Aiwu Li
- Department of Traditional Chinese Medicine, NanFang Hospital, Guangdong, Guangzhou, China
| | - Qin Fan
- TCM Molecular Biology Laboratory, School of Traditional Chinese Medicine, Southern Medical University
| |
Collapse
|
26
|
Norton ES, Da Mesquita S, Guerrero-Cazares H. SERPINA3 in glioblastoma and Alzheimer's disease. Aging (Albany NY) 2021; 13:21812-21813. [PMID: 34587119 PMCID: PMC8507276 DOI: 10.18632/aging.203603] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/24/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Emily S Norton
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Hugo Guerrero-Cazares
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
27
|
Brockman AA, Mobley BC, Ihrie RA. Histological Studies of the Ventricular-Subventricular Zone as Neural Stem Cell and Glioma Stem Cell Niche. J Histochem Cytochem 2021; 69:819-834. [PMID: 34310246 DOI: 10.1369/00221554211032003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The neural stem cell niche of the ventricular-subventricular zone supports the persistence of stem and progenitor cells in the mature brain. This niche has many notable cytoarchitectural features that affect the activity of stem cells and may also support the survival and growth of invading tumor cells. Histochemical studies of the niche have revealed many proteins that, in combination, can help to reveal stem-like cells in the normal or cancer context, although many caveats persist in the quest to consistently identify these cells in the human brain. Here, we explore the complex relationship between the persistent proliferative capacity of the neural stem cell niche and the malignant proliferation of brain tumors, with a special focus on histochemical identification of stem cells and stem-like tumor cells and an eye toward the potential application of high-dimensional imaging approaches to the field.
Collapse
Affiliation(s)
- Asa A Brockman
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Bret C Mobley
- Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rebecca A Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee.,Departments of Neurological Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
28
|
Ripari LB, Norton ES, Bodoque-Villar R, Jeanneret S, Lara-Velazquez M, Carrano A, Zarco N, Vazquez-Ramos CA, Quiñones-Hinojosa A, de la Rosa-Prieto C, Guerrero-Cázares H. Glioblastoma Proximity to the Lateral Ventricle Alters Neurogenic Cell Populations of the Subventricular Zone. Front Oncol 2021; 11:650316. [PMID: 34268110 PMCID: PMC8277421 DOI: 10.3389/fonc.2021.650316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 06/07/2021] [Indexed: 12/01/2022] Open
Abstract
Despite current strategies combining surgery, radiation, and chemotherapy, glioblastoma (GBM) is the most common and aggressive malignant primary brain tumor in adults. Tumor location plays a key role in the prognosis of patients, with GBM tumors located in close proximity to the lateral ventricles (LVs) resulting in worse survival expectancy and higher incidence of distal recurrence. Though the reason for worse prognosis in these patients remains unknown, it may be due to proximity to the subventricular zone (SVZ) neurogenic niche contained within the lateral wall of the LVs. We present a novel rodent model to analyze the bidirectional signaling between GBM tumors and cells contained within the SVZ. Patient-derived GBM cells expressing GFP and luciferase were engrafted at locations proximal, intermediate, and distal to the LVs in immunosuppressed mice. Mice were either sacrificed after 4 weeks for immunohistochemical analysis of the tumor and SVZ or maintained for survival analysis. Analysis of the GFP+ tumor bulk revealed that GBM tumors proximal to the LV show increased levels of proliferation and tumor growth than LV-distal counterparts and is accompanied by decreased median survival. Conversely, numbers of innate proliferative cells, neural stem cells (NSCs), migratory cells and progenitors contained within the SVZ are decreased as a result of GBM proximity to the LV. These results indicate that our rodent model is able to accurately recapitulate several of the clinical aspects of LV-associated GBM, including increased tumor growth and decreased median survival. Additionally, we have found the neurogenic and cell division process of the SVZ in these adult mice is negatively influenced according to the presence and proximity of the tumor mass. This model will be invaluable for further investigation into the bidirectional signaling between GBM and the neurogenic cell populations of the SVZ.
Collapse
Affiliation(s)
- Luisina B. Ripari
- Department of Medical Sciences, Facultad de Medicina de Albacete, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Emily S. Norton
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, United States
- Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, United States
- Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Raquel Bodoque-Villar
- Translational Research Unit, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Stephanie Jeanneret
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, United States
- Faculty of Psychology and Sciences of Education, University of Geneva, Geneva, Switzerland
| | | | - Anna Carrano
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, United States
| | - Natanael Zarco
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, United States
| | | | | | - Carlos de la Rosa-Prieto
- Department of Medical Sciences, Facultad de Medicina de Albacete, Universidad de Castilla-La Mancha, Albacete, Spain
| | | |
Collapse
|
29
|
Lathia JD. Fountain of chaos: cerebrospinal fluid enhancement of cancer stem cells in glioblastoma. Neuro Oncol 2021; 23:530-532. [PMID: 33749775 DOI: 10.1093/neuonc/noab026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Justin D Lathia
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case, Western Reserve University, Cleveland, Ohio, USA.,Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
30
|
Carrano A, Zarco N, Phillipps J, Lara-Velazquez M, Suarez-Meade P, Norton ES, Chaichana KL, Quiñones-Hinojosa A, Asmann YW, Guerrero-Cázares H. Human Cerebrospinal Fluid Modulates Pathways Promoting Glioblastoma Malignancy. Front Oncol 2021; 11:624145. [PMID: 33747938 PMCID: PMC7969659 DOI: 10.3389/fonc.2021.624145] [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: 10/30/2020] [Accepted: 02/05/2021] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma (GBM) is the most common and devastating primary cancer of the central nervous system in adults. High grade gliomas are able to modify and respond to the brain microenvironment. When GBM tumors infiltrate the Subventricular zone (SVZ) they have a more aggressive clinical presentation than SVZ-distal tumors. We suggest that cerebrospinal fluid (CSF) contact contributes to enhance GBM malignant characteristics in these tumors. We evaluated the impact of human CSF on GBM, performing a transcriptome analysis on human primary GBM cells exposed to CSF to measure changes in gene expression profile and their clinical relevance on disease outcome. In addition we evaluated the proliferation and migration changes of CSF-exposed GBM cells in vitro and in vivo. CSF induced transcriptomic changes in pathways promoting cell malignancy, such as apoptosis, survival, cell motility, angiogenesis, inflammation, and glucose metabolism. A genetic signature extracted from the identified transcriptional changes in response to CSF proved to be predictive of GBM patient survival using the TCGA database. Furthermore, CSF induced an increase in viability, proliferation rate, and self-renewing capacity, as well as the migratory capabilities of GBM cells in vitro. In vivo, GBM cells co-injected with human CSF generated larger and more proliferative tumors compared to controls. Taken together, these results provide direct evidence that CSF is a key player in determining tumor growth and invasion through the activation of complex gene expression patterns characteristic of a malignant phenotype. These findings have diagnostic and therapeutic implications for GBM patients. The changes induced by CSF contact might play a role in the increased malignancy of SVZ-proximal GBM.
Collapse
Affiliation(s)
- Anna Carrano
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | - Natanael Zarco
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | - Jordan Phillipps
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Paola Suarez-Meade
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | - Emily S Norton
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biochemical Sciences, Mayo Clinic, Jacksonville, FL, United States.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Kaisorn L Chaichana
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Yan W Asmann
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, United States
| | | |
Collapse
|