1
|
Li M, Nie H, He Q, Zhang Z, Yu S, Wang T, Fu B. Astrocytic Gap Junctions protein Cx43/Cx30 modulate EAAT1 and glutamate to mediate cerebral ischemia-reperfusion injury. Brain Res 2025; 1847:149306. [PMID: 39491663 DOI: 10.1016/j.brainres.2024.149306] [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: 08/06/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
The gap connexins of astrocytes play a crucial role in facilitating neuronal coordination and maintaining the homeostasis of the central nervous system. Cx30/Cx43 are the main proteins constituting these gap junctions, and the glutamate transporter EAAT1 associates with nerve injury. However, the role and mechanism underlying the changes of astrocytic connexins and EAAT1 during cerebral ischemia-reperfusion injury remain unclear. In this study, we investigated the expressions of Cx30, Cx43, and EAAT1 in OGD/R-treated astrocytes and in a MCAO/R animal model using gap junction inhibitors and siRNAs targeting Cx43 and Cx30. The differences of cell viability, malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), reactive oxygen species (ROS) and glutamate in cells and tissues were detected. Our results indicate that OGD/R exposure leads to the decline of astrocyte activity, which, in turn, adversely affects neuronal health. Ischemia-reperfusion induced increasing Cx43 and EAAT1 expression and decreasing Cx30 expression in astrocytes and animal brain tissue. Moreover, ischemia-reperfusion resulted in heightened MDA and ROS levels and reduced CAT and SOD activities in both astrocytes and the surrounding brain tissue. The release of glutamate from astrocytes and its concentration in animal brain tissue significantly increased following ischemia-reperfusion. Inhibition Cx43 expression through Gap26 or siRNA effectively mitigated the increase in EAAT1 and glutamate levels, as well as the oxidative stress changes induced by ischemia-reperfusion. Therefore, Brain astrocytes may mediate the effects of cerebral ischemia-reperfusion injury by influencing glutamate transporters and glutamate dynamics in response to oxidative stress through Cx30/Cx43.
Collapse
Affiliation(s)
- Min Li
- Rehabilitation Medicine Department of the Second People's Hospital Lianyungang, PR China
| | - Hongxia Nie
- Neurology Department of the Second People's Hospital Lianyungang, PR China
| | - Qianqian He
- Neurology Department of the Second People's Hospital Lianyungang, PR China
| | - Zhaoting Zhang
- Neurology Department of the Second People's Hospital Lianyungang, PR China
| | - Shanhua Yu
- Neurology Department of the Second People's Hospital Lianyungang, PR China
| | - Tiantian Wang
- Rehabilitation Medicine Department of the Second People's Hospital Lianyungang, PR China
| | - Bing Fu
- Neurology Department of the Second People's Hospital Lianyungang, PR China.
| |
Collapse
|
2
|
Suh HN, Choi GE. Wnt signaling in the tumor microenvironment: A driver of brain tumor dynamics. Life Sci 2024; 358:123174. [PMID: 39471897 DOI: 10.1016/j.lfs.2024.123174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
The Wnt signaling pathway is important for cell growth and development in the central nervous system and its associated vasculature. Thus, it is an interesting factor for establishing anti-brain cancer therapy. However, simply inhibiting the Wnt signaling pathway in patients with brain tumors is not an effective anti-cancer therapy. Due to their complex microenvironment, which comprises various cell types and signaling molecules, brain tumors pose significant challenges. It is important to understand the interplay between tumor cells and the microenvironment for developing effective therapeutic strategies for both benign and malignant brain tumors. Thus, this research focused on the role of the tumor microenvironment (TME) in brain tumor progression, particularly the involvement of Wnt-dependent signaling pathways. The brain parenchyma comprises neurons, glia, endothelial cells, and other extracellular matrix elements that can contribute to the TME. The TME components can secrete Wnt ligands or associated molecules, resulting in the aberrant activation of the Wnt signaling pathway, followed by tumor progression and therapeutic resistance. Therefore, it is essential to understand the intricate crosstalk between the Wnt signaling pathway and the TME in developing targeted therapies. This review aimed to elucidate the complexities of the brain TME and its interactions with the Wnt signaling pathways to improve treatment outcomes and our understanding of brain tumor biology.
Collapse
Affiliation(s)
- Han Na Suh
- Center for Translational Toxicologic Research, Korea Institute of Toxicology, 30 Baekhak1-gil, Jeongeup, Jeonbukdo 56212, Republic of Korea.
| | - Gee Euhn Choi
- Laboratory of Veterinary Biochemistry, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju 63243, South Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, South Korea.
| |
Collapse
|
3
|
Hatlen RR, Rajagopalan P. Investigating Trans-differentiation of Glioblastoma Cells in an In Vitro 3D Model of the Perivascular Niche. ACS Biomater Sci Eng 2023. [PMID: 37129167 DOI: 10.1021/acsbiomaterials.2c01310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Glioblastoma multiforme (GBM) is the deadliest form of brain cancer, responsible for over 50% of adult brain tumors. A specific region within the GBM environment is known as the perivascular niche (PVN). This area is defined as within approximately 100 μm of vasculature and plays an important role in the interactions between endothelial cells (ECs), astrocytes, GBM cells, and stem cells. We have designed a 3D in vitro model of the PVN comprising either collagen Type 1 or HyStem-C, human umbilical vein ECs (HUVECs), and LN229 (GBM) cells. HUVECs were encapsulated within the hydrogels to form vascular networks. After 7 days, LN229 cells were co-cultured to investigate changes in both cell types. Over a 14 day culture period, we measured alterations in HUVEC networks, the contraction of the hydrogels, trans-differentiation of LN229 cells, and the concentrations of two chemokines; CXCL12 and TGF-β. Increased cellular proliferation ranging from 10- to 16-fold was exhibited in co-cultures from days 8 to 14. This was accompanied with a decrease in the height of hydrogels of up to 68%. These changes in the biomaterial scaffold indicate that LN229-HUVEC interactions promote changes to the matrix. TGF-β and CXCL12 secretion increased approximately 2-2.6-fold each from day 8 to 14 in all co-cultures. The expression of CXCL12 correlated with cell colocalization, indicating a chemotactic role in enabling the migration of LN229 cells toward HUVECs in co-cultures. von Willebrand factor (vWF) was co-expressed with glial fibrillary acidic protein (GFAP) in up to 15% of LN229 cells after 24 h in co-culture. Additionally, when LN229 cells were co-cultured with human brain microvascular ECs, the percentages of GFAP+/vWF+ cells were up to 20% higher than that in co-cultures with HUVECs in collagen (2.2 mg/mL) and HyStem-C gels on day 14. The expression of vWF indicates the early stages of trans-differentiation of LN229 cells to an EC phenotype. Designing in vitro models of trans-differentiation may provide additional insights into how vasculature and cellular phenotypes are altered in GBM.
Collapse
Affiliation(s)
- Rosalyn R Hatlen
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
4
|
Hatlen RR, Rajagopalan P. Environmental interplay: Stromal cells and biomaterial composition influence in the glioblastoma microenvironment. Acta Biomater 2021; 132:421-436. [PMID: 33276155 DOI: 10.1016/j.actbio.2020.11.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
Glioblastoma multiforme (GBM) is the most deadly form of brain cancer. Recurrence is common, and established therapies have not been able to significantly extend overall patient survival. One platform through which GBM research can progress is to design biomimetic systems for discovery and investigation into the mechanisms of invasion, cellular properties, as well as the efficacy of therapies. In this review, 2D and 3D GBM in vitro cultures will be discussed. We focus on the effects of biomaterial properties, interactions between stromal cells, and vascular influence on cancer cell survival and progression. This review will summarize critical findings in each of these areas and how they have led to a more comprehensive scientific understanding of GBM. STATEMENT OF SIGNIFICANCE: Glioblastoma multiforme (GBM) is the most deadly form of brain cancer. Recurrence is common, and established therapies have not been able to significantly extend overall patient survival. One platform through which GBM research can progress is to design biomimetic systems for discovery and investigation into the mechanisms of invasion, cellular properties, as well as the efficacy of therapies. In this review, 2D and 3D GBM in vitro cultures will be discussed. We focus on the effects of biomaterial properties, interactions between stromal cells and vascular influence on cancer cell survival and progression. This review will summarize critical findings in each of these areas and how they have lead to a more comprehensive scientific understanding of GBM.
Collapse
Affiliation(s)
- Rosalyn R Hatlen
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, United States
| | | |
Collapse
|
5
|
Chou FJ, Liu Y, Lang F, Yang C. D-2-Hydroxyglutarate in Glioma Biology. Cells 2021; 10:cells10092345. [PMID: 34571995 PMCID: PMC8464856 DOI: 10.3390/cells10092345] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Isocitrate dehydrogenase (IDH) mutations are common genetic abnormalities in glioma, which result in the accumulation of an "oncometabolite", D-2-hydroxyglutarate (D-2-HG). Abnormally elevated D-2-HG levels result in a distinctive pattern in cancer biology, through competitively inhibiting α-ketoglutarate (α-KG)/Fe(II)-dependent dioxgenases (α-KGDDs). Recent studies have revealed that D-2-HG affects DNA/histone methylation, hypoxia signaling, DNA repair, and redox homeostasis, which impacts the oncogenesis of IDH-mutated cancers. In this review, we will discuss the current understanding of D-2-HG in cancer biology, as well as the emerging opportunities in therapeutics in IDH-mutated glioma.
Collapse
|
6
|
Characterization of Hevin (SPARCL1) Immunoreactivity in Postmortem Human Brain Homogenates. Neuroscience 2021; 467:91-109. [PMID: 34033869 DOI: 10.1016/j.neuroscience.2021.05.017] [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: 03/12/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 11/22/2022]
Abstract
Hevin is a matricellular glycoprotein that plays important roles in neural developmental processes such as neuronal migration, synaptogenesis and synaptic plasticity. In contrast to other matricellular proteins whose expression decreases when development is complete, hevin remains highly expressed, suggesting its involvement in adult brain function. In vitro studies have shown that hevin can have different post-translational modifications. However, the glycosylation pattern of hevin in the human brain remains unknown, as well as its relative distribution and localization. The present study provides the first thorough characterization of hevin protein expression by Western blot in postmortem adult human brain. Our results demonstrated two major specific immunoreactive bands for hevin: an intense band migrating around 130 kDa, and a band migrating around 100 kDa. Biochemical assays revealed that both hevin bands have a different glycosylation pattern. Subcellular fractionation showed greater expression in membrane-enriched fraction than in cytosolic preparation, and a higher expression in prefrontal cortex (PFC) compared to hippocampus (HIP), caudate nucleus (CAU) and cerebellum (CB). We confirmed that a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) and matrixmetalloproteinase 3 (MMP-3) proteases digestion led to an intense double band with similar molecular weight to that described as SPARC-like fragment (SLF). Finally, hevin immunoreactivity was also detected in human astrocytoma, meningioma, cerebrospinal fluid and serum samples, but was absent from any blood cell type.
Collapse
|
7
|
Brenet M, Martínez S, Pérez-Nuñez R, Pérez LA, Contreras P, Díaz J, Avalos AM, Schneider P, Quest AFG, Leyton L. Thy-1 (CD90)-Induced Metastatic Cancer Cell Migration and Invasion Are β3 Integrin-Dependent and Involve a Ca 2+/P2X7 Receptor Signaling Axis. Front Cell Dev Biol 2021; 8:592442. [PMID: 33511115 PMCID: PMC7835543 DOI: 10.3389/fcell.2020.592442] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/04/2020] [Indexed: 01/21/2023] Open
Abstract
Cancer cell adhesion to the vascular endothelium is an important step in tumor metastasis. Thy-1 (CD90), a cell adhesion molecule expressed in activated endothelial cells, has been implicated in melanoma metastasis by binding to integrins present in cancer cells. However, the signaling pathway(s) triggered by this Thy-1-Integrin interaction in cancer cells remains to be defined. Our previously reported data indicate that Ca2+-dependent hemichannel opening, as well as the P2X7 receptor, are key players in Thy-1-αVβ3 Integrin-induced migration of reactive astrocytes. Thus, we investigated whether this signaling pathway is activated in MDA-MB-231 breast cancer cells and in B16F10 melanoma cells when stimulated with Thy-1. In both cancer cell types, Thy-1 induced a rapid increase in intracellular Ca2+, ATP release, as well as cell migration and invasion. Connexin and Pannexin inhibitors decreased cell migration, implicating a requirement for hemichannel opening in Thy-1-induced cell migration. In addition, cell migration and invasion were precluded when the P2X7 receptor was pharmacologically blocked. Moreover, the ability of breast cancer and melanoma cells to transmigrate through an activated endothelial monolayer was significantly decreased when the β3 Integrin was silenced in these cancer cells. Importantly, melanoma cells with silenced β3 Integrin were unable to metastasize to the lung in a preclinical mouse model. Thus, our results suggest that the Ca2+/hemichannel/ATP/P2X7 receptor-signaling axis triggered by the Thy-1-αVβ3 Integrin interaction is important for cancer cell migration, invasion and transvasation. These findings open up the possibility of therapeutically targeting the Thy-1-Integrin signaling pathway to prevent metastasis.
Collapse
Affiliation(s)
- Marianne Brenet
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Samuel Martínez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ramón Pérez-Nuñez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Leonardo A Pérez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Pamela Contreras
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Jorge Díaz
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Andrew F G Quest
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies of Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
8
|
Xing L, Yang T, Cui S, Chen G. Connexin Hemichannels in Astrocytes: Role in CNS Disorders. Front Mol Neurosci 2019; 12:23. [PMID: 30787868 PMCID: PMC6372977 DOI: 10.3389/fnmol.2019.00023] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
In the central nervous system (CNS), astrocytes form networks interconnected by gap junctions made from connexins of the subtypes Cx30 and Cx43. When unopposed by an adjoining hemichannel, astrocytic connexins can act as hemichannels to control the release of small molecules such as ATP and glutamate into the extracellular space. Accruing evidence indicates that astrocytic connexins are crucial for the coordination and maintenance of physiologic CNS activity. Here we provide an update on the role of astrocytic connexins in neurodegenerative disorders, glioma, and ischemia. In addition, we address the regulation of Cx43 in chronic pain.
Collapse
Affiliation(s)
- LingYan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - ShuSen Cui
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Gang Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
9
|
Towner RA, Smith N, Saunders D, Brown CA, Cai X, Ziegler J, Mallory S, Dozmorov MG, Coutinho De Souza P, Wiley G, Kim K, Kang S, Kong DS, Kim YT, Fung KM, Wren JD, Battiste J. OKN-007 Increases temozolomide (TMZ) Sensitivity and Suppresses TMZ-Resistant Glioblastoma (GBM) Tumor Growth. Transl Oncol 2019; 12:320-335. [PMID: 30468988 PMCID: PMC6251232 DOI: 10.1016/j.tranon.2018.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 02/06/2023] Open
Abstract
Treatment of glioblastoma (GBM) remains a challenge using conventional chemotherapy, such as temozolomide (TMZ), and is often ineffective as a result of drug resistance. We have assessed a novel nitrone-based agent, OKN-007, and found it to be effective in decreasing tumor volumes and increasing survival in orthotopic GBM xenografts by decreasing cell proliferation and angiogenesis and increasing apoptosis. In this study, we assessed combining OKN-007 with TMZ in vivo in a human G55 GBM orthotopic xenograft model and in vitro in TMZ-resistant and TMZ-sensitive human GBM cell lines. For the in vivo studies, magnetic resonance imaging was used to assess tumor growth and vascular alterations. Percent animal survival was also determined. For the in vitro studies, cell growth, IC50 values, RNA-seq, RT-PCR, and ELISA were used to assess growth inhibition, possible mechanism-of actions (MOAs) associated with combined OKN-007 + TMZ versus TMZ alone, and gene and protein expression levels, respectively. Microarray analysis of OKN-007-treated rat F98 glioma tumors was also carried out to determine possible MOAs of OKN-007 in glioma-bearing animals either treated or not treated with OKN-007. OKN-007 seems to elicit its effect on GBM tumors via inhibition of tumorigenic TGF-β1, which affects the extracellular matrix. When combined with TMZ, OKN-007 significantly increases percent survival, decreases tumor volumes, and normalizes tumor blood vasculature in vivo compared to untreated tumors and seems to affect TMZ-resistant GBM cells possibly via IDO-1, SUMO2, and PFN1 in vitro. Combined OKN-007 + TMZ may be a potentially potent treatment strategy for GBM patients.
Collapse
Affiliation(s)
- Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of PathologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Chase A Brown
- Arthritis and Clinical Immunology Research Program, Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xue Cai
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jadith Ziegler
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of PathologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Graham Wiley
- Clinical Genomics Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kyeongsoon Kim
- Department of Pharmaceutical Engineering, Inje University, Gimhae-si, Gyeongsangnam-do, Republic of Korea; Oblato, Inc., Princeton, NJ, USA
| | | | - Doo-Sik Kong
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Young-Tae Kim
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Kar-Ming Fung
- Department of PathologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jonathan D Wren
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Arthritis and Clinical Immunology Research Program, Division of Genomics and Data Sciences, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - James Battiste
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of NeurologyUniversity of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
10
|
Zhang X, Ding K, Wang J, Li X, Zhao P. Chemoresistance caused by the microenvironment of glioblastoma and the corresponding solutions. Biomed Pharmacother 2018; 109:39-46. [PMID: 30391707 DOI: 10.1016/j.biopha.2018.10.063] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/03/2018] [Accepted: 10/12/2018] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary human brain tumor. Although comprehensive therapies combining radiotherapy and chemotherapy after surgery can prolong survival, the prognosis is still poor with a median survival of only 14.6 months. Chemoresistance is one of the major causes of relapse as well as poor survival in glioma patients. Therefore, novel strategies to overcome chemoresistance are desperately needed for improved treatment of human GBM. Recent studies have demonstrated that the tumor microenvironment plays a critical role in the chemoresistance of various tumor types, which makes it a suitable target in anti-cancer therapies, as well as a valuable biomarker for prognostic purposes. This review focuses on chemoresistance in GBM induced by stromal cells, including the endothelium of blood vessels, astrocytes, and myeloid cells, as well as non-cellular factors in the tumor microenvironment. Corresponding therapies are discussed, including progressive strategies involving 3-dimensional models integrating engineering as well as biological advances.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Kaikai Ding
- Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, PR China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China; Department of Biomedicine, University of Bergen, 5009, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China
| | - Peng Zhao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Institute of Brain and Brain-Inspired Science, Shandong University, PR China; Shandong Key Laboratory of Brain Function Remodeling, PR China.
| |
Collapse
|
11
|
Qi XT, Zhan JS, Xiao LM, Li L, Xu HX, Fu ZB, Zhang YH, Zhang J, Jia XH, Ge G, Chai RC, Gao K, Yu ACH. The Unwanted Cell Migration in the Brain: Glioma Metastasis. Neurochem Res 2017; 42:1847-1863. [PMID: 28478595 DOI: 10.1007/s11064-017-2272-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/12/2017] [Accepted: 04/17/2017] [Indexed: 12/19/2022]
Abstract
Cell migration is identified as a highly orchestrated process. It is a fundamental and essential phenomenon underlying tissue morphogenesis, wound healing, and immune response. Under dysregulation, it contributes to cancer metastasis. Brain is considered to be the most complex organ in human body containing many types of neural cells with astrocytes playing crucial roles in monitoring both physiological and pathological functions. Astrocytoma originates from astrocytes and its most malignant type is glioblastoma multiforme (WHO Grade IV astrocytoma), which is capable to infiltrate widely into the neighboring brain tissues making a complete resection of tumors impossible. Very recently, we have reviewed the mechanisms for astrocytes in migration. Given the fact that astrocytoma shares many histological features with astrocytes, we therefore attempt to review the mechanisms for glioma cells in migration and compare them to normal astrocytes, hoping to obtain a better insight into the dysregulation of migratory mechanisms contributing to their metastasis in the brain.
Collapse
Affiliation(s)
- Xue Tao Qi
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Jiang Shan Zhan
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Li Ming Xiao
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Lina Li
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China.
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China.
- Hai Kang Life (Beijing) Corporation Ltd., Sino-I Campus No.1, Beijing Economic-Technological Development Area, Beijing, 100176, China.
- Hai Kang Life Corporation Ltd., Hong Kong Science Park, Shatin, New Territories, Hong Kong, China.
| | - Han Xiao Xu
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Department of Human Anatomy, Guizhou Medical University, Guian New Area, Guiyang, Guizhou, 550025, China
| | - Zi Bing Fu
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Yan Hao Zhang
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
| | - Jing Zhang
- Department of Pathology, Peking University Health Science Center and Peking University Third Hospital, Beijing, 100191, China
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Xi Hua Jia
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Hai Kang Life (Beijing) Corporation Ltd., Sino-I Campus No.1, Beijing Economic-Technological Development Area, Beijing, 100176, China
- Hai Kang Life Corporation Ltd., Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
| | - Guo Ge
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Department of Human Anatomy, Guizhou Medical University, Guian New Area, Guiyang, Guizhou, 550025, China
| | - Rui Chao Chai
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Hai Kang Life (Beijing) Corporation Ltd., Sino-I Campus No.1, Beijing Economic-Technological Development Area, Beijing, 100176, China
- Hai Kang Life Corporation Ltd., Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
| | - Kai Gao
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Albert Cheung Hoi Yu
- Laboratory for Functional Study of Astrocytes, Neuroscience Research Institute, Peking University, 38 Xue Yuan Road, Beijing, 100191, China.
- Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Key Laboratory for Neuroscience, Ministry of Education, Peking University Health Science Center, Beijing, 100191, China.
- National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China.
- Hai Kang Life (Beijing) Corporation Ltd., Sino-I Campus No.1, Beijing Economic-Technological Development Area, Beijing, 100176, China.
- Hai Kang Life Corporation Ltd., Hong Kong Science Park, Shatin, New Territories, Hong Kong, China.
- Laboratory of Translational Medicine, Institute of Systems Biomedicine, Peking University, Beijing, 100191, China.
| |
Collapse
|
12
|
Okolie O, Bago JR, Schmid RS, Irvin DM, Bash RE, Miller CR, Hingtgen SD. Reactive astrocytes potentiate tumor aggressiveness in a murine glioma resection and recurrence model. Neuro Oncol 2016; 18:1622-1633. [PMID: 27298311 DOI: 10.1093/neuonc/now117] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 05/04/2016] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Surgical resection is a universal component of glioma therapy. Little is known about the postoperative microenvironment due to limited preclinical models. Thus, we sought to develop a glioma resection and recurrence model in syngeneic immune-competent mice to understand how surgical resection influences tumor biology and the local microenvironment. METHODS We genetically engineered cells from a murine glioma mouse model to express fluorescent and bioluminescent reporters. Established allografts were resected using image-guided microsurgery. Postoperative tumor recurrence was monitored by serial imaging, and the peritumoral microenvironment was characterized by histopathology and immunohistochemistry. Coculture techniques were used to explore how astrocyte injury influences tumor aggressiveness in vitro. Transcriptome and secretome alterations in injured astrocytes was examined by RNA-seq and Luminex. RESULTS We found that image-guided resection achieved >90% reduction in tumor volume but failed to prevent both local and distant tumor recurrence. Immunostaining for glial fibrillary acidic protein and nestin showed that resection-induced injury led to temporal and spatial alterations in reactive astrocytes within the peritumoral microenvironment. In vitro, we found that astrocyte injury induced transcriptome and secretome alterations and promoted tumor proliferation, as well as migration. CONCLUSIONS This study demonstrates a unique syngeneic model of glioma resection and recurrence in immune-competent mice. Furthermore, this model provided insights into the pattern of postsurgical tumor recurrence and changes in the peritumoral microenvironment, as well as the impact of injured astrocytes on glioma growth and invasion. A better understanding of the postsurgical tumor microenvironment will allow development of targeted anticancer agents that improve surgery-mediated effects on tumor biology.
Collapse
Affiliation(s)
- Onyinyechukwu Okolie
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - Juli R Bago
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - Ralf S Schmid
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - David M Irvin
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - Ryan E Bash
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - C Ryan Miller
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| | - Shawn D Hingtgen
- Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (O.O., J.R.B., S.D.H.); Division of Neuropathology, Department of Pathology and Laboratory Medicine, Department of Neurology, and Neuroscience Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.R.M.); Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (R.S.S., D.M.I., R.E.B., C.R.M., S.D.H.); Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (S.D.H.)
| |
Collapse
|
13
|
Robel S, Sontheimer H. Glia as drivers of abnormal neuronal activity. Nat Neurosci 2016; 19:28-33. [PMID: 26713746 PMCID: PMC4966160 DOI: 10.1038/nn.4184] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/04/2015] [Indexed: 12/13/2022]
Abstract
Reactive astrocytes have been proposed to become incompetent bystanders in epilepsy as a result of cellular changes rendering them unable to perform important housekeeping functions. Indeed, successful surgical treatment of mesiotemporal lobe epilepsy hinges on the removal of the glial scar. New research now extends the role of astrocytes, suggesting that they may drive the disease process by impairing the inhibitory action of neuronal GABA receptors. Here we discuss studies that include hyperexcitability resulting from impaired supply of astrocytic glutamine for neuronal GABA synthesis, and epilepsy resulting from genetically induced astrogliosis or malignant transformation, both of which render the inhibitory neurotransmitter GABA excitatory. In these examples, glial cells alter the expression or function of neuronal proteins involved in excitability. Although epilepsy has traditionally been thought of as a disease caused by changes in neuronal properties exclusively, these new findings challenge us to consider the contribution of glial cells as drivers of epileptogenesis in acquired epilepsies.
Collapse
Affiliation(s)
- Stefanie Robel
- Virginia Tech Carilion Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, Virginia, USA
| | - Harald Sontheimer
- Virginia Tech Carilion Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, Virginia, USA
| |
Collapse
|
14
|
Mechanisms regulating glioma invasion. Cancer Lett 2015; 362:1-7. [PMID: 25796440 DOI: 10.1016/j.canlet.2015.03.015] [Citation(s) in RCA: 241] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/10/2015] [Accepted: 03/11/2015] [Indexed: 01/10/2023]
Abstract
Glioblastoma (GBM) is the most aggressive, deadliest, and most common brain malignancy in adults. Despite the advances made in surgical techniques, radiotherapy and chemotherapy, the median survival for GBM patients has remained at a mere 14 months. GBM poses several unique challenges to currently available treatments for the disease. For example, GBM cells have the propensity to aggressively infiltrate/invade into the normal brain tissues and along the vascular tracks, which prevents complete resection of all malignant cells and limits the effect of localized radiotherapy while sparing normal tissue. Although anti-angiogenic treatment exerts anti-edematic effect in GBM, unfortunately, tumors progress with acquired increased invasiveness. Therefore, it is an important task to gain a deeper understanding of the intrinsic and post-treatment invasive phenotypes of GBM in hopes that the gained knowledge would lead to novel GBM treatments that are more effective and less toxic. This review will give an overview of some of the signaling pathways that have been shown to positively and negatively regulate GBM invasion, including, the PI3K/Akt, Wnt, sonic hedgehog-GLI1, and microRNAs. The review will also discuss several approaches to cancer therapies potentially altering GBM invasiveness.
Collapse
|
15
|
Chen W, Wang D, Du X, He Y, Chen S, Shao Q, Ma C, Huang B, Chen A, Zhao P, Qu X, Li X. Glioma cells escaped from cytotoxicity of temozolomide and vincristine by communicating with human astrocytes. Med Oncol 2015; 32:43. [PMID: 25631631 DOI: 10.1007/s12032-015-0487-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/23/2015] [Indexed: 12/14/2022]
Abstract
Resistance to chemotherapeutic drugs remains a great obstacle to successful treatment of gliomas. Understanding the mechanism of glioma chemoresistance is conducive to develop effective strategies to overcome resistance. Astrocytes are the major stromal cells in the brain and have been demonstrated to play a key role in the malignant phenotype of gliomas. However, little is known regarding its role in glioma chemoresistance. In our study, we established a co-culture system of human astrocytes and glioma in vitro to simulate tumor microenvironment. Our results showed that astrocytes significantly reduced glioma cell apoptosis induced by the chemotherapeutic drugs temozolomide and vincristine. This protective effect was dependent on direct contact between astrocytes and glioma cells through Cx43-GJC. Moreover, in human glioma specimens, we found astrocytes infiltrating around the tumor, with a reactive appearance, suggesting that these astrocytes would play the same chemoprotective effect on gliomas in vivo. Our results expand the understanding of the interaction between astrocytes and glioma cells and provide a possible explanation for unsatisfactory clinical outcomes of chemotherapeutic drugs. Cx43-GJC between astrocytes and glioma cells may be a potential target for overcoming chemoresistance in gliomas clinically.
Collapse
Affiliation(s)
- Weiliang Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Oxygen Glucose Deprivation/Reperfusion Astrocytes Promotes Primary Neural Stem/Progenitor Cell Proliferation by Releasing High-Mobility Group Box 1. Neurochem Res 2014; 39:1440-50. [DOI: 10.1007/s11064-014-1333-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/06/2014] [Accepted: 05/10/2014] [Indexed: 01/09/2023]
|
17
|
Abstract
As the emergence of cancer is most frequent in proliferating tissues, replication errors are considered to be at the base of this disease. This review concentrates mainly on two neural cancers, neuroblastoma and glioma, with completely different backgrounds that are well documented with respect to their ontogeny. Although clinical data on other cancers of the nervous system are available, usually little can be said about their origins. Neuroblastoma is initiated in the embryo at a moment when the nervous system (NS) is in full expansion and occasionally genomic damage can lead to neoplasia. Glioma, to the contrary, occurs in the adult brain supposed to be mostly in a postmitotic state. According to current consensus, neural stem cells located in the subventricular zone (SVZ) in the adult are thought to accumulate enough genomic mutations to diverge on a carcinogenic course leading to diverse forms of glioma. After weighing the pros and cons of this current hypothesis in this review, it will be argued that this may be improbable, yielding to the original old concept of glial origin of glioma.
Collapse
|
18
|
Jha MK, Kim JH, Suk K. Proteome of brain glia: the molecular basis of diverse glial phenotypes. Proteomics 2013; 14:378-98. [PMID: 24124134 DOI: 10.1002/pmic.201300236] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 07/16/2013] [Accepted: 07/30/2013] [Indexed: 12/11/2022]
Abstract
Several different types of nonneuronal glial cells with diverse phenotypes are present in the CNS, and all have distinct indispensible functions. Although glial cells primarily provide neurons with metabolic and structural support in the healthy brain, they may switch phenotype from a "resting" to a "reactive" state in response to pathological insults. Furthermore, this reactive gliosis is an invariant feature of the pathogeneses of CNS maladies. The glial proteome serves as a signature of glial phenotype, and not only executes physiological functions, but also acts as a molecular mediator of the reactive glial phenotype. The glial proteome is also involved in intra- and intercellular communications as exemplified by glia-glia and neuron-glia interactions. The utilization of authoritative proteomic tools and the bioinformatic analyses have helped to profile the brain glial proteome and explore the molecular mechanisms of diverse glial phenotypes. Furthermore, technologic innovations have equipped the field of "glioproteomics" with refined tools for studies of the expression, interaction, and function of glial proteins in the healthy and in the diseased CNS. Glioproteomics is expected to contribute to the elucidation of the molecular mechanisms of CNS pathophysiology and to the discovery of biomarkers and theragnostic targets in CNS disorders.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science & Engineering Institute, Kyungpook National University School of Medicine, Daegu, South Korea
| | | | | |
Collapse
|