1
|
Poh QH, Rai A, Cross J, Greening DW. HB-EGF-loaded nanovesicles enhance trophectodermal spheroid attachment and invasion. Proteomics 2024; 24:e2200145. [PMID: 38214697 DOI: 10.1002/pmic.202200145] [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: 08/14/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024]
Abstract
The ability of trophectodermal cells (outer layer of the embryo) to attach to the endometrial cells and subsequently invade the underlying matrix are critical stages of embryo implantation during successful pregnancy establishment. Extracellular vesicles (EVs) have been implicated in embryo-maternal crosstalk, capable of reprogramming endometrial cells towards a pro-implantation signature and phenotype. However, challenges associated with EV yield and direct loading of biomolecules limit their therapeutic potential. We have previously established generation of cell-derived nanovesicles (NVs) from human trophectodermal cells (hTSCs) and their capacity to reprogram endometrial cells to enhance adhesion and blastocyst outgrowth. Here, we employed a rapid NV loading strategy to encapsulate potent implantation molecules such as HB-EGF (NVHBEGF). We show these loaded NVs elicit EGFR-mediated effects in recipient endometrial cells, activating kinase phosphorylation sites that modulate their activity (AKT S124/129, MAPK1 T185/Y187), and downstream signalling pathways and processes (AKT signal transduction, GTPase activity). Importantly, they enhanced target cell attachment and invasion. The phosphoproteomics and proteomics approach highlight NVHBEGF-mediated short-term signalling patterns and long-term reprogramming capabilities on endometrial cells which functionally enhance trophectodermal-endometrial interactions. This proof-of-concept study demonstrates feasibility in enhancing the functional potency of NVs in the context of embryo implantation.
Collapse
Affiliation(s)
- Qi Hui Poh
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Victoria, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Alin Rai
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Jonathon Cross
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Zhang YH, Sun XT, Guo RF, Feng GY, Gao HL, Zhong ML, Tian LW, Qiu ZY, Cui YW, Li JY, Zhao P. AβPP-tau-HAS1 axis trigger HAS1-related nuclear speckles and gene transcription in Alzheimer's disease. Matrix Biol 2024; 129:29-43. [PMID: 38518923 DOI: 10.1016/j.matbio.2024.03.003] [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: 09/25/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
As the backbone of the extracellular matrix (ECM) and the perineuronal nets (PNNs), hyaluronic acid (HA) provides binding sites for proteoglycans and other ECM components. Although the pivotal of HA has been recognized in Alzheimer's disease (AD), few studies have addressed the relationship between AD pathology and HA synthases (HASs). Here, HASs in different regions of AD brains were screened in transcriptomic database and validated in AβPP/PS1 mice. We found that HAS1 was distributed along the axon and nucleus. Its transcripts were reduced in AD patients and AβPP/PS1 mice. Phosphorylated tau (p-tau) mediates AβPP-induced cytosolic-nuclear translocation of HAS1, and negatively regulated the stability, monoubiquitination, and oligomerization of HAS1, thus reduced the synthesis and release of HA. Furthermore, non-ubiquitinated HAS1 mutant lost its enzyme activity, and translocated from the cytosol into the nucleus, forming nuclear speckles (NS). Unlike the splicing-related NS, less than 1 % of the non-ubiquitinated HAS1 co-localized with SRRM2, proving the regulatory role of HAS1 in gene transcription, indirectly. Thus, differentially expressed genes (DEGs) related to both non-ubiquitinated HAS1 mutant and AD were screened using transcriptomic datasets. Thirty-nine DEGs were identified, with 64.1 % (25/39) showing consistent results in both datasets. Together, we unearthed an important function of the AβPP-p-tau-HAS1 axis in microenvironment remodeling and gene transcription during AD progression, involving the ubiquitin-proteasome, lysosome, and NS systems.
Collapse
Affiliation(s)
- Ya-Hong Zhang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University
| | - Xing-Tong Sun
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University
| | - Rui-Fang Guo
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University
| | - Gang-Yi Feng
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University
| | - Hui-Ling Gao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University
| | - Man-Li Zhong
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University
| | - Li-Wen Tian
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University
| | - Zhong-Yi Qiu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University
| | - Yu-Wei Cui
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University
| | - Jia-Yi Li
- Health Sciences Institute, China Medical University; Neuronal Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University.
| | - Pu Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University; Lead contact.
| |
Collapse
|
3
|
Ortega JA, Soares de Aguiar GP, Chandravanshi P, Levy N, Engel E, Álvarez Z. Exploring the properties and potential of the neural extracellular matrix for next-generation regenerative therapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1962. [PMID: 38723788 DOI: 10.1002/wnan.1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024]
Abstract
The extracellular matrix (ECM) is a dynamic and complex network of proteins and molecules that surrounds cells and tissues in the nervous system and orchestrates a myriad of biological functions. This review carefully examines the diverse interactions between cells and the ECM, as well as the transformative chemical and physical changes that the ECM undergoes during neural development, aging, and disease. These transformations play a pivotal role in shaping tissue morphogenesis and neural activity, thereby influencing the functionality of the central nervous system (CNS). In our comprehensive review, we describe the diverse behaviors of the CNS ECM in different physiological and pathological scenarios and explore the unique properties that make ECM-based strategies attractive for CNS repair and regeneration. Addressing the challenges of scalability, variability, and integration with host tissues, we review how advanced natural, synthetic, and combinatorial matrix approaches enhance biocompatibility, mechanical properties, and functional recovery. Overall, this review highlights the potential of decellularized ECM as a powerful tool for CNS modeling and regenerative purposes and sets the stage for future research in this exciting field. This article is categorized under: Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- J Alberto Ortega
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Gisele P Soares de Aguiar
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Palash Chandravanshi
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Natacha Levy
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elisabeth Engel
- IMEM-BRT Group, Department of Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), Barcelona, Spain
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Zaida Álvarez
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
4
|
Ma J, Fang X, Chen M, Wang Y, Zhang L. Hyaluronan-CD44 Interaction Regulates Mouse Retinal Progenitor Cells Migration, Proliferation and Neuronal Differentiation. Stem Cell Rev Rep 2023; 19:2929-2942. [PMID: 37707669 PMCID: PMC10661819 DOI: 10.1007/s12015-023-10622-1] [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] [Accepted: 08/30/2023] [Indexed: 09/15/2023]
Abstract
Cell-based therapies have shown great potential because of their abilities to replace dying retinal neuron cells and preserve vision. The migration, proliferation and differentiation of retinal progenitor cells(RPCs) plays a vital role in the integration of the RPCs into the retina when transplanted into the host. Our study aimed to explore the effects of Hyaluronan(HA)-CD44 interactions on the regulation of RPCs migration, proliferation and differentiation, and investigate the underlying regulatory mechanisms. We found that CD44 was expressed in RPCs, and the HA-CD44 interaction markedly improved RPCs adhesion and migration. The stimulation of microRNA-21(miR-21) expression by the HA-CD44 interaction was protein kinase C (PKC)/Nanog-dependent in RPCs. Treatment of RPCs with PKC- or Nanog-specific ASODN or miR-21 antagomir effectively blocked HA-mediated RPCs adhesion and migration. Moreover, Rho-Kinase(ROK)/ Grb2-associated binders(Gab-1) associated phosphatidylinositol 3-kinase(PI3K)/AKT signalling activation was required for HA-CD44 interaction mediated RPCs proliferation and neuronal differentiation. Our findings demonstrated new roles for the HA-CD44 interaction in regulating the migration, proliferation and neuronal differentiation of RPCs. HA-CD44 signalling could represent a novel approach to controlling RPC fates, and the findings may be instructive for the application of RPCs for future therapeutic applications.
Collapse
Affiliation(s)
- Jian Ma
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Xiaoyun Fang
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Min Chen
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yao Wang
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Li Zhang
- Eye Center, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| |
Collapse
|
5
|
Berdiaki A, Neagu M, Spyridaki I, Kuskov A, Perez S, Nikitovic D. Hyaluronan and Reactive Oxygen Species Signaling—Novel Cues from the Matrix? Antioxidants (Basel) 2023; 12:antiox12040824. [PMID: 37107200 PMCID: PMC10135151 DOI: 10.3390/antiox12040824] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Hyaluronan (HA) is a naturally occurring non-sulfated glycosaminoglycan (GAG) localized to the cell surface and the tissue extracellular matrix (ECM). It is composed of disaccharides containing glucuronic acid and N-acetylglucosamine, is synthesized by the HA synthase (HAS) enzymes and is degraded by hyaluronidase (HYAL) or reactive oxygen and nitrogen species (ROS/RNS) actions. HA is deposited as a high molecular weight (HMW) polymer and degraded to low molecular weight (LMW) fragments and oligosaccharides. HA affects biological functions by interacting with HA-binding proteins (hyaladherins). HMW HA is anti-inflammatory, immunosuppressive, and antiangiogenic, whereas LMW HA has pro-inflammatory, pro-angiogenetic, and oncogenic effects. ROS/RNS naturally degrade HMW HA, albeit at enhanced levels during tissue injury and inflammatory processes. Thus, the degradation of endothelial glycocalyx HA by increased ROS challenges vascular integrity and can initiate several disease progressions. Conversely, HA exerts a vital role in wound healing through ROS-mediated HA modifications, which affect the innate immune system. The normal turnover of HA protects against matrix rigidification. Insufficient turnover leads to increased tissue rigidity, leading to tissue dysfunction. Both endogenous and exogenous HMW HA have a scavenging capacity against ROS. The interactions of ROS/RNS with HA are more complex than presently perceived and present an important research topic.
Collapse
|
6
|
Ritch SJ, Noman ASM, Goyeneche AA, Telleria CM. The metastatic capacity of high-grade serous ovarian cancer cells changes along disease progression: inhibition by mifepristone. Cancer Cell Int 2022; 22:397. [PMID: 36494669 PMCID: PMC9733158 DOI: 10.1186/s12935-022-02822-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Simplistic two-dimensional (2D) in vitro assays have long been the standard for studying the metastatic abilities of cancer cells. However, tri-dimensional (3D) organotypic models provide a more complex environment, closer to that seen in patients, and thereby provide a more accurate representation of their true capabilities. Our laboratory has previously shown that the antiprogestin and antiglucocorticoid mifepristone can reduce the growth, adhesion, migration, and invasion of various aggressive cancer cells assessed using 2D assays. In this study, we characterize the metastatic capabilities of high-grade serous ovarian cancer cells generated along disease progression, in both 2D and 3D assays, and the ability of cytostatic doses of mifepristone to inhibit them. METHODS High-grade serous ovarian cancer cells collected from two separate patients at different stages of their disease were used throughout the study. The 2D wound healing and Boyden chamber assays were used to study migration, while a layer of extracellular matrix was added to the Boyden chamber to study invasion. A 3D organotypic model, composed of fibroblasts embedded in collagen I and topped with a monolayer of mesothelial cells was used to further study cancer cell adhesion and mesothelial displacement. All assays were studied in cells, which were originally harvested from two patients at different stages of disease progression, in the absence or presence of cytostatic doses of mifepristone. RESULTS 2D in vitro assays demonstrated that the migration and invasive rates of the cells isolated from both patients decreased along disease progression. Conversely, in both patients, cells representing late-stage disease demonstrated a higher adhesion capacity to the 3D organotypic model than those representing an early-stage disease. This adhesive behavior is associated with the in vivo tumor capacity of the cells. Regardless of these differences in adhesive, migratory, and invasive behavior among the experimental protocols used, cytostatic doses of mifepristone were able to inhibit the adhesion, migration, and invasion rates of all cells studied, regardless of their basal capabilities over simplistic or organotypic metastatic in vitro model systems. Finally, we demonstrate that when cells acquire the capacity to grow spontaneously as spheroids, they do attach to a 3D organotypic model system when pre-incubated with conditioned media. Of relevance, mifepristone was able to cause dissociation of these multicellular structures. CONCLUSION Differences in cellular behaviours were observed between 2 and 3D assays when studying the metastatic capabilities of high-grade serous ovarian cancer cells representing disease progression. Mifepristone inhibited these metastatic capabilities in all assays studied.
Collapse
Affiliation(s)
- Sabrina J. Ritch
- grid.14709.3b0000 0004 1936 8649Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada
| | - Abu Shadat M. Noman
- grid.413089.70000 0000 9744 3393Department of Biochemistry and Molecular Biology, Chittagong University, Chittagong, Bangladesh
| | - Alicia A. Goyeneche
- grid.14709.3b0000 0004 1936 8649Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada ,grid.63984.300000 0000 9064 4811Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC Canada
| | - Carlos M. Telleria
- grid.14709.3b0000 0004 1936 8649Experimental Pathology Unit, Department of Pathology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC Canada ,grid.63984.300000 0000 9064 4811Cancer Research Program, Research Institute, McGill University Health Centre, Montreal, QC Canada
| |
Collapse
|
7
|
MiR-182 Inhibition Protects Against Experimental Stroke in vivo and Mitigates Astrocyte Injury and Inflammation in vitro via Modulation of Cortactin Activity. Neurochem Res 2022; 47:3682-3696. [PMID: 35951202 PMCID: PMC10069410 DOI: 10.1007/s11064-022-03718-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/08/2022] [Accepted: 07/27/2022] [Indexed: 10/15/2022]
Abstract
Ischemic stroke remains a devastating cerebrovascular disease that accounts for a high proportion of mortality and disability worldwide. MicroRNAs (miRNAs) are a class of small non-coding RNAs that are responsible for regulation of post-transcriptional gene expression, and growing evidence supports a role for miRNAs in stroke injury and recovery. The current study examined the role of miR-182 in experimental stroke using both in vitro and in vivo models of ischemic injury. Brain levels of miR-182 significantly increased after transient middle cerebral artery occlusion (MCAO) in mice and in primary astrocyte cultures subjected to combined oxygen-glucose deprivation/reperfusion (OGD/R) injury. In vivo, stroke volume and neurological score were significantly improved by pre-treatment with miR-182 antagomir. Astrocyte cultures stressed with OGD/R resulted in mitochondrial fragmentation and downregulation of cortactin, an actin-binding protein. Inhibition of miR-182 significantly preserved cortactin expression, reduced mitochondrial fragmentation and improved astrocyte survival after OGD/R. In parallel, lipopolysaccharide (LPS)-induced nitric-oxide release in astrocyte cultures was significantly reduced by miR-182 inhibition, translating to reduced injury in primary neuronal cultures subjected to conditioned medium from LPS-treated astrocytes. These findings identify miR-182 and/or cortactin as potential clinical targets to preserve mitochondrial structure and mitigate neuroinflammation and cell death after ischemic stroke.
Collapse
|
8
|
Pintér P, Alpár A. The Role of Extracellular Matrix in Human Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms231911085. [PMID: 36232390 PMCID: PMC9569603 DOI: 10.3390/ijms231911085] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
The dense neuropil of the central nervous system leaves only limited space for extracellular substances free. The advent of immunohistochemistry, soon followed by advanced diagnostic tools, enabled us to explore the biochemical heterogeneity and compartmentalization of the brain extracellular matrix in exploratory and clinical research alike. The composition of the extracellular matrix is critical to shape neuronal function; changes in its assembly trigger or reflect brain/spinal cord malfunction. In this study, we focus on extracellular matrix changes in neurodegenerative disorders. We summarize its phenotypic appearance and biochemical characteristics, as well as the major enzymes which regulate and remodel matrix establishment in disease. The specifically built basement membrane of the central nervous system, perineuronal nets and perisynaptic axonal coats can protect neurons from toxic agents, and biochemical analysis revealed how the individual glycosaminoglycan and proteoglycan components interact with these molecules. Depending on the site, type and progress of the disease, select matrix components can either proactively trigger the formation of disease-specific harmful products, or reactively accumulate, likely to reduce tissue breakdown and neuronal loss. We review the diagnostic use and the increasing importance of medical screening of extracellular matrix components, especially enzymes, which informs us about disease status and, better yet, allows us to forecast illness.
Collapse
Affiliation(s)
- Panka Pintér
- Department of Anatomy, Semmelweis University, 1113 Budapest, Hungary
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, 1113 Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, 1051 Budapest, Hungary
- Correspondence:
| |
Collapse
|
9
|
Weng X, Maxwell-Warburton S, Hasib A, Ma L, Kang L. The membrane receptor CD44: novel insights into metabolism. Trends Endocrinol Metab 2022; 33:318-332. [PMID: 35249813 DOI: 10.1016/j.tem.2022.02.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/17/2022]
Abstract
CD44, a cell-surface glycoprotein, has long been studied as a cancer molecule due to its essential role in physiological activities in normal cells and pathological activities in cancer cells, such as cell proliferation, adhesion, and migration; angiogenesis; inflammation; and cytoskeleton rearrangement. Yet, recent evidence suggests a role of CD44 in metabolism, especially insulin resistance in obesity and diabetes. In line with the current concept of fibroinflammation in obesity and insulin resistance, CD44 as the main receptor of the extracellular matrix component, hyaluronan (HA), has been shown to regulate diet-induced insulin resistance in muscle and other insulin-sensitive tissues. In this review, we integrate current evidence for a role of CD44 in regulating glucose and lipid homeostasis and speculate about its involvement in the pathogenesis of chronic metabolic diseases, including obesity and diabetes. We summarize the current development of CD44-targeted therapies and discuss its potential for the use in treating metabolic diseases.
Collapse
Affiliation(s)
- Xiong Weng
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | | | - Annie Hasib
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Lifeng Ma
- School of Medicine, Xizang Minzhu University, Xianyang, Shaanxi, China
| | - Li Kang
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK.
| |
Collapse
|
10
|
Chelyshev YA, Kabdesh IM, Mukhamedshina YO. Extracellular Matrix in Neural Plasticity and Regeneration. Cell Mol Neurobiol 2022; 42:647-664. [PMID: 33128689 PMCID: PMC11441266 DOI: 10.1007/s10571-020-00986-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix (ECM) is a fundamental component of biological tissues. The ECM in the central nervous system (CNS) is unique in both composition and function. Functions such as learning, memory, synaptogenesis, and plasticity are regulated by numerous ECM molecules. The neural ECM acts as a non-specific physical barrier that modulates neuronal plasticity and axon regeneration. There are two specialized types of ECM in the CNS, diffuse perisynaptic ECM and condensed ECM, which selectively surround the perikaryon and initial part of dendritic trees in subtypes of neurons, forming perineuronal nets. This review presents the current knowledge about the role of important neuronal ECM molecules in maintaining the basic functions of a neuron, including electrogenesis and the ability to form neural circuits. The review mainly focuses on the role of ECM components that participate in the control of key events such as cell survival, axonal growth, and synaptic remodeling. Particular attention is drawn to the numerous molecular partners of the main ECM components. These regulatory molecules are integrated into the cell membrane or disposed into the matrix itself in solid or soluble form. The interaction of the main matrix components with molecular partners seems essential in molecular mechanisms controlling neuronal functions. Special attention is paid to the chondroitin sulfate proteoglycan 4, type 1 transmembrane protein, neural-glial antigen 2 (NG2/CSPG4), whose cleaved extracellular domain is such a molecular partner that it not only acts directly on neural and vascular cells, but also exerts its influence indirectly by binding to resident ECM molecules.
Collapse
Affiliation(s)
- Yurii A Chelyshev
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Ilyas M Kabdesh
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kremlevskaya St 18, Kazan, Tatarstan, Russia, 420008
| | - Yana O Mukhamedshina
- Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia.
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kremlevskaya St 18, Kazan, Tatarstan, Russia, 420008.
| |
Collapse
|
11
|
Yu G, Zhang Y, Ning B. Reactive Astrocytes in Central Nervous System Injury: Subgroup and Potential Therapy. Front Cell Neurosci 2022; 15:792764. [PMID: 35002629 PMCID: PMC8733560 DOI: 10.3389/fncel.2021.792764] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Traumatic central nervous system (CNS) injury, which includes both traumatic brain injury (TBI) and spinal cord injury (SCI), is associated with irreversible loss of neurological function and high medical care costs. Currently, no effective treatment exists to improve the prognosis of patients. Astrocytes comprise the largest population of glial cells in the CNS and, with the advancements in the field of neurology, are increasingly recognized as having key functions in both the brain and the spinal cord. When stimulated by disease or injury, astrocytes become activated and undergo a series of changes, including alterations in gene expression, hypertrophy, the loss of inherent functions, and the acquisition of new ones. Studies have shown that astrocytes are highly heterogeneous with respect to their gene expression profiles, and this heterogeneity accounts for their observed context-dependent phenotypic diversity. In the inured CNS, activated astrocytes play a dual role both as regulators of neuroinflammation and in scar formation. Identifying the subpopulations of reactive astrocytes that exert beneficial or harmful effects will aid in deciphering the pathological mechanisms underlying CNS injuries and ultimately provide a theoretical basis for the development of effective strategies for the treatment of associated conditions. Following CNS injury, as the disease progresses, astrocyte phenotypes undergo continuous changes. Although current research methods do not allow a comprehensive and accurate classification of astrocyte subpopulations in complex pathological contexts, they can nonetheless aid in understanding the roles of astrocytes in disease. In this review, after a brief introduction to the pathology of CNS injury, we summarize current knowledge regarding astrocyte activation following CNS injury, including: (a) the regulatory factors involved in this process; (b) the functions of different astrocyte subgroups based on the existing classification of astrocytes; and (c) attempts at astrocyte-targeted therapy.
Collapse
Affiliation(s)
- GuiLian Yu
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Zhang
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bin Ning
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
12
|
Nelson DW, Gilbert RJ. Extracellular Matrix-Mimetic Hydrogels for Treating Neural Tissue Injury: A Focus on Fibrin, Hyaluronic Acid, and Elastin-Like Polypeptide Hydrogels. Adv Healthc Mater 2021; 10:e2101329. [PMID: 34494398 PMCID: PMC8599642 DOI: 10.1002/adhm.202101329] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/23/2021] [Indexed: 12/19/2022]
Abstract
Neurological and functional recovery is limited following central nervous system injury and severe injury to the peripheral nervous system. Extracellular matrix (ECM)-mimetic hydrogels are of particular interest as regenerative scaffolds for the injured nervous system as they provide 3D bioactive interfaces that modulate cellular response to the injury environment and provide naturally degradable scaffolding for effective tissue remodeling. In this review, three unique ECM-mimetic hydrogels used in models of neural injury are reviewed: fibrin hydrogels, which rely on a naturally occurring enzymatic gelation, hyaluronic acid hydrogels, which require chemical modification prior to chemical crosslinking, and elastin-like polypeptide (ELP) hydrogels, which exhibit a temperature-sensitive gelation. The hydrogels are reviewed by summarizing their unique biological properties, their use as drug depots, and their combination with other biomaterials, such as electrospun fibers and nanoparticles. This review is the first to focus on these three ECM-mimetic hydrogels for their use in neural tissue engineering. Additionally, this is the first review to summarize the use of ELP hydrogels for nervous system applications. ECM-mimetic hydrogels have shown great promise in preclinical models of neural injury and future advancements in their design and use can likely lead to viable treatments for patients with neural injury.
Collapse
Affiliation(s)
- Derek W Nelson
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
| |
Collapse
|
13
|
Chun H, Lim J, Park KD, Lee CJ. Inhibition of monoamine oxidase B prevents reactive astrogliosis and scar formation in stab wound injury model. Glia 2021; 70:354-367. [PMID: 34713936 DOI: 10.1002/glia.24110] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 11/07/2022]
Abstract
Reactive astrocytes manifest molecular, structural, and functional alterations under various pathological conditions. We have previously demonstrated that the reactive astrocytes of the stab wound injury model (STAB) display aberrant cellular gamma-aminobutyric acid (GABA) content and tonic GABA release, whereas the active astrocytes under enriched environment (EE) express high levels of proBDNF. However, the role of monoamine oxidase B (MAO-B) in reactive astrogliosis and hypertrophy still remains unknown. Here, we investigate the role of MAO-B, a GABA-producing enzyme, in reactive astrogliosis in STAB. We observed that the genetic removal of MAO-B significantly reduced the hypertrophy, scar formation, and GABA production of reactive astrocytes, whereas the MAO-B overexpression under glial fibrillary acidic protein (GFAP) promoter enhanced the levels of GFAP and GABA. Furthermore, we found that one of the by-products of the MAO-B action, H2 O2 , but not GABA, was sufficient and necessary for the hypertrophy of reactive astrocytes. Notably, we identified two potent pharmacological tools to attenuate scar-forming astrogliosis-the recently developed reversible MAO-B inhibitor, KDS2010, and an H2 O2 scavenger, crisdesalazine (AAD-2004). Our results implicate that inhibiting MAO-B activity has dual beneficial effects in preventing astrogliosis and scar-formation under brain injury, and that the MAO-B/H2 O2 pathway can be a useful therapeutic target with a high clinical potential.
Collapse
Affiliation(s)
- Heejung Chun
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Ki Duk Park
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, Republic of Korea.,Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea.,IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
14
|
The structure and function of protein kinase C-related kinases (PRKs). Biochem Soc Trans 2021; 49:217-235. [PMID: 33522581 PMCID: PMC7925014 DOI: 10.1042/bst20200466] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/29/2020] [Accepted: 01/07/2021] [Indexed: 11/17/2022]
Abstract
The protein kinase C-related kinase (PRK) family of serine/threonine kinases, PRK1, PRK2 and PRK3, are effectors for the Rho family small G proteins. An array of studies have linked these kinases to multiple signalling pathways and physiological roles, but while PRK1 is relatively well-characterized, the entire PRK family remains understudied. Here, we provide a holistic overview of the structure and function of PRKs and describe the molecular events that govern activation and autoregulation of catalytic activity, including phosphorylation, protein interactions and lipid binding. We begin with a structural description of the regulatory and catalytic domains, which facilitates the understanding of their regulation in molecular detail. We then examine their diverse physiological roles in cytoskeletal reorganization, cell adhesion, chromatin remodelling, androgen receptor signalling, cell cycle regulation, the immune response, glucose metabolism and development, highlighting isoform redundancy but also isoform specificity. Finally, we consider the involvement of PRKs in pathologies, including cancer, heart disease and bacterial infections. The abundance of PRK-driven pathologies suggests that these enzymes will be good therapeutic targets and we briefly report some of the progress to date.
Collapse
|
15
|
PKN2 is involved in aggregation and spheroid formation of fibroblasts in suspension culture by regulating cell motility and N-cadherin expression. Biochem Biophys Rep 2021; 25:100895. [PMID: 33437883 PMCID: PMC7787963 DOI: 10.1016/j.bbrep.2020.100895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 01/04/2023] Open
Abstract
The role of Protein Kinase N2 (PKN2, also known as PRK2/PKNγ) in cell aggregate/spheroid formation in suspension culture was investigated using immortalized fibroblasts established from PKN2flox/flox mouse embryos. PKN2flox/flox cells formed cell aggregates in flat bottom low attachment well plates, such as 2% agar and poly-2-hydroxyethymethacrylate coated plates, however, Cre;PKN2flox/flox cells in which PKN2 was depleted by the introduction of Cre-recombinase rarely formed aggregates. Time-lapse analysis revealed that the velocity of Cre;PKN2flox/flox cell motility was significantly lower than that of PKN2flox/flox in a low attachment flat-bottom plate, which likely resulted in a lower cell-cell contact frequency among Cre;PKN2flox/flox cells. Conversely, Cre;PKN2flox/flox cells could form initial cell aggregates in U-bottom low attachment well plates, however, the succeeding compaction process was delayed in Cre;PKN2flox/flox cells with decreased roundness, although PKN2flox/flox cells underwent compaction in a round shape spheroid within 24 h. Immunoblot analysis revealed that the preparation of the cell suspension from adherent conditions using trypsin/EDTA treatment significantly decreased the expression of N-cadherin in both PKN2flox/flox and Cre;PKN2flox/flox cells. The N-cadherin expression level recovered time-dependently; however, the recovery of N-cadherin was significantly delayed in Cre;PKN2flox/flox cells compared to PKN2flox/flox cells. Reverse transcription quantitative PCR revealed that N-cadherin mRNA in Cre;PKN2flox/flox cells was significantly lower than that of PKN2flox/flox cells. These results suggest that PKN2 controls the velocity of cell motility and the transcription of N-cadherin in fibroblasts, leading to cell aggregation and compaction for spheroid formation in suspension culture. PKN2 is involved in initial fibroblast aggregation by regulating cell motility. PKN2 is involved in compaction of fibroblasts in suspension. N-cadherin protein level seems to be a key element for compaction of fibroblasts. PKN2 controls transcription of N-cadherin mRNA in fibroblasts.
Collapse
|
16
|
Sun J, Wu J, Hua F, Chen Y, Zhan F, Xu G. Sleep Deprivation Induces Cognitive Impairment by Increasing Blood-Brain Barrier Permeability via CD44. Front Neurol 2020; 11:563916. [PMID: 33329306 PMCID: PMC7728917 DOI: 10.3389/fneur.2020.563916] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Sleep deprivation occurs frequently in older adults, which can result in delirium and cognitive impairment. CD44 is a key molecular in blood-brain barrier (BBB) regulation. However, whether CD44 participates in the role of sleep deprivation in cognitive impairment remains unclear. In this study, the effect of sleep deprivation on cognitive ability, tissue inflammation, BBB permeability, and astrocyte activity were evaluated in vivo. The differentially expressed genes (DEGs) were identified by RNA sequencing. A CD44 overexpression in the BBB model was performed in vitro to assess the effect and mechanisms of CD44. Sleep deprivation impaired the learning and memory ability and increased the levels of inflammatory cytokines, along with increased BBB permeability and activated astrocytes in hippocampus tissue. RNA sequencing of the hippocampus tissue revealed that 329 genes were upregulated in sleep deprivation-induced mice compared to control mice, and 147 genes were downregulated. GO and pathways showed that DEGs were mainly involved in BBB permeability and astrocyte activation, including nervous system development, neuron development, and brain development, and neuroactive ligand-receptor interaction. Moreover, the PCR analysis revealed that CD44 was dramatically increased in mice with sleep deprivation induction. The overexpression of CD44 in astrocytes promoted BBB permeability in vitro and induced the expression of the downstream gene NANOG. Our results indicate that sleep deprivation upregulated CD44 expression in hippocampus tissue, and increased BBB permeability, resulting in cognitive impairment.
Collapse
Affiliation(s)
- Jing Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jusheng Wu
- Department of Anesthesiology, Zhuji People's Hospital of Zhejiang Province, Shaoxing, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yong Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fenfang Zhan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Sánchez N, González-Ramírez MC, Contreras EG, Ubilla A, Li J, Valencia A, Wilson A, Green JBA, Tucker AS, Gaete M. Balance Between Tooth Size and Tooth Number Is Controlled by Hyaluronan. Front Physiol 2020; 11:996. [PMID: 32982773 PMCID: PMC7476214 DOI: 10.3389/fphys.2020.00996] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/22/2020] [Indexed: 12/20/2022] Open
Abstract
While the function of proteins and genes has been widely studied during vertebrate development, relatively little work has addressed the role of carbohydrates. Hyaluronan (HA), also known as hyaluronic acid, is an abundant carbohydrate in embryonic tissues and is the main structural component of the extracellular matrix of epithelial and mesenchymal cells. HA is able to absorb large quantities of water and can signal by binding to cell-surface receptors. During organ development and regeneration, HA has been shown to regulate cell proliferation, cell shape, and migration. Here, we have investigated the function of HA during molar tooth development in mice, in which, similar to humans, new molars sequentially bud off from a pre-existing molar. Using an ex vivo approach, we found that inhibiting HA synthesis in culture leads to a significant increase in proliferation and subsequent size of the developing molar, while the formation of sequential molars was inhibited. By cell shape analysis, we observed that inhibition of HA synthesis caused an elongation and reorientation of the major cell axes, indicating that disruption to cellular orientation and shape may underlie the observed phenotype. Lineage tracing demonstrated the retention of cells in the developing first molar (M1) at the expense of the generation of a second molar (M2). Our results highlight a novel role for HA in controlling proliferation, cell orientation, and migration in the developing tooth, impacting cellular decisions regarding tooth size and number.
Collapse
Affiliation(s)
- Natalia Sánchez
- Department of Anatomy, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | - Angélica Ubilla
- Department of Anatomy, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jingjing Li
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Anyeli Valencia
- Department of Anatomy, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrés Wilson
- Department of Anatomy, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jeremy B A Green
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Abigail S Tucker
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Marcia Gaete
- Department of Anatomy, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
18
|
Jensen G, Holloway JL, Stabenfeldt SE. Hyaluronic Acid Biomaterials for Central Nervous System Regenerative Medicine. Cells 2020; 9:E2113. [PMID: 32957463 PMCID: PMC7565873 DOI: 10.3390/cells9092113] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
Hyaluronic acid (HA) is a primary component of the brain extracellular matrix and functions through cellular receptors to regulate cell behavior within the central nervous system (CNS). These behaviors, such as migration, proliferation, differentiation, and inflammation contribute to maintenance and homeostasis of the CNS. However, such equilibrium is disrupted following injury or disease leading to significantly altered extracellular matrix milieu and cell functions. This imbalance thereby inhibits inherent homeostatic processes that support critical tissue health and functionality in the CNS. To mitigate the damage sustained by injury/disease, HA-based tissue engineering constructs have been investigated for CNS regenerative medicine applications. HA's effectiveness in tissue healing and regeneration is primarily attributed to its impact on cell signaling and the ease of customizing chemical and mechanical properties. This review focuses on recent findings to highlight the applications of HA-based materials in CNS regenerative medicine.
Collapse
Affiliation(s)
- Gregory Jensen
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ 85224, USA;
| | - Julianne L. Holloway
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ 85224, USA;
| | - Sarah E. Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
19
|
Diverse Roles for Hyaluronan and Hyaluronan Receptors in the Developing and Adult Nervous System. Int J Mol Sci 2020; 21:ijms21175988. [PMID: 32825309 PMCID: PMC7504301 DOI: 10.3390/ijms21175988] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023] Open
Abstract
Hyaluronic acid (HA) plays a vital role in the extracellular matrix of neural tissues. Originally thought to hydrate tissues and provide mechanical support, it is now clear that HA is also a complex signaling molecule that can regulate cell processes in the developing and adult nervous systems. Signaling properties are determined by molecular weight, bound proteins, and signal transduction through specific receptors. HA signaling regulates processes such as proliferation, differentiation, migration, and process extension in a variety of cell types including neural stem cells, neurons, astrocytes, microglia, and oligodendrocyte progenitors. The synthesis and catabolism of HA and the expression of HA receptors are altered in disease and influence neuroinflammation and disease pathogenesis. This review discusses the roles of HA, its synthesis and breakdown, as well as receptor expression in neurodevelopment, nervous system function and disease.
Collapse
|
20
|
López-Carvallo JA, Mazón-Suástegui JM, Hernández-Oñate MÁ, Tovar-Ramírez D, Abasolo-Pacheco F, Morelos-Castro RM, Arcos-Ortega GF. Transcriptome analysis of Catarina scallop (Argopecten ventricosus) juveniles treated with highly-diluted immunomodulatory compounds reveals activation of non-self-recognition system. PLoS One 2020; 15:e0233064. [PMID: 32407349 PMCID: PMC7224555 DOI: 10.1371/journal.pone.0233064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022] Open
Abstract
Marine bivalve hatchery productivity is continuously challenged by apparition and propagation of new diseases, mainly those related to vibriosis. Disinfectants and antibiotics are frequently overused to prevent pathogen presence, generating a potential negative impact on the environment. Recently, the use of highly diluted compounds with immunostimulant properties in marine organisms has been trailed successfully to activate the self-protection mechanisms of marine bivalves. Despite their potential as immunostimulants, little is known about their way of action. To understand their effect, a comparative transcriptomic analysis was performed with Argopecten ventricosus juveniles. The experimental design consisted of four treatments formulated from pathogenic Vibrio lysates at two dilutions: [(T1) Vibrio parahaemolyticus and Vibrio alginolyticus 1D; (T2) V. parahaemolyticus and V. alginolyticus 7C]; minerals [(T3) PhA+SiT 7C], scorpion venom [(T4) ViT 31C]; and one control (C1) hydro-alcoholic solution (ethanol 1%). The RNA sequencing (RNAseq) analysis showed a higher modulation of differentially expressed genes (DEG) in mantle tissue compared to gill tissue. The scallops that showed a higher number of DEG related to immune response in mantle tissue corresponded to T1 (V. parahaemolyticus and V. alginolyticus lysate) and T3 (Silicea terra® - Phosphoric acid®). The transcriptome analysis allowed understanding some interactions between A. ventricosus juveniles and highly-diluted treatments.
Collapse
Affiliation(s)
- Jesús Antonio López-Carvallo
- Laboratorio Experimental de Cultivo de Moluscos, Centro de Investigaciones Biológicas del Noroeste, La Paz, México
| | - José Manuel Mazón-Suástegui
- Laboratorio Experimental de Cultivo de Moluscos, Centro de Investigaciones Biológicas del Noroeste, La Paz, México
| | - Miguel Ángel Hernández-Oñate
- CONACyT, Centro de Investigación en Alimentación y Desarrollo A.C, Hermosillo, Sonora, México
- * E-mail: (GFAO); (MAHO)
| | - Dariel Tovar-Ramírez
- Laboratorio de Fisiología Comparada y Genómica Funcional, Centro de Investigaciones Biológicas del Noroeste, La Paz, México
| | - Fernando Abasolo-Pacheco
- Facultad de Ciencias Agrarias, Universidad Técnica Estatal de Quevedo, Quevedo, Los Ríos, Ecuador
| | - Rosa María Morelos-Castro
- Laboratorio de Imunogenómica Marina, Centro de Investigaciones Biológicas del Noroeste, La Paz, México
| | - Guadalupe Fabiola Arcos-Ortega
- Laboratorio de Imunogenómica Marina, Centro de Investigaciones Biológicas del Noroeste, La Paz, México
- * E-mail: (GFAO); (MAHO)
| |
Collapse
|
21
|
Patel H, Li J, Herrero A, Kroboth J, Byron A, Kriegsheim AV, Brunton V, Carragher N, Hurd T, Frame M. Novel roles of PRK1 and PRK2 in cilia and cancer biology. Sci Rep 2020; 10:3902. [PMID: 32127582 PMCID: PMC7054267 DOI: 10.1038/s41598-020-60604-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 02/10/2020] [Indexed: 12/24/2022] Open
Abstract
PRK1 and PRK2 are two closely related AGC-family serine/threonine protein kinases. Here we demonstrate novel roles for them at cilia and in cancer biology. In both instances serum withdrawal leads to increased activating PRK1 and PRK2 phosphorylation (pPRK1/pPRK2) and their depletion results in reduced spheroid growth. pPRK1/pPRK2 localise to the transition zone of cilia and their co-depletion results in reduced cilia size, impaired planer polarity and impaired cilia associated signalling. High PRK2 (but not PRK1) expression correlates with poor outcome in patients with basal-like/Triple Negative (TN) Breast Cancer (BC) where there is also higher expression relative to other BC tumour subtypes. In agreement, depletion of PRK1 and PRK2 in mouse TNBC cells, or CRISPR/Cas9 mediated deletion of PRK2 alone, significantly reduces cell proliferation and spheroid growth. Finally proteomic analysis to identify PRK2 binding partners in mouse TNBC cells revealed proteins that are important for both cilia and BC biology. Taken together these data demonstrate novel roles for PRK1 and PRK2 at cilia and in BC biology and in the case of PRK2 in particular, identifies it as a novel TNBC therapeutic target.
Collapse
Affiliation(s)
- Hitesh Patel
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom.
- University of Sussex, Sussex Drug Discovery Centre, School of Life Sciences, Brighton, BN1 9QJ, United Kingdom.
| | - Jun Li
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Ana Herrero
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Jakob Kroboth
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Adam Byron
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Alex Von Kriegsheim
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Valerie Brunton
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Neil Carragher
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| | - Toby Hurd
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom.
| | - Margaret Frame
- University of Edinburgh, Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Crewe Road South, Edinburgh, United Kingdom
| |
Collapse
|
22
|
Ding HY, Xie YN, Dong Q, Kimata K, Nishida Y, Ishiguro N, Zhuo LS. Roles of hyaluronan in cardiovascular and nervous system disorders. J Zhejiang Univ Sci B 2019; 20:428-436. [PMID: 31090268 DOI: 10.1631/jzus.b1900155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hyaluronan is a widely occurring extracellular matrix molecule, which is not only a supporting structural component, but also an active regulator of cellular functions. The chemophysical and biological properties of hyaluronan are greatly affected by its molecular size and several hyaluronan-binding proteins, making hyaluronan a fascinating molecule with great functional diversity. This review summarizes our current understanding of the roles of hyaluronan in cardiovascular and nervous system disorders, such as atherosclerosis, myocardial infarction, and stroke, with the aim to provide a foundation for future research and clinical trials.
Collapse
Affiliation(s)
- Hong-Yan Ding
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ya-Nan Xie
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Koji Kimata
- Multidisciplinary Pain Center, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Yoshihiro Nishida
- Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Naoki Ishiguro
- Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Li-Sheng Zhuo
- Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| |
Collapse
|
23
|
Cell-matrix tension contributes to hypoxia in astrocyte-seeded viscoelastic hydrogels composed of collagen and hyaluronan. Exp Cell Res 2019; 376:49-57. [PMID: 30658092 DOI: 10.1016/j.yexcr.2019.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/08/2019] [Accepted: 01/14/2019] [Indexed: 11/22/2022]
Abstract
Astrocyte activation is crucial for wound contraction and glial scar formation following central nervous system injury, but the mechanism by which activation leads to astrocyte contractility and matrix reorganization in the central nervous system (CNS) is unknown. Current means to measure cell traction forces within three-dimensional scaffolds are limited to analyzing individual or small groups of cells, within extracellular matrices, whereas gap junctions and other cell-cell adhesions connect astrocytes to form a functional syncytium within the glial scar. Here, we measure the viscoelastic properties of cell-seeded hydrogels to yield insight into the collective contractility of astrocytes as they exert tension on the surrounding matrix and change its bulk mechanical properties. Our results indicate that incorporation of the CNS matrix component hyaluronan into a collagen hydrogel increases expression of the intermediate filament protein GFAP and results in a higher shear storage modulus of the cell/matrix composite, establishing the correlation between astrocyte activation and increased cell contractility. The effects of thrombin and blebbistatin, known mediators of actomyosin-mediated contraction, verify that cell-matrix tension dictates the hydrogel mechanical properties. Viability assays indicate that increased cell traction exacerbates cell death at the center of the scaffold, and message level analysis reveals that cells in the hyaluronan-containing matrix have a ~ 3-fold increase in HIF-1α gene expression. Overall, these findings suggest that astrocyte activation not only increases cell traction, but may also contribute to hypoxia near sites of central nervous system injury.
Collapse
|
24
|
Powell MA, Black RT, Smith TL, Reeves TM, Phillips LL. Matrix Metalloproteinase 9 and Osteopontin Interact to Support Synaptogenesis in the Olfactory Bulb after Mild Traumatic Brain Injury. J Neurotrauma 2019; 36:1615-1631. [PMID: 30444175 DOI: 10.1089/neu.2018.5994] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Olfactory receptor axons reinnervate the olfactory bulb (OB) after chemical or transection lesion. Diffuse brain injury damages the same axons, but the time course and regulators of OB reinnervation are unknown. Gelatinases (matrix metalloproteinase [MMP]2, MMP9) and their substrate osteopontin (OPN) are candidate mediators of synaptogenesis after central nervous system (CNS) insult, including olfactory axon damage. Here, we examined the time course of MMP9, OPN, and OPN receptor CD44 response to diffuse OB injury. FVBV/NJ mice received mild midline fluid percussion insult (mFPI), after which MMP9 activity and both OPN and CD44 protein expression were measured. Diffuse mFPI induced time-dependent increase in OB MMP9 activity and elevated the cell signaling 48-kD OPN fragment. This response was bimodal at 1 and 7 days post-injury. MMP9 activity was also correlated with 7-day reduction in a second 32-kD OPN peptide. CD44 increase peaked at 3 days, delayed relative to MMP9/OPN response. MMP9 and OPN immunohistochemistry suggested that deafferented tufted and mitral neurons were the principal sites for these molecular interactions. Analysis of injured MMP9 knockout (KO) mice showed that 48-kD OPN production was dependent on OB MMP9 activity, but with no KO effect on CD44 induction. Olfactory marker protein (OMP), used to identify injured olfactory axons, revealed persistent axon damage in the absence of MMP9. MMP9 KO ultrastructure at 21 days post-injury indicated that persistent OMP reduction was paired with delayed removal of degenerated axons. These results provide evidence that diffuse, concussive brain trauma induces a post-injury interaction between MMP9, OPN, and CD44, which mediates synaptic plasticity and reinnervation within the OB.
Collapse
Affiliation(s)
- Melissa A Powell
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virgina
| | - Raiford T Black
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virgina
| | - Terry L Smith
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virgina
| | - Thomas M Reeves
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virgina
| | - Linda L Phillips
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virgina
| |
Collapse
|
25
|
Mitochondrial Neuroglobin Is Necessary for Protection Induced by Conditioned Medium from Human Adipose-Derived Mesenchymal Stem Cells in Astrocytic Cells Subjected to Scratch and Metabolic Injury. Mol Neurobiol 2018; 56:5167-5187. [PMID: 30536184 DOI: 10.1007/s12035-018-1442-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022]
Abstract
Astrocytes are specialized cells capable of regulating inflammatory responses in neurodegenerative diseases or traumatic brain injury. In addition to playing an important role in neuroinflammation, these cells regulate essential functions for the preservation of brain tissue. Therefore, the search for therapeutic alternatives to preserve these cells and maintain their functions contributes in some way to counteract the progress of the injury and maintain neuronal survival in various brain pathologies. Among these strategies, the conditioned medium from human adipose-derived mesenchymal stem cells (CM-hMSCA) has been reported with a potential beneficial effect against several neuropathologies. In this study, we evaluated the potential effect of CM-hMSCA in a model of human astrocytes (T98G cells) subjected to scratch injury. Our findings demonstrated that CM-hMSCA regulates the cytokines IL-2, IL-6, IL-8, IL-10, GM-CSF, and TNF-α, downregulates calcium at the cytoplasmic level, and regulates mitochondrial dynamics and the respiratory chain. These actions are accompanied by modulation of the expression of different proteins involved in signaling pathways such as AKT/pAKT and ERK1/2/pERK, and may mediate the localization of neuroglobin (Ngb) at the cellular level. We also confirmed that Ngb mediated the protective effects of CM-hMSCA through regulation of proteins involved in survival pathways and oxidative stress. In conclusion, regulation of brain inflammation combined with the recovery of fundamental cellular aspects in the face of injury makes CM-hMSCA a promising candidate for the protection of astrocytes in brain pathologies.
Collapse
|
26
|
Malinovskaya NA, Morgun AV, Pisareva NV, Osipova ED, Boytsova EB, Panina YA, Zhukov EL, Medvedeva NN, Salmina AB. Changes in the Permeability and Expression of Markers of the Structural and Functional Integrity of the Blood–Brain Barrier under Early Postnatal Hypoxia in vivo. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418030078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
27
|
Valkonen M, Haapasalo H, Rilla K, Tyynelä-Korhonen K, Soini Y, Pasonen-Seppänen S. Elevated expression of hyaluronan synthase 2 associates with decreased survival in diffusely infiltrating astrocytomas. BMC Cancer 2018; 18:664. [PMID: 29914429 PMCID: PMC6006557 DOI: 10.1186/s12885-018-4569-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/31/2018] [Indexed: 11/30/2022] Open
Abstract
Background Diffusely infiltrating astrocytomas originate from astrocytic glial cells or their precursor cells and are the most common type of brain tumors in adults. In this retrospective study, we investigated the content of hyaluronan, its cell surface receptor, CD44 and the expression of hyaluronan metabolizing enzymes, in these aggressive tumors. Hyaluronan is the main component of extracellular matrix in the brain. In many tumors, aberrant hyaluronan metabolism implicates aggressive disease progression and metastatic potential. Methods Our material consisted of 163 diffusely infiltrating astrocytomas (WHO grades II-IV). Tumor samples were processed into tissue microarray (TMA) blocks. The TMA sections were stained for hyaluronan, CD44, hyaluronan synthases 1–3 (HAS1–3) and hyaluronidase 2 (HYAL2). The immunostaining results were compared with χ2 –test or with Kruskal-Wallis test for correlation with clinicopathological parameters and survival analyses were done with Kaplan-Meier log rank test and Cox regression. Results Hyaluronan and CD44 were strongly expressed in astrocytic gliomas but their expression did not correlate with WHO grade or any other clinicopathological parameters whereas high HAS2 staining intensity was observed in IDH1 negative tumors (p = 0.003). In addition, in non-parametric tests increased HAS2 staining intensity correlated with increased cell proliferation (p = 0.013) and in log rank test with decreased overall survival of patients (p = 0.001). In the Cox regression analysis HAS2 expression turned out to be a significant independent prognostic factor (p = 0.008). Conclusions This study indicates that elevated expression of HAS2 is associated with glioma progression and suggests that HAS2 has a prognostic significance in diffusely infiltrating astrocytomas. Electronic supplementary material The online version of this article (10.1186/s12885-018-4569-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mari Valkonen
- Institute of Biomedicine, University of Eastern Finland, 70211, Kuopio, Finland
| | - Hannu Haapasalo
- Department of Pathology, University of Tampere and Fimlab Laboratories, Tampere, Finland
| | - Kirsi Rilla
- Institute of Biomedicine, University of Eastern Finland, 70211, Kuopio, Finland
| | | | - Ylermi Soini
- Institute of Clinical Medicine/ Clinical Pathology, University of Eastern Finland, Kuopio, Finland.,Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland.,Cancer Center of Eastern Finland, Kuopio, Finland
| | | |
Collapse
|
28
|
George N, Geller HM. Extracellular matrix and traumatic brain injury. J Neurosci Res 2018; 96:573-588. [PMID: 29344975 DOI: 10.1002/jnr.24151] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/21/2017] [Accepted: 08/14/2017] [Indexed: 12/27/2022]
Abstract
The brain extracellular matrix (ECM) plays a crucial role in both the developing and adult brain by providing structural support and mediating cell-cell interactions. In this review, we focus on the major constituents of the ECM and how they function in both normal and injured brain, and summarize the changes in the composition of the ECM as well as how these changes either promote or inhibit recovery of function following traumatic brain injury (TBI). Modulation of ECM composition to facilitates neuronal survival, regeneration and axonal outgrowth is a potential therapeutic target for TBI treatment.
Collapse
Affiliation(s)
- Naijil George
- Laboratory of Developmental Neurobiology, Cell Biology and Physiology Center, NHLBI, NIH, Bethesda, MD, 20892-1603, USA
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, Cell Biology and Physiology Center, NHLBI, NIH, Bethesda, MD, 20892-1603, USA
| |
Collapse
|
29
|
Nastase MV, Janicova A, Wygrecka M, Schaefer L. Signaling at the Crossroads: Matrix-Derived Proteoglycan and Reactive Oxygen Species Signaling. Antioxid Redox Signal 2017; 27:855-873. [PMID: 28510506 DOI: 10.1089/ars.2017.7165] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Proteoglycans (PGs), besides their structural contribution, have emerged as dynamic components that mediate a multitude of cellular events. The various roles of PGs are attributed to their structure, spatial localization, and ability to act as ligands and receptors. Reactive oxygen species (ROS) are small mediators that are generated in physiological and pathological conditions. Besides their reactivity and ability to induce oxidative stress, a growing body of data suggests that ROS signaling is more relevant than direct radical damage in development of human pathologies. Recent Advances: Cell surface transmembrane PGs (syndecans, cluster of differentiation 44) represent receptors in diverse and complex transduction networks, which involve redox signaling with implications in cancer, fibrosis, renal dysfunction, or Alzheimer's disease. Through NADPH oxidase (NOX)-dependent ROS, the extracellular PG, hyaluronan is involved in osteoclastogenesis and cancer. The ROS sources, NOX1 and NOX4, increase biglycan-induced inflammation, while NOX2 is a negative regulator. CRITICAL ISSUES The complexity of the mechanisms that bring ROS into the light of PG biology might be the foundation of a new research area with significant promise for understanding health and disease. Important aspects need to be investigated in PG/ROS signaling: the discovery of specific targets of ROS, the precise ROS-induced chemical modifications of these targets, and the study of their pathological relevance. FUTURE DIRECTIONS As we become more and more aware of the interactions between PG and ROS signaling underlying intracellular communication and cell fate decisions, it is quite conceivable that this field will allow to identify new therapeutic targets.-Antioxid. Redox Signal. 27, 855-873.
Collapse
Affiliation(s)
- Madalina-Viviana Nastase
- 1 Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität , Frankfurt am Main, Germany .,2 National Institute for Chemical-Pharmaceutical Research and Development , Bucharest, Romania
| | - Andrea Janicova
- 1 Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität , Frankfurt am Main, Germany
| | - Malgorzata Wygrecka
- 3 Department of Biochemistry, Faculty of Medicine, Justus Liebig University , Giessen, Germany
| | - Liliana Schaefer
- 1 Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe Universität , Frankfurt am Main, Germany
| |
Collapse
|
30
|
Baez-Jurado E, Hidalgo-Lanussa O, Guio-Vega G, Ashraf GM, Echeverria V, Aliev G, Barreto GE. Conditioned Medium of Human Adipose Mesenchymal Stem Cells Increases Wound Closure and Protects Human Astrocytes Following Scratch Assay In Vitro. Mol Neurobiol 2017; 55:5377-5392. [PMID: 28936798 DOI: 10.1007/s12035-017-0771-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/11/2017] [Indexed: 12/16/2022]
Abstract
Astrocytes perform essential functions in the preservation of neural tissue. For this reason, these cells can respond with changes in gene expression, hypertrophy, and proliferation upon a traumatic brain injury event (TBI). Different therapeutic strategies may be focused on preserving astrocyte functions and favor a non-generalized and non-sustained protective response over time post-injury. A recent strategy has been the use of the conditioned medium of human adipose mesenchymal stem cells (CM-hMSCA) as a therapeutic strategy for the treatment of various neuropathologies. However, although there is a lot of information about its effect on neuronal protection, studies on astrocytes are scarce and its specific action in glial cells is not well explored. In the present study, the effects of CM-hMSCA on human astrocytes subjected to scratch assay were assessed. Our findings indicated that CM-hMSCA improved cell viability, reduced nuclear fragmentation, and preserved mitochondrial membrane potential. These effects were accompanied by morphological changes and an increased polarity index thus reflecting the ability of astrocytes to migrate to the wound stimulated by CM-hMSCA. In conclusion, CM-hMSCA may be considered as a promising therapeutic strategy for the protection of astrocyte function in brain pathologies.
Collapse
Affiliation(s)
- Eliana Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
| | - Oscar Hidalgo-Lanussa
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
| | - Gina Guio-Vega
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Valentina Echeverria
- Research & Development Service, Bay Pines VA Healthcare System, Bay Pines, FL, 33744, USA.,Fac. Cs de la Salud, Universidad San Sebastián, Lientur 1457, 4080871, Concepción, Chile
| | - Gjumrakch Aliev
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow Region, 142432, Russia.,GALLY International Biomedical Research Consulting LLC, San Antonio, TX, 78229, USA.,School of Health Science and Healthcare Administration, University of Atlanta, Johns Creek, GA, 30097, USA
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, D.C., Colombia. .,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
31
|
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
|
32
|
Chiu CT, Kuo SN, Hung SW, Yang CY. Combined Treatment with Hyaluronic Acid and Mesalamine Protects Rats from Inflammatory Bowel Disease Induced by Intracolonic Administration of Trinitrobenzenesulfonic Acid. Molecules 2017; 22:molecules22060904. [PMID: 28556814 PMCID: PMC6152619 DOI: 10.3390/molecules22060904] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/26/2017] [Accepted: 05/28/2017] [Indexed: 12/31/2022] Open
Abstract
Drugs such as mesalamine (5-ASA) are currently recommended for the treatment of inflammatory bowel disease (IBD). To reduce the frequency of their administration and improve their therapeutic effect, this study investigated the adhesion efficacy, wound healing promotion, and decrease in inflammation in ulcers in the colonic tissue of rats with colitis after combined treatment with hyaluronic acid (HA) and 5-ASA (IBD98-M). HA-fluoresceinamine (FL) conjugates successfully adhered to the mucosal layer and were conjugated in the vascular tissue. In addition, macroscopic and microscopic observations indicated that colonic injuries reduced significantly after treatment with IBD98-M. Compared with PBS and 5-ASA treatment alone, treatment with IBD98-M more effectively reduced bowel inflammation and promoted colonic mucosal healing in TNBS-induced colitis. IBD98-M treatment also reduced myeloperoxidase activity and the expression levels of cyclooxygenase 2 and tumor necrosis factor-αin the colitis tissue. In conclusion, IBD98-M treatment strongly promoted wound healing in colonic injuries and significantly inhibited MPO activity in the inflamed colon tissue of rats. Combined treatment with HA and 5-ASA can accelerate wound healing and reduce inflammatory reaction in rat colitis.
Collapse
Affiliation(s)
| | - Sheng-Nan Kuo
- Holy Stone Healthcare Co., Ltd., Taipei 11493, Taiwan.
| | - Shao-Wen Hung
- Division of Animal Resource, Animal Technology Laboratories, Agricultural Technology Research Institute, Hsinchu City 30093, Taiwan.
| | - Cheng-Yao Yang
- Division of Animal Resource, Animal Technology Laboratories, Agricultural Technology Research Institute, Hsinchu City 30093, Taiwan.
| |
Collapse
|
33
|
Chao MW, Yang CH, Lin PT, Yang YH, Chuang YC, Chung MC, Tseng CY. Exposure to PM 2.5 causes genetic changes in fetal rat cerebral cortex and hippocampus. ENVIRONMENTAL TOXICOLOGY 2017; 32:1412-1425. [PMID: 27539004 DOI: 10.1002/tox.22335] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/29/2016] [Accepted: 07/30/2016] [Indexed: 05/06/2023]
Abstract
PM2.5 travels along the respiratory tract and enters systemic blood circulation. Studies have shown that PM2.5 increases the incidence of various diseases not only in adults but also in newborn infants. It causes chronic inflammation in pregnant women and retards fetal development. In this study, pregnant rats were exposed to PM2.5 for extended periods of time and it was found that PM2.5 exposure increased immune cells in mother rats. In addition, cytokines and free radicals rapidly accumulated in the amniotic fluid and indirectly affected the fetuses. The authors collected cerebral cortex and hippocampus samples at E18 and analyzed changes of miRNA levels. Expression levels of cortical miR-6315, miR-3588, and miR-466b-5p were upregulated, and positively correlated with the genes Pkn2 (astrocyte migration), Gorab (neuritogenesis), and Mobp (allergic encephalomyelitis). In contrast, PM2.5 decreased expression of miR-338-5p and let-7e-5p, both related to mental development. Further, PM2.5 exposure increased miR-3560 and let-7b-5p in the hippocampus, two proteins that regulate genes Oxct1 and Lin28b that control ketogenesis and glycosylation, and neural cell differentiation, respectively. miR-99b-5p, miR-92b-5p, and miR-99a-5p were decreased, leading to reduced expression of Kbtbd8 and Adam11 which reduced cell mitosis, migration, and differentiation, and inhibited learning abilities and motor coordination of the fetus. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1412-1425, 2017.
Collapse
Affiliation(s)
- Ming-Wei Chao
- Department of Bioscience Technology College of Science, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
- Center for Nanotechnology, College of Science, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Chin-Hua Yang
- Department of Diagnostic Radiology, Taoyuan General Hospital, Taoyaun, 310, Taiwan
- Departmewnt of Biomedical Engineering and Environmental Science, National Tsing Hua University, East District, Hsinchu 300, Taiwan
| | - Po-Ting Lin
- Department of Mechanical Engineering, College of Engineering, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Yu-Hsiu Yang
- Department of Biomedical Engineering College of Engineering, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Yu-Chen Chuang
- Department of Biomedical Engineering College of Engineering, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Meng-Chi Chung
- Department of Bioscience Technology College of Science, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| | - Chia-Yi Tseng
- Center for Nanotechnology, College of Science, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
- Department of Biomedical Engineering College of Engineering, Chung Yuan Christian University, Zhongli district, Taoyaun, 320, Taiwan
| |
Collapse
|
34
|
Baez-Jurado E, Vega GG, Aliev G, Tarasov VV, Esquinas P, Echeverria V, Barreto GE. Blockade of Neuroglobin Reduces Protection of Conditioned Medium from Human Mesenchymal Stem Cells in Human Astrocyte Model (T98G) Under a Scratch Assay. Mol Neurobiol 2017; 55:2285-2300. [PMID: 28332151 DOI: 10.1007/s12035-017-0481-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/03/2017] [Indexed: 12/25/2022]
Abstract
Previous studies have indicated that paracrine factors (conditioned medium) increase wound closure and reduce reactive oxygen species in a traumatic brain injury in vitro model. Although the beneficial effects of conditioned medium from human adipose tissue-derived mesenchymal stem cells (hMSCA-CM) have been previously suggested for various neurological diseases, their actions on astrocytic cells are not well understood. In this study, we have explored the effect of hMSCA-CM on human astrocyte model (T98G cells) subjected to scratch assay. Our results indicated that hMSCA-CM improved cell viability, reduced nuclear fragmentation, attenuated the production of reactive oxygen species, and preserved mitochondrial membrane potential and ultrastructural parameters. In addition, hMSCA-CM upregulated neuroglobin in T98G cells and the genetic silencing of this protein prevented the protective action of hMSCA-CM on damaged cells, suggesting that neuroglobin is mediating, at least in part, the protective effect of hMSCA-CM. Overall, this evidence suggests that the use of hMSCA-CM is a promising therapeutic strategy for the protection of astrocytic cells in central nervous system (CNS) pathologies.
Collapse
Affiliation(s)
- Eliana Baez-Jurado
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Gina Guio Vega
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - Gjumrakch Aliev
- Institute of Physiologically Active Compounds Russian Academy of Sciences, Chernogolovka, 142432, Russia
- GALLY International Biomedical Research Consulting LLC, San Antonio, TX, 78229, USA
- School of Health Science and Healthcare Administration, University of Atlanta, Johns Creek, GA, 30097, USA
| | - Vadim V Tarasov
- Institute of Pharmacy and Translational Medicine, Sechenov First Moscow State Medical University, 2-4 Bolshaya Pirogovskaya st., 119991, Moscow, Russia
| | - Paula Esquinas
- Facultad Medicina Veterinaria y Zootecnia, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Valentina Echeverria
- Facultad Ciencias de la Salud, Universidad San Sebastián, Lientur 1457, 4030000, Concepción, Chile
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia.
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
35
|
Danno S, Kubouchi K, Mehruba M, Abe M, Natsume R, Sakimura K, Eguchi S, Oka M, Hirashima M, Yasuda H, Mukai H. PKN2 is essential for mouse embryonic development and proliferation of mouse fibroblasts. Genes Cells 2017; 22:220-236. [PMID: 28102564 DOI: 10.1111/gtc.12470] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/21/2016] [Indexed: 12/17/2022]
Abstract
PKN2, a member of the protein kinase N (PKN) family, has been suggested by in vitro culture cell experiments to bind to Rho/Rac GTPases and contributes to cell-cell contact and cell migration. To unravel the in vivo physiological function of PKN2, we targeted the PKN2 gene. Constitutive disruption of the mouse PKN2 gene resulted in growth retardation and lethality before embryonic day (E) 10.5. PKN2-/- embryo did not undergo axial turning and showed insufficient closure of the neural tube. Mouse embryonic fibroblasts (MEFs) derived from PKN2-/- embryos at E9.5 failed to grow. Cre-mediated ablation of PKN2 in PKN2flox/flox MEFs obtained from E14.5 embryos showed impaired cell proliferation, and cell cycle analysis of these MEFs showed a decrease in S-phase population. Our results show that PKN2 is essential for mouse embryonic development and cell-autonomous proliferation of primary MEFs in culture. Comparison of the PKN2-/- phenotype with the phenotypes of PKN1 and PKN3 knockout strains suggests that PKN2 has distinct nonredundant functions in vivo, despite the structural similarity and evolutionary relationship among the three isoforms.
Collapse
Affiliation(s)
- Sally Danno
- Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Koji Kubouchi
- Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Mona Mehruba
- Graduate School of Medicine, Kobe University, Kobe, 650-0017, Japan
| | - Manabu Abe
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Rie Natsume
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Satoshi Eguchi
- Department of Medical Biology, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Masahiro Oka
- Division of Dermatology, Tohoku Medical and Pharmaceutical University, 1-12-1 Fukumuro, Miyagino-ku, Sendai, 983-8512, Japan
| | | | - Hiroki Yasuda
- Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Hideyuki Mukai
- Biosignal Research Center, Kobe University, Kobe, 657-8501, Japan
| |
Collapse
|
36
|
Zhan JS, Gao K, Chai RC, Jia XH, Luo DP, Ge G, Jiang YW, Fung YWW, Li L, Yu ACH. Astrocytes in Migration. Neurochem Res 2017; 42:272-282. [PMID: 27837318 DOI: 10.1007/s11064-016-2089-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/20/2016] [Accepted: 10/21/2016] [Indexed: 12/30/2022]
Abstract
Cell migration is a fundamental phenomenon that underlies tissue morphogenesis, wound healing, immune response, and cancer metastasis. Great progresses have been made in research methodologies, with cell migration identified as a highly orchestrated process. Brain is considered the most complex organ in the human body, containing many types of neural cells with astrocytes playing crucial roles in monitoring normal functions of the central nervous system. Astrocytes are mostly quiescent under normal physiological conditions in the adult brain but become migratory after injury. Under most known pathological conditions in the brain, spinal cord and retina, astrocytes are activated and become hypertrophic, hyperplastic, and up-regulating GFAP based on the grades of severity. These three observations are the hallmark in glia scar formation-astrogliosis. The reactivation process is initiated with structural changes involving cell process migration and ended with cell migration. Detailed mechanisms in astrocyte migration have not been studied extensively and remain largely unknown. Here, we therefore attempt to review the mechanisms in migration of astrocytes.
Collapse
Affiliation(s)
- 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, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, 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, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, 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, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, 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
| | - 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, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, 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
| | - Dao Peng Luo
- 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, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, 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, 550025, Guizhou, 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, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, 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, 550025, Guizhou, China
| | - Yu Wu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Yin-Wan Wendy Fung
- 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, Beijing, 100191, China
- Key Laboratory for Neuroscience, Ministry of Education, 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
| | - 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, Beijing, 100191, China.
- Key Laboratory for Neuroscience, Ministry of Education, 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.
| | - 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, Beijing, 100191, China.
- Key Laboratory for Neuroscience, Ministry of Education, National Health and Family Planning Commission, Peking University Health Science Center, Beijing, 100191, China.
- Laboratory of Translational Medicine, Institute of Systems Biomedicine, Peking University, Beijing, 100191, China.
- Hai Kang Life (Beijing) Corporation Ltd., Sino-I Campus No.1, Beijing Economic-Technological Development Area, Beijing, 100176, China.
| |
Collapse
|
37
|
PKN2 and Cdo interact to activate AKT and promote myoblast differentiation. Cell Death Dis 2016; 7:e2431. [PMID: 27763641 PMCID: PMC5133968 DOI: 10.1038/cddis.2016.296] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/10/2016] [Accepted: 08/22/2016] [Indexed: 01/01/2023]
Abstract
Skeletal myogenesis is coordinated by multiple signaling pathways that control cell adhesion/migration, survival and differentiation accompanied by muscle-specific gene expression. A cell surface protein Cdo is involved in cell contact-mediated promyogenic signals through activation of p38MAPK and AKT. Protein kinase C-related kinase 2 (PKN2/PRK2) is implicated in regulation of various biological processes, including cell migration, adhesion and death. It has been shown to interact with and inhibit AKT thereby inducing cell death. This led us to investigate the role of PKN2 in skeletal myogenesis and the crosstalk between PKN2 and Cdo. Like Cdo, PKN2 was upregulated in C2C12 myoblasts during differentiation and decreased in cells with Cdo depletion caused by shRNA or cultured on integrin-independent substratum. This decline of PKN2 levels resulted in diminished AKT activation during myoblast differentiation. Consistently, PKN2 overexpression-enhanced C2C12 myoblast differentiation, whereas PKN2-depletion impaired it, without affecting cell survival. PKN2 formed complexes with Cdo, APPL1 and AKT via its C-terminal region and this interaction appeared to be important for induction of AKT activity as well as myoblast differentiation. Furthermore, PKN2-enhanced MyoD-responsive reporter activities by mediating the recruitment of BAF60c and MyoD to the myogenin promoter. Taken together, PKN2 has a critical role in cell adhesion-mediated AKT activation during myoblast differentiation.
Collapse
|
38
|
Tarus D, Hamard L, Caraguel F, Wion D, Szarpak-Jankowska A, van der Sanden B, Auzély-Velty R. Design of Hyaluronic Acid Hydrogels to Promote Neurite Outgrowth in Three Dimensions. ACS APPLIED MATERIALS & INTERFACES 2016; 8:25051-25059. [PMID: 27598554 DOI: 10.1021/acsami.6b06446] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A hyaluronic acid (HA)-based extracellular matrix (ECM) platform with independently tunable stiffness and density of cell-adhesive peptide (RGD, arginine-glycine-aspartic acid) that mimics key biochemical and mechanical features of brain matrix has been designed. We demonstrated here its utility in elucidating ECM regulation of neural progenitor cell behavior and neurite outgrowth. The analysis of neurite outgrowth in 3-D by two-photon microscopy showed several important results in the development of these hydrogels. First, the ability of neurites to extend deeply into these soft HA-based matrices even in the absence of cell-adhesive ligand further confirms the potential of HA hydrogels for central nervous system (CNS) regeneration. Second, the behavior of hippocampal neural progenitor cells differed markedly between the hydrogels with a storage modulus of 400 Pa and those with a modulus of 800 Pa. We observed an increased outgrowth and density of neurites in the softest hydrogels (G' = 400 Pa). Interestingly, cells seeded on the surface of the hydrogels functionalized with the RGD ligand experienced an optimum in neurite outgrowth as a function of ligand density. Surprinsingly, neurites preferentially progressed inside the gels in a vertical direction, suggesting that outgrowth is directed by the hydrogel structure. This work may provide design principles for the development of hydrogels to facilitate neuronal regeneration in the adult brain.
Collapse
Affiliation(s)
- Dominte Tarus
- Grenoble Alpes University , Centre de Recherches sur les Macromolécules Végétales (CERMAV-CNRS), 601, rue de la Chimie, BP 53, Grenoble 38041 Cedex 9, France
| | - Lauriane Hamard
- Platform Intravital Microscopy, France Life Imaging, Grenoble Alpes University , INSERM U1205, 17 rue des Martyrs, 38054 Grenoble, France
| | - Flavien Caraguel
- Platform Intravital Microscopy, France Life Imaging, Grenoble Alpes University , INSERM U1205, 17 rue des Martyrs, 38054 Grenoble, France
| | - Didier Wion
- Grenoble Alpes University , INSERM U1205, 17 rue des Martyrs, 38054 Grenoble, France
| | - Anna Szarpak-Jankowska
- Grenoble Alpes University , Centre de Recherches sur les Macromolécules Végétales (CERMAV-CNRS), 601, rue de la Chimie, BP 53, Grenoble 38041 Cedex 9, France
| | - Boudewijn van der Sanden
- Platform Intravital Microscopy, France Life Imaging, Grenoble Alpes University , INSERM U1205, 17 rue des Martyrs, 38054 Grenoble, France
| | - Rachel Auzély-Velty
- Grenoble Alpes University , Centre de Recherches sur les Macromolécules Végétales (CERMAV-CNRS), 601, rue de la Chimie, BP 53, Grenoble 38041 Cedex 9, France
| |
Collapse
|
39
|
Konopka A, Zeug A, Skupien A, Kaza B, Mueller F, Chwedorowicz A, Ponimaskin E, Wilczynski GM, Dzwonek J. Cleavage of Hyaluronan and CD44 Adhesion Molecule Regulate Astrocyte Morphology via Rac1 Signalling. PLoS One 2016; 11:e0155053. [PMID: 27163367 PMCID: PMC4862642 DOI: 10.1371/journal.pone.0155053] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 04/22/2016] [Indexed: 11/19/2022] Open
Abstract
Communication of cells with their extracellular environment is crucial to fulfill their function in physiological and pathophysiological conditions. The literature data provide evidence that such a communication is also important in case of astrocytes. Mechanisms that contribute to the interaction between astrocytes and extracellular matrix (ECM) proteins are still poorly understood. Hyaluronan is the main component of ECM in the brain, where its major receptor protein CD44 is expressed by a subset of astrocytes. Considering the fact that functions of astrocytes are tightly coupled with changes in their morphology (e.g.: glutamate clearance in the synaptic cleft, migration, astrogliosis), we investigated the influence of hyaluronan cleavage by hyaluronidase, knockdown of CD44 by specific shRNA and CD44 overexpression on astrocyte morphology. Our results show that hyaluronidase treatment, as well as knockdown of CD44, in astrocytes result in a "stellate"-like morphology, whereas overexpression of CD44 causes an increase in cell body size and changes the shape of astrocytes into flattened cells. Moreover, as a dynamic reorganization of the actin cytoskeleton is supposed to be responsible for morphological changes of cells, and this reorganization is controlled by small GTPases of the Rho family, we hypothesized that GTPase Rac1 acts as a downstream effector for hyaluronan and CD44 in astrocytes. We used FRET-based biosensor and a dominant negative mutant of Rac1 to investigate the involvement of Rac1 activity in hyaluronidase- and CD44-dependent morphological changes of astrocytes. Both, hyaluronidase treatment and knockdown of CD44, enhances Rac1 activity while overexpression of CD44 reduces the activity state in astrocytes. Furthermore, morphological changes were blocked by specific inhibition of Rac1 activity. These findings indicate for the first time that regulation of Rac1 activity is responsible for hyaluronidase and CD44-driven morphological changes of astrocytes.
Collapse
Affiliation(s)
- Anna Konopka
- Laboratory of Molecular and Systemic Neuromorphology, The Nencki Institute of Experimental Biology, 02–093, Warsaw, ul. Pasteura 3, Poland
| | - Andre Zeug
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, 30625, Hannover, Germany
| | - Anna Skupien
- Laboratory of Molecular and Systemic Neuromorphology, The Nencki Institute of Experimental Biology, 02–093, Warsaw, ul. Pasteura 3, Poland
| | - Beata Kaza
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology, 02–093, Warsaw, ul. Pasteura 3, Poland
| | - Franziska Mueller
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, 30625, Hannover, Germany
| | - Agnieszka Chwedorowicz
- Laboratory of Molecular and Systemic Neuromorphology, The Nencki Institute of Experimental Biology, 02–093, Warsaw, ul. Pasteura 3, Poland
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, 30625, Hannover, Germany
| | - Grzegorz M. Wilczynski
- Laboratory of Molecular and Systemic Neuromorphology, The Nencki Institute of Experimental Biology, 02–093, Warsaw, ul. Pasteura 3, Poland
| | - Joanna Dzwonek
- Laboratory of Molecular and Systemic Neuromorphology, The Nencki Institute of Experimental Biology, 02–093, Warsaw, ul. Pasteura 3, Poland
| |
Collapse
|
40
|
Ventorp F, Barzilay R, Erhardt S, Samuelsson M, Träskman-Bendz L, Janelidze S, Weizman A, Offen D, Brundin L. The CD44 ligand hyaluronic acid is elevated in the cerebrospinal fluid of suicide attempters and is associated with increased blood-brain barrier permeability. J Affect Disord 2016; 193:349-54. [PMID: 26796235 DOI: 10.1016/j.jad.2015.12.069] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/07/2015] [Accepted: 12/26/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND The glycosaminoglycan hyaluronic acid (HA) is an important component of the extracellular matrix (ECM) in the brain. CD44 is a cell adhesion molecule that binds to HA in the ECM and is present on astrocytes, microglia and certain neurons. Cell adhesion molecules have been reported to be involved in anxiety and mood disorders. CD44 levels are decreased in the cerebrospinal fluid (CSF) of depressed individuals, and the CD44 gene has been identified in brain GWAS studies as a possible risk gene for suicidal behavior. METHOD We measured the CSF levels of HA and the soluble CD44 (sCD44) in suicide attempters (n=94) and in healthy controls (n=45) using ELISA and electrochemiluminescence assays. We also investigated other proteins known to interact with CD44, such as osteopontin and the matrix metalloproteinases MMP1, MMP3 and MMP9. RESULTS The suicide attempters had higher CSF levels of HA (p=.003) and MMP9 (p=.004). The CSF levels of HA correlated with BBB-permeability (rho=0.410, p<.001) and MMP9 correlated with sCD44 levels (rho=0.260, p=.005). LIMITATIONS Other relevant biological contributors to suicidal behavior is not addressed in parallel to the specific role of CD44-HA signaling. The gender distribution of the patients from whom CSF was analyzed was uneven. CONCLUSIONS Increased BBB-permeability and HA levels might be a results of increased neuroinflammation and can play a role in the pathobiology of suicidal behavior. The CD44 signaling pathway might be considered a novel target for intervention in mood disorders.
Collapse
Affiliation(s)
- F Ventorp
- Psychoimmunology Unit, Division of Psychiatry, Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Psychiatry and Behavioral Medicine, Michigan State University, Grand Rapids, Michigan, United States.
| | - R Barzilay
- Laboratory of Neuroscience, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Petach-Tikva, Israel; Research Unit at Geha Mental Health Center, Petach-Tikva, Israel
| | - S Erhardt
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - M Samuelsson
- Psychiatry Section, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - L Träskman-Bendz
- Psychoimmunology Unit, Division of Psychiatry, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - S Janelidze
- Psychoimmunology Unit, Division of Psychiatry, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - A Weizman
- Research Unit at Geha Mental Health Center, Petach-Tikva, Israel; Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Rabin Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - D Offen
- Laboratory of Neuroscience, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Petach-Tikva, Israel
| | - L Brundin
- Psychoimmunology Unit, Division of Psychiatry, Department of Clinical Sciences, Lund University, Lund, Sweden; Department of Psychiatry and Behavioral Medicine, Michigan State University, Grand Rapids, Michigan, United States; Laboratory of Behavioral Medicine, Van Andel Research Institute, Grand Rapids, Michigan, United States
| |
Collapse
|
41
|
Petrilli AM, Fernández-Valle C. Role of Merlin/NF2 inactivation in tumor biology. Oncogene 2016; 35:537-48. [PMID: 25893302 PMCID: PMC4615258 DOI: 10.1038/onc.2015.125] [Citation(s) in RCA: 300] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/20/2015] [Accepted: 03/16/2015] [Indexed: 01/13/2023]
Abstract
Merlin (Moesin-ezrin-radixin-like protein, also known as schwannomin) is a tumor suppressor protein encoded by the neurofibromatosis type 2 gene NF2. Loss of function mutations or deletions in NF2 cause neurofibromatosis type 2 (NF2), a multiple tumor forming disease of the nervous system. NF2 is characterized by the development of bilateral vestibular schwannomas. Patients with NF2 can also develop schwannomas on other cranial and peripheral nerves, as well as meningiomas and ependymomas. The only potential treatment is surgery/radiosurgery, which often results in loss of function of the involved nerve. There is an urgent need for chemotherapies that slow or eliminate tumors and prevent their formation in NF2 patients. Interestingly NF2 mutations and merlin inactivation also occur in spontaneous schwannomas and meningiomas, as well as other types of cancer including mesothelioma, glioma multiforme, breast, colorectal, skin, clear cell renal cell carcinoma, hepatic and prostate cancer. Except for malignant mesotheliomas, the role of NF2 mutation or inactivation has not received much attention in cancer, and NF2 might be relevant for prognosis and future chemotherapeutic approaches. This review discusses the influence of merlin loss of function in NF2-related tumors and common human cancers. We also discuss the NF2 gene status and merlin signaling pathways affected in the different tumor types and the molecular mechanisms that lead to tumorigenesis, progression and pharmacological resistance.
Collapse
Affiliation(s)
- Alejandra M. Petrilli
- Department of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Cristina Fernández-Valle
- Department of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| |
Collapse
|
42
|
A. Elawady M, M. Elmaghrabi M, Ebrahim N, A. Elawady M, Sabry D, Shamaa A, Ragaei A. Therapeutic Potential of Bone Marrow Derived Mesenchymal Stem Cells in Modulating Astroglyosis of Surgical Induced Experimental Spinal Cord Injury. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/abb.2016.76024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
43
|
He X, Liao W, Li Y, Wang Y, Chen Q, Jin J, He S. Upregulation of hyaluronan-mediated motility receptor in hepatocellular carcinoma predicts poor survival. Oncol Lett 2015; 10:3639-3646. [PMID: 26788183 DOI: 10.3892/ol.2015.3773] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 09/15/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of cancer across the world. Hyaluronic acid (HA) has been reported to serve an important role in tumor extension, progression, migration and invasion. In addition, the receptor for HA-mediated motility (RHAMM) has been demonstrated to be overexpressed in different types of cancer. However, whether the upregulation of RHAMM contributes to hepatocarcinogenesis of HCC remains unclear. The present study examined the RHAMM expression in 187 HCC patients by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC). RHAMM expression was significantly upregulated in liver cancer tissues compared with that observed in adjacent normal liver tissues. The IHC analysis demonstrated that RHAMM was overexpressed in 18 (72.0%) of the 25 HCC tissues. Furthermore, overexpression of RHAMM was associated with tumor-node-metastasis (TNM), the presence of vascular invasion and recurrence. Notably, the present study indicated that the overexpression of RHAMM was closely associated with the shorter disease-free and overall survival, so it may be a potential independent predictor for disease-free and overall survival of HCC patients. In conclusion, the upregulation of RHAMM is associated with HCC progression and prognosis; and it may be a potential independent predictor of disease-free and overall survival of HCC following surgical resection.
Collapse
Affiliation(s)
- Xiaohu He
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China; Laboratory of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin, Guangxi 541001, P.R. China
| | - Yulan Li
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin, Guangxi 541001, P.R. China
| | - Yongqin Wang
- Laboratory of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin, Guangxi 541001, P.R. China
| | - Qian Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China; Laboratory of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Junfei Jin
- Laboratory of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin, Guangxi 541001, P.R. China
| | - Songqing He
- Laboratory of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin, Guangxi 541001, P.R. China
| |
Collapse
|
44
|
Mishra JP, Cohen D, Zamperone A, Nesic D, Muesch A, Stein M. CagA of Helicobacter pylori interacts with and inhibits the serine-threonine kinase PRK2. Cell Microbiol 2015; 17:1670-82. [PMID: 26041307 DOI: 10.1111/cmi.12464] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/12/2015] [Accepted: 05/24/2015] [Indexed: 12/22/2022]
Abstract
CagA is a multifunctional toxin of Helicobacter pylori that is secreted into host epithelial cells by a type IV secretion system. Following host cell translocation, CagA interferes with various host-cell signalling pathways. Most notably this toxin is involved in the disruption of apical-basolateral cell polarity and cell adhesion, as well as in the induction of cell proliferation, migration and cell morphological changes. These are processes that also play an important role in epithelial-to-mesenchymal transition and cancer cell invasion. In fact, CagA is considered as the only known bacterial oncoprotein. The cellular effects are triggered by a variety of CagA activities including the inhibition of serine-threonine kinase Par1b/MARK2 and the activation of tyrosine phosphatase SHP-2. Additionally, CagA was described to affect the activity of Src family kinases and C-terminal Src kinase (Csk) suggesting that interference with multiple cellular kinase- and phosphatase-associated signalling pathways is a major function of CagA. Here, we describe the effect of CagA on protein kinase C-related kinase 2 (PRK2), which acts downstream of Rho GTPases and is known to affect cytoskeletal rearrangements and cell polarity. CagA interacts with PRK2 and inhibits its kinase activity. Because PRK2 has been linked to cytoskeletal rearrangements and establishment of cell polarity, we suggest that CagA may hijack PRK2 to further manipulate cancer-related signalling pathways.
Collapse
Affiliation(s)
- Jyoti Prasad Mishra
- Department of Health Sciences, School of Arts and Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | - David Cohen
- Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Dragana Nesic
- Laboratory of Structural Microbiology, The Rockefeller University, New York, NY, USA
| | - Anne Muesch
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Markus Stein
- Department of Health Sciences, School of Arts and Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| |
Collapse
|
45
|
Signaling molecules regulating phenotypic conversions of astrocytes and glial scar formation in damaged nerve tissues. Neurochem Int 2014; 78:35-42. [DOI: 10.1016/j.neuint.2014.08.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/17/2014] [Accepted: 08/22/2014] [Indexed: 12/21/2022]
|
46
|
Son SR, Sarkar SK, Linh NTB, Padalhin AR, Kim BR, Jung HI, Lee BT. Platelet-rich plasma encapsulation in hyaluronic acid/gelatin-BCP hydrogel for growth factor delivery in BCP sponge scaffold for bone regeneration. J Biomater Appl 2014; 29:988-1002. [DOI: 10.1177/0885328214551373] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Microporous calcium phosphate based synthetic bone substitutes are used for bone defect healing. Different growth factor loading has been investigated for enhanced bone regeneration. The platelet is a cellular component of blood which naturally contains a pool of necessary growth factors that mediate initiation, continuation, and completion of cellular mechanism of healing. In this work, we have investigated the encapsulation and immobilization of platelet-rich plasma (PRP) with natural polymers like hyaluronic acid (HA) and gelatin (Gel) and loading them in a biphasic calcium phosphate (BCP) scaffold, for a synthetic-allologous hybrid scaffold. Effect of PRP addition in small doses was evaluated for osteogenic potential in vitro and in vivo. BCP (10%) mixed HA–Gel hydrogel with or without PRP, was loaded into a BCP sponge scaffold. We investigated the hydrogel-induced improvement in mechanical property and PRP-mediated enhancement in biocompatibility. In vitro studies for cytotoxicity, cell attachment, and proliferation were carried out using MC3T3-E1 pre-osteoblast cells. In in vitro studies, the cell count, cell proliferation, and cell survival were higher in the scaffold with PRP loading than without PRP. However, in the in vivo studies using a rat model, the PRP scaffold was not superior to the scaffold without PRP. This discrepancy was investigated in terms of the interaction of PRP in the in vivo environment.
Collapse
Affiliation(s)
- So-Ra Son
- Department of Regenerative Medicine, Ssangyong-dong, Chungnam, Republic of Korea
| | - Swapan Kumar Sarkar
- Instititue of Tissue Regeneration, College of Medicine, Soonchunhyang University, Ssangyong-dong, Chungnam, Republic of Korea
| | - Nguyen-Thuy Ba Linh
- Instititue of Tissue Regeneration, College of Medicine, Soonchunhyang University, Ssangyong-dong, Chungnam, Republic of Korea
| | - Andrew R Padalhin
- Department of Regenerative Medicine, Ssangyong-dong, Chungnam, Republic of Korea
| | - Bo Ram Kim
- Department of Regenerative Medicine, Ssangyong-dong, Chungnam, Republic of Korea
| | - Hae Il Jung
- Department of Surgery, Soonchunhyang University, Cheonan Hospital, Cheonan, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, Ssangyong-dong, Chungnam, Republic of Korea
- Instititue of Tissue Regeneration, College of Medicine, Soonchunhyang University, Ssangyong-dong, Chungnam, Republic of Korea
| |
Collapse
|
47
|
Karbownik MS, Nowak JZ. Hyaluronan: towards novel anti-cancer therapeutics. Pharmacol Rep 2014; 65:1056-74. [PMID: 24399703 DOI: 10.1016/s1734-1140(13)71465-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 05/16/2013] [Indexed: 12/17/2022]
Abstract
The understanding of the role of hyaluronan in physiology and various pathological conditions has changed since the complex nature of its synthesis, degradation and interactions with diverse binding proteins was revealed. Initially perceived only as an inert component of connective tissue, it is now known to be involved in multiple signaling pathways, including those involved in cancer pathogenesis and progression. Hyaluronan presents a mixture of various length polymer molecules from finely fragmented oligosaccharides, polymers intermediate in size, to huge aggregates of high molecular weight hyaluronan. While large molecules promote tissue integrity and quiescence, the generation of breakdown products enhances signaling transduction, contributing to the pro-oncogenic behavior of cancer cells. Low molecular weight hyaluronan has well-established angiogenic properties, while the smallest hyaluronan oligomers may counteract tumor development. These equivocal properties make the role of hyaluronan in cancer biology very complex. This review surveys recent data on hyaluronan biosynthesis, metabolism, and interactions with its binding proteins called hyaladherins (CD44, RHAMM), providing themolecular background underlying its differentiated biological activity. In particular, the article critically presents current ideas on actual role of hyaluronan in cancer. The paper additionally maps a path towards promising novel anti-cancer therapeutics which target hyaluronan metabolic enzymes and hyaladherins, and constitute hyaluronan-based drug delivery systems.
Collapse
Affiliation(s)
- Michał S Karbownik
- Department of Pharmacology, Medical University of Lodz, Żeligowskiego 7/9, PL 90-752 Łódź, Poland. ;
| | | |
Collapse
|
48
|
Thauerer B, Zur Nedden S, Baier-Bitterlich G. Protein Kinase C-Related Kinase (PKN/PRK). Potential Key-Role for PKN1 in Protection of Hypoxic Neurons. Curr Neuropharmacol 2014; 12:213-8. [PMID: 24851086 PMCID: PMC4023452 DOI: 10.2174/1570159x11666131225000518] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 08/20/2013] [Accepted: 12/10/2013] [Indexed: 12/13/2022] Open
Abstract
Serine/threonine protein kinase C-related kinase (PKN/PRK) is a family of three isoenzymes (PKN1, PKN2,
PKN3), which are widely distributed in eukaryotic organisms and share the same overall domain structure. The Nterminal
region encompasses a conserved repeated domain, termed HR1a-c as well as a HR2/C2 domain. The
serine/threonine kinase domain is found in the C-terminal region of the protein and shows high sequence homology to
other members of the PKC superfamily.
In neurons, PKN1 is the most abundant isoform and has been implicated in a variety of functions including cytoskeletal
organization and neuronal differentiation and its deregulation may contribute to neuropathological processes such as
amyotrophic lateral sclerosis and Alzheimer’s disease. We have recently identified a candidate role of PKN1 in the
regulation of neuroprotective processes during hypoxic stress. Our key findings were that: 1) the activity of PKN1 was
significantly increased by hypoxia (1% O2) and neurotrophins (nerve growth factor and purine nucleosides); 2) Neuronal
cells, deficient of PKN1 showed a decrease of cell viability and neurite formation along with a disturbance of the F-actinassociated
cytoskeleton; 3) Purine nucleoside-mediated neuroprotection during hypoxia was severely hampered in PKN1
deficient neuronal cells, altogether suggesting a potentially critical role of PKN1 in neuroprotective processes.
This review gives an up-to-date overview of the PKN family with a special focus on the neuroprotective role of PKN1 in
hypoxia.
Collapse
Affiliation(s)
- Bettina Thauerer
- Medical University of Innsbruck, Biocenter/ Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Stephanie Zur Nedden
- Medical University of Innsbruck, Biocenter/ Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Gabriele Baier-Bitterlich
- Medical University of Innsbruck, Biocenter/ Neurobiochemistry, Innrain 80-82, A-6020 Innsbruck, Austria
| |
Collapse
|
49
|
Matrix hyaluronan-activated CD44 signaling promotes keratinocyte activities and improves abnormal epidermal functions. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1912-9. [PMID: 24819962 DOI: 10.1016/j.ajpath.2014.03.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 03/13/2014] [Accepted: 03/18/2014] [Indexed: 11/23/2022]
Abstract
Hyaluronan (HA), a major component of the extracellular matrix, is enriched in skin tissues, particularly the epidermis. HA binds to a ubiquitous, abundant, and functionally important family of cell surface receptors, CD44. This article reviews the current evidence for HA/CD44-mediated activation of RhoGTPase signaling and calcium mobilization, leading to the regulation of keratinocyte activities and various epidermal functions. It further discusses the role of HA-mediated CD44 interactions with unique downstream effectors, such as RhoGTPases (RhoA and Rac1), Rho-kinase, protein kinase-Nγ, and phosphoinositide-specific phospholipases (phospholipases Cε and Cγ1) in coordinating certain intracellular signaling pathways, such as calcium mobilization, phosphatidylinositol 3-kinase-AKT activation, cortactin-actin binding, and actin-associated cytoskeleton reorganization; generating the onset of important keratinocyte activities, such as cell adhesion, proliferation, migration, and differentiation; and performing epidermal functions. Topical application of selective HA fragments (large versus small HA) to the skin of wild-type mice (but not CD44 knockout mice) improves keratinocyte-associated epidermal functions and accelerates permeability barrier recovery and skin wound healing. Consequently, specific HA fragment (large versus small HA)-mediated signaling events (through the CD44 receptor) are required for keratinocyte activities, which offer new HA-based therapeutic options for patients experiencing epidermal dysfunction and skin damage as well as aging-related skin diseases, such as epidermal thinning (atrophy), permeability barrier dysfunction, and chronic nonhealing wounds.
Collapse
|
50
|
Torrente D, Avila MF, Cabezas R, Morales L, Gonzalez J, Samudio I, Barreto GE. Paracrine factors of human mesenchymal stem cells increase wound closure and reduce reactive oxygen species production in a traumatic brain injury in vitro model. Hum Exp Toxicol 2013; 33:673-84. [PMID: 24178889 DOI: 10.1177/0960327113509659] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Traumatic brain injury (TBI) consists of a primary and a secondary insult characterized by a biochemical cascade that plays a crucial role in cell death in the brain. Despite the major improvements in the acute care of head injury victims, no effective strategies exist for preventing the secondary injury cascade. This lack of success might be due to that most treatments are aimed at targeting neuronal population, even if studies show that astrocytes play a key role after a brain damage. In this work, we propose a new model of in vitro traumatic brain-like injury and use paracrine factors released by human mesenchymal stem cells (hMSCs) as a neuroprotective strategy. Our results demonstrate that hMSC-conditioned medium increased wound closure and proliferation at 12 h and reduced superoxide production to control conditions. This was accompanied by changes in cell morphology and polarity index, as both parameters reflect the ability of cells to migrate toward the wound. These findings indicate that hMSC is an important regulator of oxidative stress production, enhances cells migration, and shall be considered as a useful neuroprotective approach for brain recovery following injury.
Collapse
Affiliation(s)
- D Torrente
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| | - M F Avila
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| | - R Cabezas
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| | - L Morales
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| | - J Gonzalez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| | - I Samudio
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| | - G E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| |
Collapse
|