1
|
Neili NE, AbdelKafi-Koubaa Z, Jebali J, Kaidi K, Sahraoui G, Ahmed MB, Srairi-Abid N, Marrakchi N, Doghri R, ELBini I. Modulation of αv integrins by lebecetin, a viper venom-derived molecule, in experimental neuroinflammation and demyelination models. Sci Rep 2024; 14:22398. [PMID: 39333683 PMCID: PMC11436777 DOI: 10.1038/s41598-024-73259-1] [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] [Received: 05/06/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
Several neurodegenerative diseases, such as multiple sclerosis and Parkinson's disease, are linked to alterations in myelin content or structure. Transmembrane receptors such as integrins could be involved in these alterations. In the present study, we investigated the role of αv-integrins in experimental models of neuroinflammation and demyelination with the use of lebecetin (LCT), a C-lectin protein purified from Macrovipera lebetina viper venom, as an αv-integrin modulator. In a model of neuroinflammation, LCT inhibited the upregulation of αv, β3, β5, α5, and β1 integrins, as well as the associated release of pro-inflammatory factor IL-6 and chemokine CXCL-10, and decreased the expression of phosphorylated NfκB. The subsequent "indirect culture" between reactive astrocytes and oligodendrocytes showed a down-regulation of αv and β3 integrins versus upregulation of β1 one, accompanied by a reduced expression of myelin basic protein (MBP). Treatment of oligodendrocytes with LCT rectified the changes in integrin and MBP expression. Through Western blot quantification, LCT was shown to upregulate the expression levels of PI3K and p-mTOR while downregulating expression levels of p-AKT in oligodendrocytes, suggesting the neuroprotective and pro-myelinating effects of LCT may be related to the PI3K/mTor/AKT pathway. Concomitantly, we found that LCT promoted remyelination by tracking the increased expression of MBP in the brains of cuprizone-intoxicated mice. These results point to an involvement of integrins in not only neuroinflammation but demyelination as well. Thus, targeting αv integrins could offer potential therapeutic avenues for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Nour-Elhouda Neili
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Zaineb AbdelKafi-Koubaa
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Saleh Azaiez Institute, Tunis, Tunisia
| | - Jed Jebali
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Khouloud Kaidi
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Ghada Sahraoui
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Saleh Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Melika Ben Ahmed
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections (LR16IPT02), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Najet Srairi-Abid
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Naziha Marrakchi
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Raoudha Doghri
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Saleh Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Ines ELBini
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia.
| |
Collapse
|
2
|
Glotzbach K, Stamm N, Weberskirch R, Faissner A. Cationic Hydrogels Modulate Neural Stem and Progenitor Cell Proliferation and Differentiation Behavior in Dependence of Cationic Moiety Concentration in 2D Cell Culture. ACS Biomater Sci Eng 2024; 10:3148-3163. [PMID: 38227432 DOI: 10.1021/acsbiomaterials.3c01668] [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] [Indexed: 01/17/2024]
Abstract
The central nervous system (CNS) has a limited regenerative capacity because a hostile environment prevents tissue regeneration after damage or injury. Neural stem/progenitor cells (NSPCs) are considered a potential resource for CNS repair, which raises the issue of adequate cultivation and expansion procedures. Cationic charge supports the survival and adhesion of NSPCs. Typically, tissue culture plates with cationic coatings, such as poly-l-ornithine (PLO), have been used to culture these cell types (NSPCs). Yet presently, little is known about the impact of cationic charge concentration on the viability, proliferation, and differentiation capacity of NSPCs. Therefore, we have recently developed well-defined, fully synthetic hydrogel systems G1 (gel 1) to G6 (gel 6) that allow for the precise control of the concentration of the cationic trimethylaminoethyl acrylate (TMAEA) molecule associated with the polymer in a range from 0.06 to 0.91 μmol/mg. When murine NSPCs were cultured on these gels under differentiation conditions, we observed a strong correlation of cationic charge concentration with NSPC survival. In particular, neurons were preferentially formed on gels with a higher cationic charge concentration, whereas astrocytes and oligodendrocytes favored weakly charged or even neutral gel surfaces. To test the properties of the gels under proliferative conditions, the NSPCs were cultivated in the presence of fibroblast growth factor 2 (FGF2). The cytokine significantly increased the number of NSPCs but delayed the differentiation toward neurons and glia cells. Thus, the hydrogels are compatible with the survival, expansion, and differentiation of NSPCs and may be useful to create supportive environments in transplantation approaches.
Collapse
Affiliation(s)
- Kristin Glotzbach
- Department of Cell Morphology and Molecular Neurobiology, Ruhr Universität Bochum, Bochum 44801, Germany
| | - Nils Stamm
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund 44227, Germany
| | - Ralf Weberskirch
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund 44227, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr Universität Bochum, Bochum 44801, Germany
| |
Collapse
|
3
|
Staal L, Plösch T, Kunovac Kallak T, Sundström Poromaa I, Wertheim B, Olivier JDA. Sex-Specific Transcriptomic Changes in the Villous Tissue of Placentas of Pregnant Women Using a Selective Serotonin Reuptake Inhibitor. ACS Chem Neurosci 2024; 15:1074-1083. [PMID: 38421943 PMCID: PMC10958514 DOI: 10.1021/acschemneuro.3c00621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/09/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024] Open
Abstract
About 5% of pregnant women are treated with selective serotonin reuptake inhibitor (SSRI) antidepressants to treat their depression. SSRIs influence serotonin levels, a key factor in neural embryonic development, and their use during pregnancy has been associated with adverse effects on the developing embryo. However, the role of the placenta in transmitting these negative effects is not well understood. In this study, we aim to elucidate how disturbances in the maternal serotonergic system affect the villous tissue of the placenta by assessing whole transcriptomes in the placentas of women with healthy pregnancies and women with depression and treated with the SSRI fluoxetine during pregnancy. Twelve placentas of the Biology, Affect, Stress, Imaging and Cognition in Pregnancy and the Puerperium (BASIC) project were selected for RNA sequencing to examine differentially expressed genes: six male infants and six female infants, equally distributed over women treated with SSRI and without SSRI treatment. Our results show that more genes in the placenta of male infants show changed expression associated with fluoxetine treatment than in placentas of female infants, stressing the importance of sex-specific analyses. In addition, we identified genes related to extracellular matrix organization to be significantly enriched in placentas of male infants born to women treated with fluoxetine. It remains to be established whether the differentially expressed genes that we found to be associated with SSRI treatment are the result of the SSRI treatment itself, the underlying depression, or a combination of the two.
Collapse
Affiliation(s)
- Laura Staal
- Neurobiology,
Groningen Institute for Evolutionary Life Sciences, University of Groningen, 9700 CC Groningen, The Netherlands
- Department
of Cardiology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Torsten Plösch
- Departments
of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Perinatal
Neurobiology, Department of Human Medicine, School of Medicine and
Health Sciences, Carl von Ossietzky University
Oldenburg, 26129 Oldenburg, Germany
| | | | | | - Bregje Wertheim
- Evolutionary
Genetics, Development & Behaviour, Groningen Institute for Evolutionary
Life Sciences, University of Groningen, 9700 CC Groningen, The Netherlands
| | - Jocelien D. A. Olivier
- Neurobiology,
Groningen Institute for Evolutionary Life Sciences, University of Groningen, 9700 CC Groningen, The Netherlands
| |
Collapse
|
4
|
Wang W, Di Nisio E, Licursi V, Cacci E, Lupo G, Kokaia Z, Galanti S, Degan P, D’Angelo S, Castagnola P, Tavella S, Negri R. Simulated Microgravity Modulates Focal Adhesion Gene Expression in Human Neural Stem Progenitor Cells. Life (Basel) 2022; 12:life12111827. [PMID: 36362982 PMCID: PMC9699612 DOI: 10.3390/life12111827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
We analyzed the morphology and the transcriptomic changes of human neural stem progenitor cells (hNSPCs) grown on laminin in adherent culture conditions and subjected to simulated microgravity for different times in a random positioning machine apparatus. Low-cell-density cultures exposed to simulated microgravity for 24 h showed cell aggregate formation and significant modulation of several genes involved in focal adhesion, cytoskeleton regulation, and cell cycle control. These effects were much more limited in hNSPCs cultured at high density in the same conditions. We also found that some of the genes modulated upon exposure to simulated microgravity showed similar changes in hNSPCs grown without laminin in non-adherent culture conditions under normal gravity. These results suggest that reduced gravity counteracts the interactions of cells with the extracellular matrix, inducing morphological and transcriptional changes that can be observed in low-density cultures.
Collapse
Affiliation(s)
- Wei Wang
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Elena Di Nisio
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Valerio Licursi
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy c/o Department of Biology and Biotechnologies “C. Darwin”, Sapienza University, 00185 Rome, Italy
| | - Emanuele Cacci
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Giuseppe Lupo
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | - Zaal Kokaia
- Lund Stem Cell Center, Department of Clinical Sciences, Lund University, 22184 Lund, Sweden
| | - Sergio Galanti
- Excise, Custom and Monopolies Agency, ADM, 00153 Rome, Italy
| | - Paolo Degan
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Sara D’Angelo
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy
| | | | - Sara Tavella
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy
| | - Rodolfo Negri
- Department of Biology and Biotechnologies “C. Darwin”, Sapienza University of Rome, 00185 Rome, Italy
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy c/o Department of Biology and Biotechnologies “C. Darwin”, Sapienza University, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
5
|
Pham L, Wright DK, O'Brien WT, Bain J, Huang C, Sun M, Casillas-Espinosa PM, Shah AD, Schittenhelm RB, Sobey CG, Brady RD, O'Brien TJ, Mychasiuk R, Shultz SR, McDonald SJ. Behavioral, axonal, and proteomic alterations following repeated mild traumatic brain injury: Novel insights using a clinically relevant rat model. Neurobiol Dis 2020; 148:105151. [PMID: 33127468 DOI: 10.1016/j.nbd.2020.105151] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/07/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022] Open
Abstract
A history of mild traumatic brain injury (mTBI) is linked to a number of chronic neurological conditions, however there is still much unknown about the underlying mechanisms. To provide new insights, this study used a clinically relevant model of repeated mTBI in rats to characterize the acute and chronic neuropathological and neurobehavioral consequences of these injuries. Rats were given four sham-injuries or four mTBIs and allocated to 7-day or 3.5-months post-injury recovery groups. Behavioral analysis assessed sensorimotor function, locomotion, anxiety, and spatial memory. Neuropathological analysis included serum quantification of neurofilament light (NfL), mass spectrometry of the hippocampal proteome, and ex vivo magnetic resonance imaging (MRI). Repeated mTBI rats had evidence of acute cognitive deficits and prolonged sensorimotor impairments. Serum NfL was elevated at 7 days post injury, with levels correlating with sensorimotor deficits; however, no NfL differences were observed at 3.5 months. Several hippocampal proteins were altered by repeated mTBI, including those associated with energy metabolism, neuroinflammation, and impaired neurogenic capacity. Diffusion MRI analysis at 3.5 months found widespread reductions in white matter integrity. Taken together, these findings provide novel insights into the nature and progression of repeated mTBI neuropathology that may underlie lingering or chronic neurobehavioral deficits.
Collapse
Affiliation(s)
- Louise Pham
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - William T O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Jesse Bain
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Cheng Huang
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Pablo M Casillas-Espinosa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Anup D Shah
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; Monash Bioinformatics Platform, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Facility, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Neurology, The Alfred Hospital, Melbourne, VIC 3004, Australia; Department of Medicine, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne, VIC 3086, Australia; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
6
|
Patel BB, McNamara MC, Pesquera-Colom LS, Kozik EM, Okuzonu J, Hashemi NN, Sakaguchi DS. Recovery of Encapsulated Adult Neural Progenitor Cells from Microfluidic-Spun Hydrogel Fibers Enhances Proliferation and Neuronal Differentiation. ACS OMEGA 2020; 5:7910-7918. [PMID: 32309700 PMCID: PMC7160838 DOI: 10.1021/acsomega.9b04214] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/20/2020] [Indexed: 06/11/2023]
Abstract
Because of the limitations imposed by traditional two-dimensional (2D) cultures, biomaterials have become a major focus in neural and tissue engineering to study cell behavior in vitro. 2D systems fail to account for interactions between cells and the surrounding environment; these cell-matrix interactions are important to guide cell differentiation and influence cell behavior such as adhesion and migration. Biomaterials provide a unique approach to help mimic the native microenvironment in vivo. In this study, a novel microfluidic technique is used to encapsulate adult rat hippocampal stem/progenitor cells (AHPCs) within alginate-based fibrous hydrogels. To our knowledge, this is the first study to encapsulate AHPCs within a fibrous hydrogel. Alginate-based hydrogels were cultured for 4 days in vitro and recovered to investigate the effects of a 3D environment on the stem cell fate. Post recovery, cells were cultured for an additional 24 or 72 h in vitro before fixing cells to determine if proliferation and neuronal differentiation were impacted after encapsulation. The results indicate that the 3D environment created within a hydrogel is one factor promoting AHPC proliferation and neuronal differentiation (19.1 and 13.5%, respectively); however, this effect is acute. By 72 h post recovery, cells had similar levels of proliferation and neuronal differentiation (10.3 and 8.3%, respectively) compared to the control conditions. Fibrous hydrogels may better mimic the natural micro-environment present in vivo and be used to encapsulate AHPCs, enhancing cell proliferation and selective differentiation. Understanding cell behavior within 3D scaffolds may lead to the development of directed therapies for central nervous system repair and rescue.
Collapse
Affiliation(s)
- Bhavika B Patel
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa 50011, United States
- Neuroscience Program, Iowa State University, Ames, Iowa 50011, United States
| | - Marilyn C McNamara
- Department of Mechanical Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Laura S Pesquera-Colom
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa 50011, United States
- Biology Program, Iowa State University, Ames, Iowa 50010, United States
| | - Emily M Kozik
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa 50011, United States
- Biology Program, Iowa State University, Ames, Iowa 50010, United States
| | - Jasmin Okuzonu
- Department of Biological and Chemical Engineering, Iowa State University, Ames, Iowa 50010, United States
| | - Nicole N Hashemi
- Department of Mechanical Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Donald S Sakaguchi
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa 50011, United States
- Neuroscience Program, Iowa State University, Ames, Iowa 50011, United States
- Biology Program, Iowa State University, Ames, Iowa 50010, United States
| |
Collapse
|
7
|
Askvig JM, Dalzell TS, Toumeh N, Kuball PT, Whiteman ST, Bye EW, Andersen MJ, McCarthy MG, Irmen RE, Bexell SH, Benolken MM, Maruska BL, Nordmann SE. Age-dependent increase in Thy-1 protein in the rat supraoptic nucleus. Heliyon 2020; 6:e03501. [PMID: 32181386 PMCID: PMC7066247 DOI: 10.1016/j.heliyon.2020.e03501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/15/2019] [Accepted: 02/24/2020] [Indexed: 11/29/2022] Open
Abstract
Mature mammalian CNS neurons often do not recover successfully following injury. To this point, unilateral lesion of the hypothalamo-neurohypophysial tract results in collateral sprouting from uninjured axons of the supraoptic nucleus (SON) in 35-day-old but not in 125-day-old rats. Thus, it appears that there are age-related changes within the SON that preclude the older rat from recovering following axotomy. We hypothesize that the intrinsic capacity for axon reorganization may depend, in part, on age-related alterations in cell adhesion molecules that allow normal astrocyte-neuron interactions in the SON. In support of our hypothesis, numerous reports have shown that Thy-1 is increased in neurons at the cessation of axon outgrowth. Therefore, we compared protein levels of Thy-1 and the Thy-1 interacting integrin subunits, alpha-v (αv), beta-3 (ß3), and beta-5 (ß5), in 35- and 125-day-old SON using western blot analysis. Our results demonstrated that there was significantly more Thy-1 protein in the 125-day-old SON compared to 35-day-old SON, but no change in the protein levels of the integrin subunits. Furthermore, we localized Thy-1-, αv integrin-, ß3 integrin-, and ß5 integrin-immunoreactivity to both neurons and astrocytes in the SON. Altogether, our results suggest that the observed increase in Thy-1 protein levels in the SON with age may contribute to an environment that prevents collateral axonal sprouting in the SON of the 125-day-old rat.
Collapse
Affiliation(s)
- Jason M Askvig
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | - Talia S Dalzell
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | - Nadia Toumeh
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | - Phillip T Kuball
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | - Sara T Whiteman
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | - Erik W Bye
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | | | | | - Riley E Irmen
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | - Sydney H Bexell
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | - Molly M Benolken
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | - Brooke L Maruska
- Department of Biology, Concordia College, Moorhead, MN, 56562, USA
| | | |
Collapse
|
8
|
Quintela-López T, Ortiz-Sanz C, Serrano-Regal MP, Gaminde-Blasco A, Valero J, Baleriola J, Sánchez-Gómez MV, Matute C, Alberdi E. Aβ oligomers promote oligodendrocyte differentiation and maturation via integrin β1 and Fyn kinase signaling. Cell Death Dis 2019; 10:445. [PMID: 31171765 PMCID: PMC6554322 DOI: 10.1038/s41419-019-1636-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/17/2019] [Accepted: 05/08/2019] [Indexed: 12/13/2022]
Abstract
Alzheimer´s disease (AD) is characterized by a progressive cognitive decline that correlates with the levels of amyloid β-peptide (Aβ) oligomers. Strong evidences connect changes of oligodendrocyte function with the onset of neurodegeneration in AD. However, the mechanisms controlling oligodendrocyte responses to Aβ are still elusive. Here, we tested the role of Aβ in oligodendrocyte differentiation, maturation, and survival in isolated oligodendrocytes and in organotypic cerebellar slices. We found that Aβ peptides specifically induced local translation of 18.5-kDa myelin basic protein (MBP) isoform in distal cell processes concomitant with an increase of process complexity of MBP-expressing oligodendrocytes. Aβ oligomers required integrin β1 receptor, Src-family kinase Fyn and Ca2+/CaMKII as effectors to modulate MBP protein expression. The pharmacological inhibition of Fyn kinase also attenuated oligodendrocyte differentiation and survival induced by Aβ oligomers. Similarly, using ex vivo organotypic cerebellar slices Aβ promoted MBP upregulation through Fyn kinase, and modulated oligodendrocyte population dynamics by inducing cell proliferation and differentiation. Importantly, application of Aβ to cerebellar organotypic slices enhanced remyelination and oligodendrocyte lineage recovery in lysolecithin (LPC)-induced demyelination. These data reveal an important role of Aβ in oligodendrocyte lineage function and maturation, which may be relevant to AD pathogenesis.
Collapse
Affiliation(s)
- Tania Quintela-López
- Department of Neuroscience, University of Basque Country (UPV/EHU), Leioa, 48940, Spain.,Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain.,CIBERNED, Leioa, 48940, Spain
| | - Carolina Ortiz-Sanz
- Department of Neuroscience, University of Basque Country (UPV/EHU), Leioa, 48940, Spain.,Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain.,CIBERNED, Leioa, 48940, Spain
| | - Mari Paz Serrano-Regal
- Department of Neuroscience, University of Basque Country (UPV/EHU), Leioa, 48940, Spain.,Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain.,CIBERNED, Leioa, 48940, Spain
| | - Adhara Gaminde-Blasco
- Department of Neuroscience, University of Basque Country (UPV/EHU), Leioa, 48940, Spain.,Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain.,CIBERNED, Leioa, 48940, Spain
| | - Jorge Valero
- Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain.,IKERBASQUE Basque Foundation for Science, Bilbao, Spain
| | - Jimena Baleriola
- Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain.,IKERBASQUE Basque Foundation for Science, Bilbao, Spain.,Department of Cell Biology and Histology, UPV/EHU, Leioa, 48940, Spain
| | - Maria Victoria Sánchez-Gómez
- Department of Neuroscience, University of Basque Country (UPV/EHU), Leioa, 48940, Spain.,Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain.,CIBERNED, Leioa, 48940, Spain
| | - Carlos Matute
- Department of Neuroscience, University of Basque Country (UPV/EHU), Leioa, 48940, Spain. .,Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain. .,CIBERNED, Leioa, 48940, Spain.
| | - Elena Alberdi
- Department of Neuroscience, University of Basque Country (UPV/EHU), Leioa, 48940, Spain. .,Achucarro Basque Center for Neuroscience, Leioa, 48940, Spain. .,CIBERNED, Leioa, 48940, Spain.
| |
Collapse
|
9
|
Nieuwenhuis B, Haenzi B, Andrews MR, Verhaagen J, Fawcett JW. Integrins promote axonal regeneration after injury of the nervous system. Biol Rev Camb Philos Soc 2018; 93:1339-1362. [PMID: 29446228 PMCID: PMC6055631 DOI: 10.1111/brv.12398] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/23/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Integrins are cell surface receptors that form the link between extracellular matrix molecules of the cell environment and internal cell signalling and the cytoskeleton. They are involved in several processes, e.g. adhesion and migration during development and repair. This review focuses on the role of integrins in axonal regeneration. Integrins participate in spontaneous axonal regeneration in the peripheral nervous system through binding to various ligands that either inhibit or enhance their activation and signalling. Integrin biology is more complex in the central nervous system. Integrins receptors are transported into growing axons during development, but selective polarised transport of integrins limits the regenerative response in adult neurons. Manipulation of integrins and related molecules to control their activation state and localisation within axons is a promising route towards stimulating effective regeneration in the central nervous system.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
| | | | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
- Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam1081 HVAmsterdamThe Netherlands
| | - James W. Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Centre of Reconstructive NeuroscienceInstitute of Experimental Medicine142 20Prague 4Czech Republic
| |
Collapse
|
10
|
Abstract
The formation of correct synaptic structures and neuronal connections is paramount for normal brain development and a functioning adult brain. The integrin family of cell adhesion receptors and their ligands play essential roles in the control of several processes regulating neuronal connectivity - including neurite outgrowth, the formation and maintenance of synapses, and synaptic plasticity - that are affected in neurodevelopmental disorders, such as autism spectrum disorders (ASDs) and schizophrenia. Many ASD- and schizophrenia-associated genes are linked to alterations in the genetic code of integrins and associated signalling pathways. In non-neuronal cells, crosstalk between integrin-mediated adhesions and the actin cytoskeleton, and the regulation of integrin activity (affinity for extracellular ligands) are widely studied in healthy and pathological settings. In contrast, the roles of integrin-linked pathways in the central nervous system remains less well defined. In this Review, we will provide an overview of the known pathways that are regulated by integrin-ECM interaction in developing neurons and in adult brain. We will also describe recent advances in the identification of mechanisms that regulate integrin activity in neurons, and highlight the interesting emerging links between integrins and neurodevelopment.
Collapse
Affiliation(s)
- Johanna Lilja
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland .,Department of Biochemistry, University of Turku, FIN-20500 Turku, Finland
| |
Collapse
|
11
|
Andreopoulou E, Arampatzis A, Patsoni M, Kazanis I. Being a Neural Stem Cell: A Matter of Character But Defined by the Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:81-118. [PMID: 29204830 DOI: 10.1007/978-3-319-69194-7_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cells that build the nervous system, either this is a small network of ganglia or a complicated primate brain, are called neural stem and progenitor cells. Even though the very primitive and the very recent neural stem cells (NSCs) share common basic characteristics that are hard-wired within their character, such as the expression of transcription factors of the SoxB family, their capacity to give rise to extremely different neural tissues depends significantly on instructions from the microenvironment. In this chapter we explore the nature of the NSC microenvironment, looking through evolution, embryonic development, maturity and even disease. Experimental work undertaken over the last 20 years has revealed exciting insight into the NSC microcosmos. NSCs are very capable in producing their own extracellular matrix and in regulating their behaviour in an autocrine and paracrine manner. Nevertheless, accumulating evidence indicates an important role for the vasculature, especially within the NSC niches of the postnatal brain; while novel results reveal direct links between the metabolic state of the organism and the function of NSCs.
Collapse
Affiliation(s)
- Evangelia Andreopoulou
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Asterios Arampatzis
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK
- School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Melina Patsoni
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece
| | - Ilias Kazanis
- Lab of Developmental Biology, Department of Biology, University of Patras, Patras, Greece.
- Wellcome Trust- MRC Cambridge Stem Cell Biology Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
12
|
Schutte RJ, Xie Y, Ng NN, Figueroa P, Pham AT, O'Dowd DK. Astrocyte-enriched feeder layers from cryopreserved cells support differentiation of spontaneously active networks of human iPSC-derived neurons. J Neurosci Methods 2017; 294:91-101. [PMID: 28746822 DOI: 10.1016/j.jneumeth.2017.07.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Human induced pluripotent stem cell (hiPSC)-derived neuronal cultures are a useful tool for studying the mechanisms of neurological disorders and developing novel therapeutics. While plating hiPSC-derived neuronal progenitors onto glial feeder layers prepared from rodent cortex has been reported to promote functional differentiation of neuronal networks, this has not been examined in detail. NEW METHOD Here we describe a method of using cryopreserved cells from primary cultures for generation of mouse astrocyte-enriched, neuron-free feeder layers that grow from 10% to 100% confluence in 1 week. RESULTS Electrophysiological analysis demonstrated that compared to biochemical substrates alone, astrocyte-enriched feeder layers support more rapid differentiation of hiPSC-derived progenitors into excitable neurons that form spontaneously active networks in culture. There was a positive correlation between the degree of astroglial confluence at the time of progenitor plating and the average frequency of postsynaptic currents 3 weeks after plating. One disadvantage to plating on 100% confluent feeder layers was a high incidence of the astroglial layer with the overlying neurons detaching from the coverslips during transfer to the recording chamber. COMPARISON WITH EXISTING METHOD(S) Prevailing methods using primary glial feeder layers can result in possible contamination with rodent neurons and an unpredictable rate of growth. We provide a reliable method of generating mouse astroglial feeder layers from cryopreserved primary cultures to support differentiation of hiPSC-derived neurons. CONCLUSIONS The ability to make astrocyte-enriched feeder layers of defined confluence from cryopreserved primary cultures will facilitate the use of human stem cell derived neuronal cultures for disease modeling.
Collapse
Affiliation(s)
- Ryan J Schutte
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - Yunyao Xie
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - Nathan N Ng
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - Priscilla Figueroa
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - An T Pham
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States
| | - Diane K O'Dowd
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, United States.
| |
Collapse
|
13
|
Metalloproteases Affecting Blood Coagulation, Fibrinolysis and Platelet Aggregation from Snake Venoms: Definition and Nomenclature of Interaction Sites. Toxins (Basel) 2016; 8:toxins8100284. [PMID: 27690102 PMCID: PMC5086644 DOI: 10.3390/toxins8100284] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 09/21/2016] [Accepted: 09/22/2016] [Indexed: 01/08/2023] Open
Abstract
Snake venom metalloproteases, in addition to their contribution to the digestion of the prey, affect various physiological functions by cleaving specific proteins. They exhibit their activities through activation of zymogens of coagulation factors, and precursors of integrins or receptors. Based on their structure–function relationships and mechanism of action, we have defined classification and nomenclature of functional sites of proteases. These metalloproteases are useful as research tools and in diagnosis and treatment of various thrombotic and hemostatic conditions. They also contribute to our understanding of molecular details in the activation of specific factors involved in coagulation, platelet aggregation and matrix biology. This review provides a ready reference for metalloproteases that interfere in blood coagulation, fibrinolysis and platelet aggregation.
Collapse
|
14
|
Ye EA, Chawla SS, Khan MZ, Sakaguchi DS. Bone marrow-derived mesenchymal stem cells (MSCs) stimulate neurite outgrowth from differentiating adult hippocampal progenitor cells. ACTA ACUST UNITED AC 2016. [DOI: 10.7243/2054-717x-3-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
15
|
Poly-L-ornithine promotes preferred differentiation of neural stem/progenitor cells via ERK signalling pathway. Sci Rep 2015; 5:15535. [PMID: 26503112 PMCID: PMC4622086 DOI: 10.1038/srep15535] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/28/2015] [Indexed: 11/09/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) replacement therapies are the most attractive strategies to restore an injured brain. Key challenges of such therapies are enriching NSPCs and directing them differentiation into specific neural cell types. Here, three biomaterial substrates Poly-L-ornithine (PO), Poly-L-lysine (PLL) and fibronectin (FN) were investigated for their effects on proliferation and differentiation of rat NSPCs, and the underlying mechanisms were also explored. The results showed PO significantly increased NSPCs proliferation and induced preferred differentiation, compared with PLL and FN. Checking protein markers of several neural cell subtypes, it is showed PO significantly induced NSPCs expressing Doublecortin (DCX) and Olig2, one for neuroblasts and young neurons and the other for young oligodendrocytes. It is suggested the ERK signaling pathway was involving in this process because an ERK antagonist U0126 could inhibit PO’s effects mentioned above, as well as an ERK pathway agonist Ceramide C6 could enhance them. Given that both neurons and oligodendrocytes are the most vulnerable cells in many neurological diseases, PO-induced preferred differentiation into neurons and oligodendrocytes is a potential paradigm for NSPCs-based therapies.
Collapse
|
16
|
Oh J, Daniels GJ, Chiou LS, Ye EA, Jeong YS, Sakaguchi DS. Multipotent adult hippocampal progenitor cells maintained as neurospheres favor differentiation toward glial lineages. Biotechnol J 2014; 9:921-33. [PMID: 24844209 DOI: 10.1002/biot.201400019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/04/2014] [Accepted: 05/16/2014] [Indexed: 12/31/2022]
Abstract
Adult hippocampal progenitor cells (AHPCs) are generally maintained as a dispersed monolayer population of multipotent neural progenitors. To better understand cell-cell interactions among neural progenitors and their influences on cellular characteristics, we generated free-floating cellular aggregates, or neurospheres, from the adherent monolayer population of AHPCs. Results from in vitro analyses demonstrated that both populations of AHPCs were highly proliferative under maintenance conditions, but AHPCs formed in neurospheres favored differentiation along a glial lineage and displayed greater migrational activity than the traditionally cultured AHPCs. To study the plasticity of AHPCs from both populations in vivo, we transplanted green fluorescent protein (GFP)-expressing AHPCs via intraocular injection into the developing rat eyes. Both AHPC populations were capable of surviving and integrating into developing host central nervous system, but considerably more GFP-positive cells were observed in the retinas transplanted with neurosphere AHPCs, compared to adherent AHPCs. These results suggest that the culture configuration during maintenance for neural progenitor cells (NPCs) influences cell fate and motility in vitro as well as in vivo. Our findings have implication for understanding different cellular characteristics of NPCs according to distinct intercellular architectures and for developing cell-based therapeutic strategies using lineage-committed NPCs.
Collapse
Affiliation(s)
- Jisun Oh
- Neuroscience Program, Iowa State University, Ames, IA, USA; Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA; Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | | | | | | | | | | |
Collapse
|
17
|
Li YC, Tsai LK, Wang JH, Young TH. A neural stem/precursor cell monolayer for neural tissue engineering. Biomaterials 2014; 35:1192-204. [DOI: 10.1016/j.biomaterials.2013.10.066] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 10/20/2013] [Indexed: 11/30/2022]
|
18
|
Jung MS, Jang HB, Lee SE, Park JH, Hwang YS. In vitro micro-mineralized tissue formation by the combinatory condition of adipose-derived stem cells, macroporous PLGA microspheres and a bioreactor. Macromol Res 2013. [DOI: 10.1007/s13233-014-2002-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
19
|
Applications of snake venom components to modulate integrin activities in cell-matrix interactions. Int J Biochem Cell Biol 2013; 45:1974-86. [PMID: 23811033 DOI: 10.1016/j.biocel.2013.06.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/29/2013] [Accepted: 06/12/2013] [Indexed: 01/23/2023]
Abstract
Snake venom proteins are broadly investigated in the different areas of life science. Direct interaction of these compounds with cells may involve a variety of mechanisms that result in diverse cellular responses leading to the activation or blocking of physiological functions of the cell. In this review, the snake venom components interacting with integrins will be characterized in context of their effect on cellular response. Currently, two major families of snake venom proteins are considered as integrin-binding molecules. The most attention has been devoted to the disintegrin family, which binds certain types of integrins through specific motifs recognized as a tri-peptide structurally localized on an integrin-binding loop. Other snake venom integrin-binding proteins belong to the C-type lectin family. Snake venom molecules bind to the cellular integrins resulting in a modulation of cell signaling and in consequence, the regulation of cell proliferation, migration and apoptosis. Therefore, snake venom research on the integrin-binding molecules may have significance in biomedicine and basic cell biology.
Collapse
|
20
|
Combination of media, biomaterials and extracellular matrix proteins to enhance the differentiation of neural stem/precursor cells into neurons. Acta Biomater 2012; 8:3035-48. [PMID: 22548842 DOI: 10.1016/j.actbio.2012.04.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 04/13/2012] [Accepted: 04/23/2012] [Indexed: 11/21/2022]
Abstract
The purpose of this study was to induce the differentiation of neural stem/precursor cells (NSPC) more towards neurons than glial cells by the combination of media, biomaterials and extracellular matrix (ECM) proteins. Considering the role of serum, 10% fetal bovine serum or its fractions were added to DMEM/F12 medium to examine the effect of the differentiation-promoting potential on cultured NSPC isolated from embryonic rat cerebral cortex. The NSPC were cultured for 7 days, after which differentiation was assayed using immunocytochemistry for lineage specific markers. It was demonstrated that molecules promoting neuron differentiation were present in serum with molecular weight <100 kDa, which could dominate the differentiation of NSPC principally into neurons in the presence of basic fibroblast growth factor. In contrast, NSPC were induced to differentiate predominantly into glial cell phenotypes in the presence of whole serum components. Based on medium containing serum fraction, semi-quantification showed that the MAP2-positive percentage of the immunoreactive ratio within migrated cells could be promoted over 85% by combining poly(ethylene-co-vinyl alcohol) biomaterial and fibronectin matrix protein. These results are very encouraging, since an environment favorable for neuronal differentiation should be useful in the development of strategies for controlling the behavior of NSPC in neuroscience research.
Collapse
|
21
|
Howk CL, Levine HA, Smiley MW, Mallapragada SK, Nilsen-Hamilton M, Oh J, Sakaguchi DS. A mathematical model for selective differentiation of neural progenitor cells on micropatterned polymer substrates. Math Biosci 2012; 238:65-79. [PMID: 22569338 DOI: 10.1016/j.mbs.2012.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 02/20/2012] [Accepted: 04/02/2012] [Indexed: 01/25/2023]
Abstract
The biological hypothesis that the astrocyte-secreted cytokine, interleukin-6 (IL6), stimulates differentiation of adult rat hippocampal progenitor cells (AHPCs) is considered from a mathematical perspective. The proposed mathematical model includes two different mechanisms for stimulation and is based on mass-action kinetics. Both biological mechanisms involve sequential binding, with one pathway solely utilizing surface receptors while the other pathway also involves soluble receptors. Choosing biologically-reasonable values for parameters, simulations of the mathematical model show good agreement with experimental results. A global sensitivity analysis is also conducted to determine both the most influential and non-influential parameters on cellular differentiation, providing additional insights into the biological mechanisms.
Collapse
Affiliation(s)
- Cory L Howk
- Department of Mathematics, Iowa State University, Ames, IA 50011, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Eva R, Andrews MR, Franssen EHP, Fawcett JW. Intrinsic mechanisms regulating axon regeneration: an integrin perspective. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012; 106:75-104. [PMID: 23211460 DOI: 10.1016/b978-0-12-407178-0.00004-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adult central nervous system (CNS) axons fail to regenerate after injury because of inhibitory factors in the surrounding environment and a low intrinsic regenerative capacity. Axons in the adult peripheral nervous system have a higher regenerative capacity, due in part to the presence of certain integrins-receptors for the extracellular matrix. Integrins are critical for axon growth during the development of the nervous system but are absent from some adult CNS axons. Here, we discuss the intrinsic mechanisms that regulate axon regeneration and examine the role of integrins. As correct localization is paramount to integrin function, we further discuss the mechanisms that regulate integrin traffic toward the axonal growth cone.
Collapse
Affiliation(s)
- Richard Eva
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
23
|
Spitzer N, Sammons GS, Butts HM, Grover LM, Price EM. Multipotent progenitor cells derived from adult peripheral blood of swine have high neurogenic potential in vitro. J Cell Physiol 2011; 226:3156-68. [PMID: 21321934 PMCID: PMC3160519 DOI: 10.1002/jcp.22670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peripheral blood-derived multipotent adult progenitor cells (PBD-MAPCs) are a novel population of stem cells, isolated from venous blood of green fluorescent protein transgenic swine, which proliferate as multicellular non-adherent spheroids. Using a simple differentiation protocol, a large proportion of these cells developed one of five distinct neural cell phenotypes, indicating that these primordial cells have high neurogenic potential. Cells exhibiting neural morphologies developed within 48 h of exposure to differentiation conditions, increased in percentage over 2 weeks, and stably maintained the neural phenotype for three additional weeks in the absence of neurogenic signaling molecules. Cells exhibited dynamic neural-like behaviors including extension and retraction of processes with growth cone-like structures rich in filamentous actin, cell migration following a leading process, and various cell-cell interactions. Differentiated cells expressed neural markers, NeuN, β-tubulin III and synaptic proteins, and progenitor cells expressed the stem cell markers nestin and NANOG. Neurally differentiated PBD-MAPCs exhibited voltage-dependent inward and outward currents and expressed voltage-gated sodium and potassium channels, suggestive of neural-like membrane properties. PBD-MAPCs expressed early neural markers and developed neural phenotypes when provided with an extracellular matrix of laminin without the addition of cytokines or growth factors, suggesting that these multipotent cells may be primed for neural differentiation. PBD-MAPCs provide a model for understanding the mechanisms of neural differentiation from non-neural sources of adult stem cells. A similar population of cells, from humans or xenogeneic sources, may offer the potential of an accessible, renewable and non-tumorigenic source of stem cells for treating neural disorders.
Collapse
Affiliation(s)
- Nadja Spitzer
- Department of Biological Sciences, Marshall University, Huntington, West Virginia 25755, USA.
| | | | | | | | | |
Collapse
|
24
|
Myers JP, Santiago-Medina M, Gomez TM. Regulation of axonal outgrowth and pathfinding by integrin-ECM interactions. Dev Neurobiol 2011; 71:901-23. [PMID: 21714101 PMCID: PMC3192254 DOI: 10.1002/dneu.20931] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Developing neurons use a combination of guidance cues to assemble a functional neural network. A variety of proteins immobilized within the extracellular matrix (ECM) provide specific binding sites for integrin receptors on neurons. Integrin receptors on growth cones associate with a number of cytosolic adaptor and signaling proteins that regulate cytoskeletal dynamics and cell adhesion. Recent evidence suggests that soluble growth factors and classic axon guidance cues may direct axon pathfinding by controlling integrin-based adhesion. Moreover, because classic axon guidance cues themselves are immobilized within the ECM and integrins modulate cellular responses to many axon guidance cues, interactions between activated receptors modulate cell signals and adhesion. Ultimately, growth cones control axon outgrowth and pathfinding behaviors by integrating distinct biochemical signals to promote the proper assembly of the nervous system. In this review, we discuss our current understanding how ECM proteins and their associated integrin receptors control neural network formation.
Collapse
Affiliation(s)
- Jonathan P Myers
- Department of Neuroscience, Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
25
|
Integrin signaling in oligodendrocytes and its importance in CNS myelination. JOURNAL OF SIGNAL TRANSDUCTION 2010; 2011:354091. [PMID: 21637375 PMCID: PMC3101883 DOI: 10.1155/2011/354091] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 10/28/2010] [Indexed: 12/22/2022]
Abstract
Multiple sclerosis is characterized by repeated demyelinating attacks of the central nervous system (CNS) white matter tracts. To tailor novel therapeutics to halt or reverse disease process, we require a better understanding of oligodendrocyte biology and of the molecular mechanisms that initiate myelination. Cell extrinsic mechanisms regulate CNS myelination through the interaction of extracellular matrix proteins and their transmembrane receptors. The engagement of one such receptor family, the integrins, initiates intracellular signaling cascades that lead to changes in cell phenotype. Oligodendrocytes express a diverse array of integrins, and the expression of these receptors is developmentally regulated. Integrin-mediated signaling is crucial to the proliferation, survival, and maturation of oligodendrocytes through the activation of downstream signaling pathways involved in cytoskeletal remodeling. Here, we review the current understanding of this important signaling axis and its role in oligodendrocyte biology and ultimately in the myelination of axons within the CNS.
Collapse
|
26
|
Solanki A, Shah S, Memoli KA, Park SY, Hong S, Lee KB. Controlling differentiation of neural stem cells using extracellular matrix protein patterns. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2010; 6:2509-13. [PMID: 20859950 PMCID: PMC2991395 DOI: 10.1002/smll.201001341] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Affiliation(s)
- Aniruddh Solanki
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Shreyas Shah
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Kevin A. Memoli
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Sung Young Park
- Department of Physics, Department of Biophysics and Chemical Biology, Interdisciplinary Program in Nano-Science and Technology, Seoul National University, Seoul, South Korea
| | - Seunghun Hong
- Interdisciplinary Program in Nano-Science and Technology, Seoul National University, Seoul, South Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA, http://rutchem.rutgers.edu/~kbleeweb/
| |
Collapse
|