Neural Stem Cell Systems to Study Brain Development and Diseases

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Stem Cells".

Deadline for manuscript submissions: closed (15 May 2021) | Viewed by 31151

Special Issue Editors

Department of Cellular, Computational and Integrative Biology - CIBIO, Università degli Studi di Trento, Via Sommarive 9, 38123 Trento, Italy
Interests: neural stem cell biology; pluripotent stem cells; iPSCs; neural progenitors; neurons; brain development; neurodevelopmental disorders; neurodegeneration
Hector Institute for Translational Brain Research (HITBR), Central Institute of Mental Health, University of Heidelberg, Medical Faculty Mannheim J5, 68159 Mannheim, Germany
Interests: neural Stem Cell biology; pluripotent stem cell differentiation; iPSCs; neurons; neurodevelopmental disorders; neuropsychiatric disorders
Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, S.S. 12 Abetone e Brennero 4, 56127 Pisa, Italy
Interests: neural stem cell biology; human cortical development; pluripotent stem cell differentiation; iPSCs; neurodevelopmental disorders; brain evolution

Special Issue Information

Dear Colleagues,

Neural stem cells (NSCs) and neural progenitors (NPs) are fundamental players in the tremendous complexity of the mammalian central nervous system (CNS). During development, these cells serve as the ultimate source of all three major neural cell types composing the mature CNS; impairments in the mechanisms controlling their specification, expansion, and behavior result in severe neurodevelopmental and neuropsychiatric disorders.

New developments in genetic and cell technologies have revolutionized how NSCs/NPs are obtained and studied, providing new insights into the decryption of the molecular and cellular codes underlying the NSC programs during neural development and into understanding how the same processes are impaired in brain diseases. This knowledge is pivotal to cell replacement approaches for the damaged brain and the generation of cellular models of brain disorders to better dissect the molecular bases of diseases and for establishing effective drug discovery avenues.

In this Special Issue, we welcome contributions (reviews and original research) covering discoveries that take advantage of the recent advances in 2D and 3D NSC-based systems to investigate neural development, to model CNS diseases, or as tools for providing new therapies for CNS disorders. These include but are not limited to (1) the optimization of new procedures and the investigation of mechanisms for the specification of defined NSC/NP identities from different sources, including their expansion and genetic modification and cell-type-specific maturation; (2) the use of patient-specific NSCs/NPs to model brain disorders; and (3) the exploitation of NSCs systems for drug discovery and cell-based and regenerative therapeutic approaches.

Dr. Luciano Conti
Dr. Philip Koch
Dr. Marco Onorati
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • neural stem cells
  • pluripotent stem cells
  • brain organoids
  • neural progenitors
  • neuronal differentiation
  • brain development
  • neuropsychiatric diseases
  • neurodevelopmental disorders
  • neurodegeneration

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

16 pages, 3277 KiB  
Article
The Combined Treatment with Chemotherapeutic Agents and the Dualsteric Muscarinic Agonist Iper-8-Naphthalimide Affects Drug Resistance in Glioblastoma Stem Cells
by Claudia Guerriero, Carlo Matera, Donatella Del Bufalo, Marco De Amici, Luciano Conti, Clelia Dallanoce and Ada Maria Tata
Cells 2021, 10(8), 1877; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10081877 - 24 Jul 2021
Cited by 8 | Viewed by 2626
Abstract
Background: Glioblastoma multiforme (GBM) is characterized by heterogeneous cell populations. Among these, the Glioblastoma Stem Cells (GSCs) fraction shares some similarities with Neural Stem Cells. GSCs exhibit enhanced resistance to conventional chemotherapy drugs. Our previous studies demonstrated that the activation of M2 muscarinic [...] Read more.
Background: Glioblastoma multiforme (GBM) is characterized by heterogeneous cell populations. Among these, the Glioblastoma Stem Cells (GSCs) fraction shares some similarities with Neural Stem Cells. GSCs exhibit enhanced resistance to conventional chemotherapy drugs. Our previous studies demonstrated that the activation of M2 muscarinic acetylcholine receptors (mAChRs) negatively modulates GSCs proliferation and survival. The aim of the present study was to analyze the ability of the M2 dualsteric agonist Iper-8-naphthalimide (N-8-Iper) to counteract GSCs drug resistance. Methods: Chemosensitivity to M2 dualsteric agonist N-8-Iper and chemotherapy drugs such as temozolomide, doxorubicin, or cisplatin was evaluated in vitro by MTT assay in two different GSC lines. Drug efflux pumps expression was evaluated by RT-PCR and qRT-PCR. Results: By using sub-toxic concentrations of N-8-Iper combined with the individual chemotherapeutic agents, we found that only low doses of the M2 agonist combined with doxorubicin or cisplatin or temozolomide were significantly able to counteract cell growth in both GSC lines. Moreover, we evaluated as the exposure to high and low doses of N-8-Iper downregulated the ATP-binding cassette (ABC) drug efflux pumps expression levels. Conclusions: Our results revealed the ability of the investigated M2 agonist to counteract drug resistance in two GSC lines, at least partially by downregulating the ABC drug efflux pumps expression. The combined effects of low doses of conventional chemotherapy and M2 agonists may thus represent a novel promising pharmacological approach to impair the GSC-drug resistance in the GBM therapy. Full article
(This article belongs to the Special Issue Neural Stem Cell Systems to Study Brain Development and Diseases)
Show Figures

Figure 1

20 pages, 3213 KiB  
Article
FOS Rescues Neuronal Differentiation of Sox2-Deleted Neural Stem Cells by Genome-Wide Regulation of Common SOX2 and AP1(FOS-JUN) Target Genes
by Miriam Pagin, Mattias Pernebrink, Mattia Pitasi, Federica Malighetti, Chew-Yee Ngan, Sergio Ottolenghi, Giulio Pavesi, Claudio Cantù and Silvia K. Nicolis
Cells 2021, 10(7), 1757; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10071757 - 12 Jul 2021
Cited by 6 | Viewed by 3383
Abstract
The transcription factor SOX2 is important for brain development and for neural stem cells (NSC) maintenance. Sox2-deleted (Sox2-del) NSC from neonatal mouse brain are lost after few passages in culture. Two highly expressed genes, Fos and Socs3, are strongly downregulated in [...] Read more.
The transcription factor SOX2 is important for brain development and for neural stem cells (NSC) maintenance. Sox2-deleted (Sox2-del) NSC from neonatal mouse brain are lost after few passages in culture. Two highly expressed genes, Fos and Socs3, are strongly downregulated in Sox2-del NSC; we previously showed that Fos or Socs3 overexpression by lentiviral transduction fully rescues NSC’s long-term maintenance in culture. Sox2-del NSC are severely defective in neuronal production when induced to differentiate. NSC rescued by Sox2 reintroduction correctly differentiate into neurons. Similarly, Fos transduction rescues normal or even increased numbers of immature neurons expressing beta-tubulinIII, but not more differentiated markers (MAP2). Additionally, many cells with both beta-tubulinIII and GFAP expression appear, indicating that FOS stimulates the initial differentiation of a “mixed” neuronal/glial progenitor. The unexpected rescue by FOS suggested that FOS, a SOX2 transcriptional target, might act on neuronal genes, together with SOX2. CUT&RUN analysis to detect genome-wide binding of SOX2, FOS, and JUN (the AP1 complex) revealed that a high proportion of genes expressed in NSC are bound by both SOX2 and AP1. Downregulated genes in Sox2-del NSC are highly enriched in genes that are also expressed in neurons, and a high proportion of the “neuronal” genes are bound by both SOX2 and AP1. Full article
(This article belongs to the Special Issue Neural Stem Cell Systems to Study Brain Development and Diseases)
Show Figures

Graphical abstract

13 pages, 3158 KiB  
Article
Functional Expression of Choline Transporters in Human Neural Stem Cells and Its Link to Cell Proliferation, Cell Viability, and Neurite Outgrowth
by Yosuke Fujita, Tomoki Nagakura, Hiroyuki Uchino, Masato Inazu and Tsuyoshi Yamanaka
Cells 2021, 10(2), 453; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10020453 - 20 Feb 2021
Cited by 4 | Viewed by 3228
Abstract
Choline and choline metabolites are essential for all cellular functions. They have also been reported to be crucial for neural development. In this work, we studied the functional characteristics of the choline uptake system in human neural stem cells (hNSCs). Additionally, we investigated [...] Read more.
Choline and choline metabolites are essential for all cellular functions. They have also been reported to be crucial for neural development. In this work, we studied the functional characteristics of the choline uptake system in human neural stem cells (hNSCs). Additionally, we investigated the effect of extracellular choline uptake inhibition on the cellular activities in hNSCs. We found that the mRNAs and proteins of choline transporter-like protein 1 (CTL1) and CTL2 were expressed at high levels. Immunostaining showed that CTL1 and CTL2 were localized in the cell membrane and partly in the mitochondria, respectively. The uptake of extracellular choline was saturable and performed by a single uptake mechanism, which was Na+-independent and pH-dependent. We conclude that CTL1 is responsible for extracellular choline uptake, and CTL2 may uptake choline in the mitochondria and be involved in DNA methylation via choline oxidation. Extracellular choline uptake inhibition caused intracellular choline deficiency in hNSCs, which suppressed cell proliferation, cell viability, and neurite outgrowth. Our findings contribute to the understanding of the role of choline in neural development as well as the pathogenesis of various neurological diseases caused by choline deficiency or choline uptake impairment. Full article
(This article belongs to the Special Issue Neural Stem Cell Systems to Study Brain Development and Diseases)
Show Figures

Graphical abstract

18 pages, 8427 KiB  
Article
Single-Cell Profiling of Coding and Noncoding Genes in Human Dopamine Neuron Differentiation
by Fredrik Nilsson, Petter Storm, Edoardo Sozzi, David Hidalgo Gil, Marcella Birtele, Yogita Sharma, Malin Parmar and Alessandro Fiorenzano
Cells 2021, 10(1), 137; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10010137 - 12 Jan 2021
Cited by 8 | Viewed by 4365
Abstract
Dopaminergic (DA) neurons derived from human pluripotent stem cells (hPSCs) represent a renewable and available source of cells useful for understanding development, developing disease models, and stem-cell therapies for Parkinson’s disease (PD). To assess the utility of stem cell cultures as an in [...] Read more.
Dopaminergic (DA) neurons derived from human pluripotent stem cells (hPSCs) represent a renewable and available source of cells useful for understanding development, developing disease models, and stem-cell therapies for Parkinson’s disease (PD). To assess the utility of stem cell cultures as an in vitro model system of human DA neurogenesis, we performed high-throughput transcriptional profiling of ~20,000 ventral midbrain (VM)-patterned stem cells at different stages of maturation using droplet-based single-cell RNA sequencing (scRNAseq). Using this dataset, we defined the cellular composition of human VM cultures at different timepoints and found high purity DA progenitor formation at an early stage of differentiation. DA neurons sharing similar molecular identities to those found in authentic DA neurons derived from human fetal VM were the major cell type after two months in culture. We also developed a bioinformatic pipeline that provided a comprehensive long noncoding RNA landscape based on temporal and cell-type specificity, which may contribute to unraveling the intricate regulatory network of coding and noncoding genes in DA neuron differentiation. Our findings serve as a valuable resource to elucidate the molecular steps of development, maturation, and function of human DA neurons, and to identify novel candidate coding and noncoding genes driving specification of progenitors into functionally mature DA neurons. Full article
(This article belongs to the Special Issue Neural Stem Cell Systems to Study Brain Development and Diseases)
Show Figures

Figure 1

13 pages, 3665 KiB  
Article
Loss of ZC4H2 and RNF220 Inhibits Neural Stem Cell Proliferation and Promotes Neuronal Differentiation
by Longlong Zhang, Maosen Ye, Liang Zhu, Jingmei Cha, Chaocui Li, Yong-Gang Yao and Bingyu Mao
Cells 2020, 9(7), 1600; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9071600 - 01 Jul 2020
Cited by 9 | Viewed by 3217
Abstract
The ubiquitin E3 ligase RNF220 and its co-factor ZC4H2 are required for multiple neural developmental processes through different targets, including spinal cord patterning and the development of the cerebellum and the locus coeruleus. Here, we explored the effects of loss of ZC4H2 and [...] Read more.
The ubiquitin E3 ligase RNF220 and its co-factor ZC4H2 are required for multiple neural developmental processes through different targets, including spinal cord patterning and the development of the cerebellum and the locus coeruleus. Here, we explored the effects of loss of ZC4H2 and RNF220 on the proliferation and differentiation of neural stem cells (NSCs) derived from mouse embryonic cortex. We showed that loss of either ZC4H2 or RNF220 inhibits the proliferation and promotes the differentiation abilities of NSCs in vitro. RNA-Seq profiling revealed 132 and 433 differentially expressed genes in the ZC4H2−/− and RNF220−/− NSCs, compared to wild type (WT) NSCs, respectively. Specifically, Cend1, a key regulator of cell cycle exit and differentiation of neuronal precursors, was found to be upregulated in both ZC4H2−/− and RNF220−/− NSCs at the mRNA and protein levels. The targets of Cend1, such as CyclinD1, Notch1 and Hes1, were downregulated both in ZC4H2−/− and RNF220−/− NSCs, whereas p53 and p21 were elevated. ZC4H2−/− and RNF220−/− NSCs showed G0/G1 phase arrest compared to WT NSCs in cell cycle analysis. These results suggested that ZC4H2 and RNF220 are likely involved in the regulation of neural stem cell proliferation and differentiation through Cend1. Full article
(This article belongs to the Special Issue Neural Stem Cell Systems to Study Brain Development and Diseases)
Show Figures

Graphical abstract

18 pages, 7524 KiB  
Article
A Matter of Choice: Inhibition of c-Rel Shifts Neuronal to Oligodendroglial Fate in Human Stem Cells
by Lucia Mercedes Ruiz-Perera, Johannes Friedrich Wilhelm Greiner, Christian Kaltschmidt and Barbara Kaltschmidt
Cells 2020, 9(4), 1037; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9041037 - 22 Apr 2020
Cited by 10 | Viewed by 3341
Abstract
The molecular mechanisms underlying fate decisions of human neural stem cells (hNSCs) between neurogenesis and gliogenesis are critical during neuronal development and neurodegenerative diseases. Despite its crucial role in the murine nervous system, the potential role of the transcription factor NF-κB in the [...] Read more.
The molecular mechanisms underlying fate decisions of human neural stem cells (hNSCs) between neurogenesis and gliogenesis are critical during neuronal development and neurodegenerative diseases. Despite its crucial role in the murine nervous system, the potential role of the transcription factor NF-κB in the neuronal development of hNSCs is poorly understood. Here, we analyzed NF-κB subunit distribution during glutamatergic differentiation of hNSCs originating from neural crest-derived stem cells. We observed several peaks of specific NF-κB subunits. The most prominent nuclear peak was shown by c-REL subunit during a period of 2–5 days after differentiation onset. Furthermore, c-REL inhibition with pentoxifylline (PTXF) resulted in a complete shift towards oligodendroglial fate, as demonstrated by the presence of OLIG2+/O4+-oligodendrocytes, which showed PDGFRα, NG2 and MBP at the transcript level. In addition c-REL impairment further produced a significant decrease in neuronal survival. Transplantation of PTXF-treated predifferentiated hNSCs into an ex vivo oxidative-stress-mediated demyelination model of mouse organotypic cerebellar slices further led to integration in the white matter and differentiation into MBP+ oligodendrocytes, validating their functionality and therapeutic potential. In summary, we present a human cellular model of neuronal differentiation exhibiting a novel essential function of NF-κB-c-REL in fate choice between neurogenesis and oligodendrogenesis which will potentially be relevant for multiple sclerosis and schizophrenia. Full article
(This article belongs to the Special Issue Neural Stem Cell Systems to Study Brain Development and Diseases)
Show Figures

Graphical abstract

Review

Jump to: Research

42 pages, 3369 KiB  
Review
Inducible Pluripotent Stem Cells to Model and Treat Inherited Degenerative Diseases of the Outer Retina: 3D-Organoids Limitations and Bioengineering Solutions
by Massimiliano Andreazzoli, Ivana Barravecchia, Chiara De Cesari, Debora Angeloni and Gian Carlo Demontis
Cells 2021, 10(9), 2489; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10092489 - 20 Sep 2021
Cited by 6 | Viewed by 3712
Abstract
Inherited retinal degenerations (IRD) affecting either photoreceptors or pigment epithelial cells cause progressive visual loss and severe disability, up to complete blindness. Retinal organoids (ROs) technologies opened up the development of human inducible pluripotent stem cells (hiPSC) for disease modeling and replacement therapies. [...] Read more.
Inherited retinal degenerations (IRD) affecting either photoreceptors or pigment epithelial cells cause progressive visual loss and severe disability, up to complete blindness. Retinal organoids (ROs) technologies opened up the development of human inducible pluripotent stem cells (hiPSC) for disease modeling and replacement therapies. However, hiPSC-derived ROs applications to IRD presently display limited maturation and functionality, with most photoreceptors lacking well-developed outer segments (OS) and light responsiveness comparable to their adult retinal counterparts. In this review, we address for the first time the microenvironment where OS mature, i.e., the subretinal space (SRS), and discuss SRS role in photoreceptors metabolic reprogramming required for OS generation. We also address bioengineering issues to improve culture systems proficiency to promote OS maturation in hiPSC-derived ROs. This issue is crucial, as satisfying the demanding metabolic needs of photoreceptors may unleash hiPSC-derived ROs full potential for disease modeling, drug development, and replacement therapies. Full article
(This article belongs to the Special Issue Neural Stem Cell Systems to Study Brain Development and Diseases)
Show Figures

Graphical abstract

31 pages, 2005 KiB  
Review
Human Neural Stem Cell Systems to Explore Pathogen-Related Neurodevelopmental and Neurodegenerative Disorders
by Matteo Baggiani, Maria Teresa Dell’Anno, Mauro Pistello, Luciano Conti and Marco Onorati
Cells 2020, 9(8), 1893; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9081893 - 12 Aug 2020
Cited by 10 | Viewed by 5874
Abstract
Building and functioning of the human brain requires the precise orchestration and execution of myriad molecular and cellular processes, across a multitude of cell types and over an extended period of time. Dysregulation of these processes affects structure and function of the brain [...] Read more.
Building and functioning of the human brain requires the precise orchestration and execution of myriad molecular and cellular processes, across a multitude of cell types and over an extended period of time. Dysregulation of these processes affects structure and function of the brain and can lead to neurodevelopmental, neurological, or psychiatric disorders. Multiple environmental stimuli affect neural stem cells (NSCs) at several levels, thus impairing the normal human neurodevelopmental program. In this review article, we will delineate the main mechanisms of infection adopted by several neurotropic pathogens, and the selective NSC vulnerability. In particular, TORCH agents, i.e., Toxoplasma gondii, others (including Zika virus and Coxsackie virus), Rubella virus, Cytomegalovirus, and Herpes simplex virus, will be considered for their devastating effects on NSC self-renewal with the consequent neural progenitor depletion, the cellular substrate of microcephaly. Moreover, new evidence suggests that some of these agents may also affect the NSC progeny, producing long-term effects in the neuronal lineage. This is evident in the paradigmatic example of the neurodegeneration occurring in Alzheimer’s disease. Full article
(This article belongs to the Special Issue Neural Stem Cell Systems to Study Brain Development and Diseases)
Show Figures

Figure 1

Back to TopTop