1,857 results on '"Neural cell"'
Search Results
102. Modulation of Cell Death in Neural Cell Lines and Transplants
- Author
-
Bredesen, Dale E., Fedoroff, Sergey, editor, Burkholder, Gary D., editor, Juurlink, Bernhard H. J., editor, Krone, Patrick H., editor, Kulyk, William M., editor, Verge, Valerie M. K., editor, and Doucette, J. Ronald, editor
- Published
- 1995
- Full Text
- View/download PDF
103. Use of Human Brain Cell Aggregates for AIDS Research in the Nervous System
- Author
-
Pulliam, Lynn, Major, Eugene O., editor, Levy, Jay A., editor, and Schoenberg, Devera, editor
- Published
- 1995
- Full Text
- View/download PDF
104. The effects of fetal neural cell conditioned medium on cell proliferation in the rat brain after traumatic brain injury
- Author
-
O. Tsupykov, M. Lisyany, D. Stanetska, and I. Govbakh
- Subjects
Transplantation ,Fetus ,business.industry ,Cell growth ,Traumatic brain injury ,Biomedical Engineering ,Rat brain ,medicine.disease ,nervous system ,Conditioned medium ,Immunology and Allergy ,Medicine ,business ,Neuroscience ,Neural cell ,Biotechnology - Abstract
Traumatic brain injury (TBI) is accompanied by an increase in the number of proliferating cells. However, the question of the nature, conditions of production and mechanisms of action of humoral factors secreted by fetal neural cells (FNCs) on reparative processes and neurogenesis in the brain after trauma and FNCs transplantation remains open. The purpose of the study was to establish the possibility of the influence of the conditioned medium of fetal neural cell cultures on the proliferative activity of Ki-67-positive cells in the cortex and subcortical structures of the rat brain after TBI. Materials and methods. TBI was simulated by dropping a metal cylinder on the rat’s head. Rats (E17-18) were used to obtain cultures of neural stem/progenitor cells. Conditioned media from cell cultures with high adhesive properties (HA-CM) and low adhesive properties (LA-CM) were used to treat the effects of experimental TBI in rats by intramuscular injection. The effect of conditioned media on the proliferative activity of Ki-67-positive cells in the cortex and subcortical structures of the brain after TBI was determined by immunohistochemical analysis using antibodies against Ki-67 protein. Results. Immunohistochemical analysis of the brain sections showed that on the 5th day after traumatic brain injury in rats there was a probable increase in the number of Ki-67-positive cells in the cortex, hippocampus and thalamus. It was found that the injection of HA-CM or LA-CM in animals with TBI increased the number of Ki-67-positive cells in the hippocampus compared with the TBI group and their value for the TBI+LA-CM group reached 59.6 ± 6.1, and for the TBI+HA-CM group – 47.2 ± 3.1 cells (p
- Published
- 2021
105. Building on a Solid Foundation: Adding Relevance and Reproducibility to Neurological Modeling Using Human Pluripotent Stem Cells
- Author
-
Erin Knock and Lisa M. Julian
- Subjects
Brain development ,Computer science ,Cell replacement ,regenerative medicine ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Human brain ,Regenerative medicine ,Neural stem cell ,Cellular and Molecular Neuroscience ,medicine.anatomical_structure ,Cellular Neuroscience ,human brain development ,disease modeling ,medicine ,Relevance (information retrieval) ,Systematic Review ,human pluripotent stem cells ,Induced pluripotent stem cell ,Neural cell ,Neuroscience ,RC321-571 ,neural stem cells - Abstract
The brain is our most complex and least understood organ. Animal models have long been the most versatile tools available to dissect brain form and function; however, the human brain is highly distinct from that of standard model organisms. In addition to existing models, access to human brain cells and tissues is essential to reach new frontiers in our understanding of the human brain and how to intervene therapeutically in the face of disease or injury. In this review, we discuss current and developing culture models of human neural tissue, outlining advantages over animal models and key challenges that remain to be overcome. Our principal focus is on advances in engineering neural cells and tissue constructs from human pluripotent stem cells (PSCs), though primary human cell and slice culture are also discussed. By highlighting studies that combine animal models and human neural cell culture techniques, we endeavor to demonstrate that clever use of these orthogonal model systems produces more reproducible, physiological, and clinically relevant data than either approach alone. We provide examples across a range of topics in neuroscience research including brain development, injury, and cancer, neurodegenerative diseases, and psychiatric conditions. Finally, as testing of PSC-derived neurons for cell replacement therapy progresses, we touch on the advancements that are needed to make this a clinical mainstay.
- Published
- 2021
- Full Text
- View/download PDF
106. Interventional Strategies for Parkinson Disease: Can Neural Precursor Cells Forge a Path Ahead?
- Author
-
Swapna Nandakumar, Rajarshi Pal, Indrani Datta, and Pradnya Shahani
- Subjects
Neurons ,Cell type ,Physiology ,SARS-CoV-2 ,Cognitive Neuroscience ,Wnt signaling pathway ,COVID-19 ,Parkinson Disease ,Cell Biology ,General Medicine ,Biology ,Biochemistry ,Neuroprotection ,Neural stem cell ,Rats ,Transplantation ,Cell therapy ,Neural Stem Cells ,Animals ,Humans ,Induced pluripotent stem cell ,Neural cell ,Neuroscience - Abstract
Neural precursor cells (NPCs), derived from pluripotent stem cells (PSCs), with their unique ability to generate multiple neuronal and glial cell types are extremely useful for understanding biological mechanisms in normal and diseased states. However, generation of specific neuronal subtypes (mature) from NPCs in large numbers adequate for cell therapy is challenging due to lack of a thorough understanding of the cues that govern their differentiation. Interestingly, neural stem cells (NSCs) themselves are in consideration for therapy given their potency to form different neural cell types, release of trophic factors, and immunomodulatory effects that confer neuroprotection. With the recent COVID-19 outbreak and its accompanying neurological indications, the immunomodulatory role of NSCs may gain additional significance in the prevention of disease progression in vulnerable populations. In this regard, small-molecule mediated NPC generation from PSCs via NSC formation has become an important strategy that ensures consistency and robustness of the process. The development of the mammalian brain occurs along the rostro-caudal axis, and the establishment of anterior identity is an early event. Wnt signaling, along with fibroblast growth factor and retinoic acid, acts as a caudalization signal. Further, the increasing amount of epigenetic data available from human fetal brain development has enhanced both our understanding of and ability to experimentally manipulate these developmental regulatory programs in vitro. However, the impact on homing and engraftment after transplantation and subsequently on therapeutic efficacy of NPCs based on their derivation strategy is not yet clear. Another formidable challenge in cell replacement therapy for neurodegenerative disorders is the mode of delivery. In this Perspective, we discuss these core ideas with insights from our preliminary studies exploring the role of PSC-derived NPCs in rat models of MPTP-induced Parkinson's disease following intranasal injections.
- Published
- 2021
107. An atlas of gene regulatory elements in adult mouse cerebrum
- Author
-
Jee Yun Han, Yunjiang Qiu, Michael Nunn, Antonio Pinto-Duarte, M. Margarita Behrens, Bing Ren, Ziyang Zhang, Naoki Kubo, Joseph R. Ecker, Sebastian Preissl, Yang Eric Li, Xinxin Wang, David U. Gorkin, Olivier Poirion, Kai Zhang, Kyle J. Gaulton, Hanqing Liu, Xiaomeng Hou, Yin Shen, Bin Li, Eran A. Mukamel, Yiming Yan, Michael Miller, Joshua Chiou, Rongxin Fang, Xiaoyu Yang, Jacinta Lucero, and Samantha Kuan
- Subjects
Epigenomics ,Male ,Regulatory Sequences, Nucleic Acid ,Inbred C57BL ,Mice ,Epigenetics in the nervous system ,Neural cell ,Neurons ,Multidisciplinary ,Cerebrum ,Chromatin ,medicine.anatomical_structure ,Regulatory sequence ,Neurological ,Stem Cell Research - Nonembryonic - Non-Human ,Single-Cell Analysis ,Sequence Analysis ,Neuroglia ,Biotechnology ,Cell type ,General Science & Technology ,1.1 Normal biological development and functioning ,Biology ,Article ,Atlases as Topic ,Underpinning research ,medicine ,Genetics ,Animals ,Humans ,Genetic Predisposition to Disease ,Gene ,Nucleic Acid ,Human Genome ,Neurosciences ,DNA ,Sequence Analysis, DNA ,Chromatin Assembly and Disassembly ,Stem Cell Research ,Olfactory bulb ,Brain Disorders ,Mice, Inbred C57BL ,Gene Expression Regulation ,Nervous System Diseases ,Regulatory Sequences ,Neuroscience - Abstract
The mammalian cerebrum performs high-level sensory perception, motor control and cognitive functions through highly specialized cortical and subcortical structures1. Recent surveys of mouse and human brains with single-cell transcriptomics2–6 and high-throughput imaging technologies7,8 have uncovered hundreds of neural cell types distributed in different brain regions, but the transcriptional regulatory programs that are responsible for the unique identity and function of each cell type remain unknown. Here we probe the accessible chromatin in more than 800,000 individual nuclei from 45 regions that span the adult mouse isocortex, olfactory bulb, hippocampus and cerebral nuclei, and use the resulting data to map the state of 491,818 candidate cis-regulatory DNA elements in 160 distinct cell types. We find high specificity of spatial distribution for not only excitatory neurons, but also most classes of inhibitory neurons and a subset of glial cell types. We characterize the gene regulatory sequences associated with the regional specificity within these cell types. We further link a considerable fraction of the cis-regulatory elements to putative target genes expressed in diverse cerebral cell types and predict transcriptional regulators that are involved in a broad spectrum of molecular and cellular pathways in different neuronal and glial cell populations. Our results provide a foundation for comprehensive analysis of gene regulatory programs of the mammalian brain and assist in the interpretation of noncoding risk variants associated with various neurological diseases and traits in humans., A comprehensive analysis of gene regulatory elements in 160 distinct cell types from the mouse cerebrum.
- Published
- 2021
108. Phenotypic variation of transcriptomic cell types in mouse motor cortex
- Author
-
Federico Scala, Leonard Hartmanis, Rickard Sandberg, Dmitry Kobak, Zheng Huan Tan, Sophie Laturnus, Philipp Berens, Jesus Ramon Castro, Hongkui Zeng, Yves Bernaerts, Xiaolong Jiang, Shalaka Mulherkar, Zizhen Yao, Matteo Bernabucci, Andreas S. Tolias, Elanine Miranda, and Cathryn R. Cadwell
- Subjects
Male ,Cell type ,Patch-Clamp Techniques ,General Science & Technology ,Biology ,Article ,chemistry.chemical_compound ,Mice ,Atlases as Topic ,Glutamates ,Cellular neuroscience ,Biocytin ,medicine ,Genetics ,Animals ,GABAergic Neurons ,Neural cell ,Neurons ,Multidisciplinary ,Neocortex ,Staining and Labeling ,Sequence Analysis, RNA ,Lysine ,Gene Expression Profiling ,Motor Cortex ,Neurosciences ,Phenotype ,Brain Disorders ,medicine.anatomical_structure ,chemistry ,Organ Specificity ,Neurological ,RNA ,Female ,Primary motor cortex ,Single-Cell Analysis ,Transcriptome ,Neuroscience ,Sequence Analysis ,Motor cortex - Abstract
Cortical neurons exhibit extreme diversity in gene expression as well as in morphological and electrophysiological properties1,2. Most existing neural taxonomies are based on either transcriptomic3,4 or morpho-electric5,6 criteria, as it has been technically challenging to study both aspects of neuronal diversity in the same set of cells7. Here we used Patch-seq8 to combine patch-clamp recording, biocytin staining, and single-cell RNA sequencing of more than 1,300 neurons in adult mouse primary motor cortex, providing a morpho-electric annotation of almost all transcriptomically defined neural cell types. We found that, although broad families of transcriptomic types (those expressing Vip, Pvalb, Sst and so on) had distinct and essentially non-overlapping morpho-electric phenotypes, individual transcriptomic types within the same family were not well separated in the morpho-electric space. Instead, there was a continuum of variability in morphology and electrophysiology, with neighbouring transcriptomic cell types showing similar morpho-electric features, often without clear boundaries between them. Our results suggest that neuronal types in the neocortex do not always form discrete entities. Instead, neurons form a hierarchy that consists of distinct non-overlapping branches at the level of families, but can form continuous and correlated transcriptomic and morpho-electrical landscapes within families.
- Published
- 2021
109. Evolution of Synapses and Neurotransmitter Systems: The Divide-and-Conquer Model for Early Neural Cell-Type Evolution
- Author
-
Pawel Burkhardt and Gáspár Jékely
- Subjects
Neurons ,Neurotransmitter Agents ,Lineage (genetic) ,biology ,General Neuroscience ,Ctenophora ,Sequencing data ,other ,Neurotransmitter systems ,biology.organism_classification ,Biological Evolution ,Homology (biology) ,Synapse ,Evolutionary biology ,Synapses ,Cambrian explosion ,Animals ,Homeobox ,Neuroscience ,Bilateria ,Neural cell - Abstract
Nervous systems evolved around 560 million years ago to coordinate and empower animal bodies. Ctenophores – one of the earliest-branching lineages – are thought to share few neuronal genes with bilaterians and may have evolved neurons convergently. Here we review our current understanding of the evolution of neuronal molecules in non-bilaterians. We also reanalyse single-cell sequencing data in light of new cell-cluster identities from a ctenophore and uncover evidence supporting the homology of one ctenophore neuron-type with neurons in Bilateria. The specific coexpression of the presynaptic proteins Unc13 and RIM with voltage-gated channels, neuropeptides and homeobox genes pinpoint a spiking sensory-peptidergic cell in the ctenophore mouth. Similar Unc13-RIM neurons may have been present in the first eumetazoans to rise to dominance only in stem Bilateria. We hypothesize that the Unc13-RIM lineage ancestrally innervated the mouth and conquered other parts of the body with the rise of macrophagy and predation during the Cambrian explosion.
- Published
- 2021
110. Culturing Mouse Fetal Neural Precursor Cells in a Free-Floating Serum-Free Condition
- Author
-
Hassan Azari
- Subjects
Fetus ,medicine.anatomical_structure ,In vivo ,Precursor cell ,Neurosphere ,Central nervous system ,Cell ,otorhinolaryngologic diseases ,medicine ,Biology ,Neural cell ,In vitro ,Cell biology - Abstract
Neural precursor cells (NPCs) are a renewable cell source that can proliferate and expand for long periods of time and give rise to the main neural cell types of the central nervous system (CNS). Establishing simple and reproducible growth culture conditions is of great importance to study the biology of NPCs and to understand the molecular basis of their behavior in healthy and diseased conditions.Here, we describe a simple free-floating , serum-free culture condition, known as the neurosphere assay, which is the most commonly used method for the isolation and expansion of NPCs harvested from the adult and fetal CNS. This culture system will result in large numbers of undifferentiated NPC progenies that represents a useful cell source for many in vitro and in vivo applications.
- Published
- 2021
111. Isolation and Enrichment of Defined Neural Cell Populations from Heterogeneous Neural Stem Cell Progeny
- Author
-
Hassan Azari
- Subjects
education.field_of_study ,medicine.diagnostic_test ,Isolation (health care) ,Cell ,Population ,Biology ,Neural stem cell ,nervous system diseases ,Flow cytometry ,Cell biology ,Transplantation ,Cell therapy ,medicine.anatomical_structure ,nervous system ,medicine ,biological phenomena, cell phenomena, and immunity ,Progenitor cell ,education ,Neural cell ,reproductive and urinary physiology - Abstract
The renewable source of neural stem cells (NSCs) with multi-lineage differentiation capability toward neurons, astrocytes, and oligodendrocytes represents an ideal supply for cell therapy of central nervous system (CNS) diseases. In spite of this, the clinical use of NSCs is hampered by heterogeneity, poor neuronal cell yield, predominant astrocytic differentiation of NSC progeny, and possible uncontrolled proliferation and tumor formation upon transplantation. The ability to generate highly enriched and defined neural cell populations from the renewable source of NSCs might overcome many of these impediments and pave the way toward their successful clinical applications.Here, we describe a simple method for NSC differentiation and subsequent purification of neuronal progenitor cells, taking advantage of size and granularity differences between neuronal cells and other NSC progeny. This highly enriched neuronal cell population provides an invaluable source of cells for both in vitro and in vivo studies.
- Published
- 2021
112. The Toxicity Evaluation of Lycopodiacea Extracts by Mice Neural Cell and Zebrafish Embryo
- Author
-
Nguyen Thi Kim Thanh, Hoang Thi My Hanh, Nguyen Lai Thanh, Nguyen Tien Dat, Pham Thi Huyen Trang, and Kieu Trung Kien
- Subjects
Toxicity ,Zebrafish embryo ,Biology ,Neural cell ,Cell biology - Abstract
Many species in the family Lycopodiacea possess highly potential medical substances, notably are Huperzine A of Huperzia serrata and alkaloids isolated from Lycopodium clavatum(α-Onocerin , Lycopodine,..). Various members of this family are found throughout of Vietnam and a few researches on its diversity as well as alkaloid isolation has been conducted, however, there is a lack of toxicology studies on this subject. In this study, three crude extracts (labeled AI.1, AI.2 and AI.3), which come from Lycopodium sp.collected from 3 different regions of Vietnam, have been tested on 2 toxicological models: primary neural cells derived from Swiss white mouse (in vitro) and Zebrafish (Danio rerio) embryos (in vivo). Our results show that the all three extracts are relatively low in toxicity, IC50 value on primary neural cell ranging from 800 - 1100 mg/L, while LC50 of fish embryos at 100 - 300 mg/L. AI.2 has the least effect of viability of both cells and embryos compare to the others. This data give the prove showed that the safety using Lycopodium as tradional medicine.
- Published
- 2021
113. Maternal exposure to polystyrene nanoplastics causes brain abnormalities in progeny
- Author
-
Young-Kyoung Ryu, Tae Kyung Kim, Jae-Ran Lee, ChiHye Chung, Jeong Yeob Baek, Nam-Soon Kim, Rumeysa Dogan, Jahong Koo, Dahun Um, Won-Ho Shin, Bohyeon Jeong, Hyung-Seok Choi, Jinyoung Jeong, Seungjae Zhang, Kyoung-Shim Kim, Subin Park, Da Yong Lee, and Wang Sik Lee
- Subjects
Microplastics ,Environmental Engineering ,Brain development ,Health, Toxicology and Mutagenesis ,Central nervous system ,Biology ,Mice ,medicine ,Environmental Chemistry ,Animals ,Humans ,Lactation ,Waste Management and Disposal ,Neural cell ,Cognitive deficit ,Ecosystem ,Brain ,Pollution ,Neural stem cell ,In vitro ,Cell biology ,medicine.anatomical_structure ,Maternal Exposure ,Polystyrenes ,Female ,medicine.symptom ,Plastics ,Water Pollutants, Chemical - Abstract
As global plastic production continues to grow, microplastics released from a massive quantity of plastic wastes have become a critical environmental concern. These microplastic particles are found in a wide range of living organisms in a diverse array of ecosystems. In this study, we investigated the biological effects of polystyrene nanoplastics (PSNPs) on development of the central nervous system using cultured neural stem cells (NSCs) and mice exposed to PSNPs during developmental stages. Our study demonstrates that maternal administration of PSNPs during gestation and lactating periods altered the functioning of NSCs, neural cell compositions, and brain histology in progeny. Similarly, our in vitro study also shows PSNP-induced molecular and functional defects in NSCs. Finally, we show that the abnormal brain development caused by exposure to high concentrations of PSNPs results in neurophysiological and cognitive deficits in a gender-specific manner. Our data demonstrate the possibility that exposure to high amounts of PSNPs may increase the risk of neurodevelopmental defects.
- Published
- 2021
114. Identification of Adeno-Associated Virus Variants for Gene Transfer Into Human Neural Cell Types by Parallel Capsid Screening
- Author
-
Lea Jessica Flitsch, Christian Stüllein, Si Wah Christina Au Yeung, Oliver Brüstle, Simon Ziegler, Julia Schlee, Kathleen Börner, Mona Mathews, Vesselina Semkova, Vera Sonntag-Buck, Mohamad Hajo, and Dirk Grimm
- Subjects
Capsid ,viruses ,medicine ,Identification (biology) ,Gene transfer ,Computational biology ,Biology ,medicine.disease_cause ,Adeno-associated virus ,Neural cell - Abstract
Human brain cells generated by in vitrocell programming provide exciting prospects for disease modeling, drug discovery and cell therapy. These applications frequently require efficient and clinically compliant tools for genetic modification of the cells. Recombinant Adeno-associated viruses (AAVs) fulfill these prerequisites for a number of reasons, including the availability of a myriad of AAV capsid variants with distinct cell type specificity (also called tropism). Here, weharnessed a customizable parallel screening approach to assessa panel of natural or synthetic AAV capsid variants for their efficacy in lineage-related human neural cell types.We identified common lead candidates suited for the transduction of directly converted,early-stage induced neural stem cells (iNSCs), induced pluripotent stem cell (iPSC)-derived later-stage, radial glia-like neuralprogenitors,as well as differentiated astrocytic and mixed neuroglial cultures.We then selected a subsetof these candidates for functional validation in iNSCs and iPSC-derived astrocytes, usingshRNA-induced downregulation of the citrate transporter SLC25A1 and overexpression of the transcription factor NGN2 for proofs-of-concept. Our study provides a comparative overview ofthe susceptibility of different human cell programming-derived brain cell types to AAV transduction and a critical discussionof the assets and limitations of the specific AAV capsidscreening approach.
- Published
- 2021
115. On-Chip Neural Cell-Cultivation System for Long-Term Observation with Multi-Electrode and Microchamber Arrays
- Author
-
Takahashi, Kazunori, Sugio, Yoshihiro, Moriguchi, Hiroyuki, Jimbo, Yasuhiko, Yasuda, Kenji, Baba, Yoshinobu, editor, Shoji, Shuichi, editor, and van den Berg, Albert, editor
- Published
- 2002
- Full Text
- View/download PDF
116. Poliovirus Neurovirulence and its Attenuation
- Author
-
Agol, Vadim I., Carrasco, Luis, editor, Sonenberg, Nahum, editor, and Wimmer, Eckard, editor
- Published
- 1993
- Full Text
- View/download PDF
117. Neural Constraints on Attention
- Author
-
Cohen, Ronald A., Puente, Antonio E., editor, Reynolds, Cecil R., editor, and Cohen, Ronald A.
- Published
- 1993
- Full Text
- View/download PDF
118. The Role of Human Immunodeficiency Virus Type 1 (HIV-1) in Neurologic Disorders of AIDS
- Author
-
Volsky, David J., Shahabuddin, Muhammad, Mizrachi, Yaffa, and Roos, Raymond P., editor
- Published
- 1992
- Full Text
- View/download PDF
119. The cadmium altered oxidative homeostasis leads to energetic metabolism rearrangement, Nrf2 activation with increased GSH production and reduced SOD1 activity in neural cells
- Author
-
Bovio, F, FUSI, PAOLA ALESSANDRA, LOTTI, MARINA, BOVIO, FEDERICA, Bovio, F, FUSI, PAOLA ALESSANDRA, LOTTI, MARINA, and BOVIO, FEDERICA
- Abstract
Il cadmio, elemento chimico ampiamente usato in ambito industriale, è considerato un contaminante ambientale con effetti tossici sugli organismi viventi. Il suo ingresso nel corpo umano può avvenire per inalazione o ingestione di cibi ed acqua contaminati, fumo di sigaretta o impiego professionale, con tratto respiratorio e gastrointestinale principalmente coinvolti nel suo assorbimento cellulare. Anche il cervello è un bersaglio della tossicità del cadmio, che può entrare nel sistema nervoso centrale tramite una maggiore permeabilità della barriera ematoencefalica o attraverso i nervi olfattivi. Infatti, l'esposizione al cadmio è stata correlata sia ad alterazioni funzionali del sistema nervoso sia a malattie neurodegenerative, come la sclerosi laterale amiotrofica (SLA). Il 90-95% dei casi di SLA sono sporadici (sALS), mentre il restante 5-10% ha origine familiare (fALS), di cui il 15-20% è attribuito a mutazioni nel gene dell’enzima antiossidante superossido dismutasi 1 (SOD1). SOD1 è un omodimero di 32 kDa, in cui ciascun monomero presenta un ponte disulfuro e due ioni metallici, il rame con ruolo catalitico e lo zinco con funzione strutturale. Poiché uno dei principali meccanismi con cui il cadmio esercita la propria tossicità è lo stress ossidativo, responsabile di un insieme di eventi avversi che culminano nella morte cellulare, scopo di questa tesi è lo studio dell'effetto neurotossico del cadmio sul metabolismo energetico nella linea cellulare umana SH-SY5Y, sulle difese antiossidanti in cellule LUHMES differenziate e sulla funzione di SOD1 in tre modelli sperimentali (proteina ricombinante in E. coli, linea cellulare SH-SY5Y e nematode Caenorhabditis elegans). La valutazione del metabolismo energetico in cellule SH-SY5Y trattate per 24 ore con dosi sub-letali di CdCl2 ha evidenziato il passaggio ad un metabolismo anaerobico; infatti cellule trattate mostrano un aumento della glicolisi, una maggiore produzione di ATP per via glicolitica e una r, The heavy metal cadmium is a widespread toxic pollutant, released into the environment mainly by anthropogenic activities. Human exposure can occur through different sources: occupationally or environmentally, with its uptake through inhalation of polluted air, cigarette smoking or ingestion of contaminated food and water. It mainly enters the human body through the respiratory and the gastrointestinal tract and it accumulates in liver and kidneys. Brain is also a target of cadmium toxicity, since this toxicant may enter the central nervous system by increasing blood brain barrier permeability or through the olfactory nerves. In fact, cadmium exposure has been related to impaired functions of the nervous system and to neurodegenerative diseases, like amyotrophic lateral sclerosis (ALS). ALS is a fatal motor neuron pathology with the 90-95% of ALS cases being sporadic (sALS), while the remaining 5-10% of familial onset (fALS); among fALS, the 15-20% is attributed to mutations in superoxide dismutase 1 (SOD1). SOD1 is an antioxidant protein responsible for superoxide anions disruption and it is a homodimeric metalloenzyme of 32 kDa mainly located in the cytoplasm, with each monomer binding one catalytic copper ion and one structural zinc ion within a disulfide bonded conformer. Since oxidative stress is one of the major mechanisms of cadmium induced toxicity and an alteration of oxidative homeostasis, through depletion of antioxidant defences, is responsible for a plethora of adverse outcoming mainly leading to cell death; we focused on cadmium effect (1) on the energetic metabolism in human neuroblastoma SH-SY5Y cell line, (2) on the oxidative defences responses in differentiated human LUHMES neural cell line and (3) on the function of human SOD1 in a three models approach (recombinant protein in E. coli, in SH-SY5Y cell line and in the nematode Caenorhabditis elegans). The evaluation of energetic metabolism of SH-SY5Y neural cells treated with sub-lethal CdCl2 doses
- Published
- 2021
120. In Silico Analysis to Explore Lineage-Independent and -Dependent Transcriptional Programs Associated with the Process of Endothelial and Neural Differentiation of Human Induced Pluripotent Stem Cells
- Author
-
Maryam Nakhaei-Nejad, Hosna Jabbari, Manijeh Pasdar, Nadia Jahroudi, and Luke Trinity
- Subjects
induced pluripotent stem cells ,Article ,03 medical and health sciences ,0302 clinical medicine ,transcription factors ,Medicine ,Epigenetics ,differentiation ,endothelial cells ,neural cells ,epigenetic regulators ,Induced pluripotent stem cell ,Transcription factor ,Neural cell ,030304 developmental biology ,0303 health sciences ,business.industry ,General Medicine ,Phenotype ,Cell biology ,Endothelial stem cell ,DNA methylation ,Stem cell ,business ,030217 neurology & neurosurgery - Abstract
Despite a major interest in understanding how the endothelial cell phenotype is established, the underlying molecular basis of this process is not yet fully understood. We have previously reported the generation of induced pluripotent stem cells (iPS) from human umbilical vein endothelial cells and differentiation of the resulting HiPS back to endothelial cells (Ec-Diff), as well as neural (Nn-Diff) cell lineage that contained both neurons and astrocytes. Furthermore, the identities of these cell lineages were established by gene array analysis. Here, we explored the same arrays to gain insight into the gene alteration processes that accompany the establishment of endothelial vs. non-endothelial neural cell phenotypes. We compared the expression of genes that code for transcription factors and epigenetic regulators when HiPS is differentiated into these endothelial and non-endothelial lineages. Our in silico analyses have identified cohorts of genes that are similarly up- or downregulated in both lineages, as well as those that exhibit lineage-specific alterations. Based on these results, we propose that genes that are similarly altered in both lineages participate in priming the stem cell for differentiation in a lineage-independent manner, whereas those that are differentially altered in endothelial compared to neural cells participate in a lineage-specific differentiation process. Specific GATA family members and their cofactors and epigenetic regulators (DNMT3B, PRDM14, HELLS) with a major role in regulating DNA methylation were among participants in priming HiPS for lineage-independent differentiation. In addition, we identified distinct cohorts of transcription factors and epigenetic regulators whose alterations correlated specifically with the establishment of endothelial vs. non-endothelial neural lineages.
- Published
- 2021
121. Micromechanical Characterisation of 3D Bioprinted neural cell models using Brillouin Microscopy
- Author
-
Joanne L. Tipper, H. Mahmodi, Irina V. Kabakova, Maryam Alsadat Rad, Elysse C. Filipe, and Thomas R. Cox
- Subjects
Brillouin zone ,Microscopy ,Self-healing hydrogels ,Biological system ,Neural cell ,Regenerative medicine ,Viscoelasticity ,Microscale chemistry ,Biofabrication - Abstract
Biofabrication of artificial 3D in vitro neural cell models that closely mimic the central nervous system (CNS) is an emerging field of research with applications from fundamental biology to regenerative medicine, and far reaching benefits for the economy, healthcare and the ethical use of animals. The micromechanical properties of such models are an important factor dictating the success of modelling outcomes in relation to accurate reproduction of the processes in native tissues. Characterising the micromechanical properties of such models non-destructively and over a prolonged span of time, however, are key challenges. Brillouin microscopy (BM) could provide a solution to this problem since this technology is non-invasive, label-free and is capable of microscale 3D imaging. In this work, the viscoelasticity of 3D bioprinted neural cell models consisting of NG 108-15 neuronal cells and GelMA hydrogels of various concentrations were investigated using BM. We demonstrate changes in the micro- and macro-scale mechanical properties of these models over a 7 day period, in which the hydrogel component of the model are found to soften as the cells grow, multiply and form stiffer spheroid-type structures. These findings signify the necessity to resolve in microscopic detail the mechanics of in vitro 3D tissue models and suggest Brillouin microscopy to be a suitable technology to bridge this gap.
- Published
- 2021
122. Transcription Factor-Based Strategies to Generate Neural Cell Types from Human Pluripotent Stem Cells
- Author
-
Isaac Canals, Ella Quist, and Henrik Ahlenius
- Subjects
Neurons ,Pluripotent Stem Cells ,Microglia ,Cell ,Dopaminergic ,Cell Differentiation ,Cell Biology ,Biology ,Inhibitory postsynaptic potential ,medicine.anatomical_structure ,medicine ,Humans ,Cell Lineage ,Induced pluripotent stem cell ,Reprogramming ,Neural cell ,Neuroscience ,Transcription factor ,Developmental Biology ,Biotechnology ,Transcription Factors - Abstract
In the last years, the use of pluripotent stem cells in studies of human biology has grown exponentially. These cells represent an infinite source for differentiation into several human cell types facilitating the investigation on biological processes, functionality of cells, or diseases mechanisms in relevant human models. In the neurobiology field, pluripotent stem cells have been extensively used to generate the main neuronal and glial cells of the brain. Traditionally, protocols following developmental cues have been applied to pluripotent stem cells to drive differentiation toward different cell lineages; however, these protocols give rise to populations with mixed identities. Interestingly, new protocols applying overexpression of lineage-specific transcription factors (TFs) have emerged and facilitated the generation of highly pure populations of specific subtypes of neurons and glial cells in an easy, reproducible, and rapid manner. In this study, we review protocols based on this strategy to generate excitatory, inhibitory, dopaminergic, and motor neurons as well as astrocytes, oligodendrocytes, and microglia. In addition, we will discuss the main applications for cells generated with these protocols, including disease modeling, drug screening, and mechanistic studies. Finally, we will discuss the advantages and disadvantages of TF-based protocols and present our view of the future in this field.
- Published
- 2021
123. Astrocyte-neuron metabolic cooperation shapes brain activity
- Author
-
Gilles Bonvento, Juan P. Bolaños, Centre National de la Recherche Scientifique (France), Commissariat à l'Ènergie Atomique et aux Ènergies Alternatives (France), Agence Nationale de la Recherche (France), French National Foundation on Alzheimer’s Disease and Related Disorders, Fondation Alzheimer, Agencia Estatal de Investigación (España), Instituto de Salud Carlos III, Junta de Castilla y León, Fundación BBVA, Fundación Ramón Areces, Service MIRCEN (MIRCEN), Université Paris-Saclay-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie François JACOB (JACOB), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA), and Association France Alzheimer, Fondation de France
- Subjects
chemistry.chemical_classification ,Neurons ,Reactive oxygen species ,Physiology ,[SDV]Life Sciences [q-bio] ,Brain ,Cell Biology ,Biology ,Neurotransmission ,medicine.anatomical_structure ,chemistry ,Astrocytes ,medicine ,Humans ,Glycolysis ,Neuron ,Energy Metabolism ,Molecular Biology ,Neuroscience ,Neural cell ,Intracellular ,Function (biology) ,Astrocyte - Abstract
The brain has almost no energy reserve, but its activity coordinates organismal function, a burden that requires precise coupling between neurotransmission and energy metabolism. Deciphering how the brain accomplishes this complex task is crucial to understand central facets of human physiology and disease mechanisms. Each type of neural cell displays a peculiar metabolic signature, forcing the intercellular exchange of metabolites that serve as both energy precursors and paracrine signals. The paradigm of this biological feature is the astrocyte-neuron couple, in which the glycolytic metabolism of astrocytes contrasts with the mitochondrial oxidative activity of neurons. Astrocytes generate abundant mitochondrial reactive oxygen species and shuttle to neurons glycolytically derived metabolites, such as L-lactate and L-serine, which sustain energy needs, conserve redox status, and modulate neurotransmitter-receptor activity. Conversely, early disruption of this metabolic cooperation may contribute to the initiation or progression of several neurological diseases, thus requiring innovative therapies to preserve brain energetics., G.B. received grants from the Centre National de la Recherche Scientifique (CNRS), the Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Agence Nationale de la Recherche (ANR 2011 MALZ-0003 MetALZ; ANR-18-C816-0008-03 ADORASTrAU), Association France Alzheimer, Fondation de France (Prix Spécial 2012), Fondation Alzheimer, and Infrastructure de Recherche Translationnelle pour les Biothérapies en Neurosciences (NeurATRIS ANR-11-INBS-0011). J.P.B. is funded by the Agencia Estatal de Investigación (PID2019-105699RB-I00 / AEI / 10.13039/501100011033 and RED2018-102576-T), Instituto de Salud Carlos III (CB16/10/00282), Junta de Castilla y León (CSI151P20 and Escalera de Excelencia CLU-2017-03), Ayudas Equipos Investigación Biomedicina 2017 Fundación BBVA, and Fundación Ramón Areces.
- Published
- 2021
124. Neural Tissue Homeostasis and Repair Is Regulated via CS and DS Proteoglycan Motifs
- Author
-
Anthony J. Hayes and James Melrose
- Subjects
0301 basic medicine ,QH301-705.5 ,Neuroregulation ,Morphogenesis ,Review ,dermatan sulfate ,Extracellular matrix ,Cell and Developmental Biology ,03 medical and health sciences ,0302 clinical medicine ,Biology (General) ,Neural cell ,Tissue homeostasis ,chondroitin sulfate ,neuroregulation ,biology ,lecticans ,Perineuronal net ,Cell Biology ,Cell biology ,030104 developmental biology ,Proteoglycan ,biology.protein ,proteoglycans ,neural repair ,Neural development ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Chondroitin sulfate (CS) is the most abundant and widely distributed glycosaminoglycan (GAG) in the human body. As a component of proteoglycans (PGs) it has numerous roles in matrix stabilization and cellular regulation. This chapter highlights the roles of CS and CS-PGs in the central and peripheral nervous systems (CNS/PNS). CS has specific cell regulatory roles that control tissue function and homeostasis. The CNS/PNS contains a diverse range of CS-PGs which direct the development of embryonic neural axonal networks, and the responses of neural cell populations in mature tissues to traumatic injury. Following brain trauma and spinal cord injury, a stabilizing CS-PG-rich scar tissue is laid down at the defect site to protect neural tissues, which are amongst the softest tissues of the human body. Unfortunately, the CS concentrated in gliotic scars also inhibits neural outgrowth and functional recovery. CS has well known inhibitory properties over neural behavior, and animal models of CNS/PNS injury have demonstrated that selective degradation of CS using chondroitinase improves neuronal functional recovery. CS-PGs are present diffusely in the CNS but also form denser regions of extracellular matrix termed perineuronal nets which surround neurons. Hyaluronan is immobilized in hyalectan CS-PG aggregates in these perineural structures, which provide neural protection, synapse, and neural plasticity, and have roles in memory and cognitive learning. Despite the generally inhibitory cues delivered by CS-A and CS-C, some CS-PGs containing highly charged CS disaccharides (CS-D, CS-E) or dermatan sulfate (DS) disaccharides that promote neural outgrowth and functional recovery. CS/DS thus has varied cell regulatory properties and structural ECM supportive roles in the CNS/PNS depending on the glycoform present and its location in tissue niches and specific cellular contexts. Studies on the fruit fly, Drosophila melanogaster and the nematode Caenorhabditis elegans have provided insightful information on neural interconnectivity and the role of the ECM and its PGs in neural development and in tissue morphogenesis in a whole organism environment.
- Published
- 2021
125. Metabolism navigates neural cell fate in development, aging and neurodegeneration
- Author
-
Jessica Lagerwall, Sophie Eichhorner, Larissa Traxler, Jerome Mertens, Davide Stefanoni, and Angelo D'Alessandro
- Subjects
0301 basic medicine ,Aging ,Neurodegenerative Disorders ,Neuroscience (miscellaneous) ,Medicine (miscellaneous) ,Review ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Immunology and Microbiology (miscellaneous) ,Alzheimer Disease ,Pathology ,medicine ,RB1-214 ,Humans ,Epigenetics ,Neural cell ,Neurons ,Mechanism (biology) ,Stem Cells ,Neurodegeneration ,Brain ,Psychiatric disorder ,Human brain ,medicine.disease ,3. Good health ,Metabolic pathway ,030104 developmental biology ,medicine.anatomical_structure ,Brain aging ,Neural development ,Metabolic Disorders ,Cell Fate Control ,Metabolic state ,Medicine ,Energy Metabolism ,Neuroscience ,030217 neurology & neurosurgery - Abstract
An uninterrupted energy supply is critical for the optimal functioning of all our organs, and in this regard the human brain is particularly energy dependent. The study of energy metabolic pathways is a major focus within neuroscience research, which is supported by genetic defects in the oxidative phosphorylation mechanism often contributing towards neurodevelopmental disorders and changes in glucose metabolism presenting as a hallmark feature in age-dependent neurodegenerative disorders. However, as recent studies have illuminated roles of cellular metabolism that span far beyond mere energetics, it would be valuable to first comprehend the physiological involvement of metabolic pathways in neural cell fate and function, and to subsequently reconstruct their impact on diseases of the brain. In this Review, we first discuss recent evidence that implies metabolism as a master regulator of cell identity during neural development. Additionally, we examine the cell type-dependent metabolic states present in the adult brain. As metabolic states have been studied extensively as crucial regulators of malignant transformation in cancer, we reveal how knowledge gained from the field of cancer has aided our understanding in how metabolism likewise controls neural fate determination and stability by directly wiring into the cellular epigenetic landscape. We further summarize research pertaining to the interplay between metabolic alterations and neurodevelopmental and psychiatric disorders, and expose how an improved understanding of metabolic cell fate control might assist in the development of new concepts to combat age-dependent neurodegenerative diseases, particularly Alzheimer's disease., Summary: Distinct metabolic states regulate the identity of neural cells during development and adulthood. Metabolic alterations due to changes in nutrient availability and aging destabilize cell identity and contribute to neurological diseases.
- Published
- 2021
126. Fabrication of PCL nanofibrous scaffold with tuned porosity for neural cell culture
- Author
-
Fatemeh Zamani, Atiyeh Abbasi, and Mohammad Amani Tehran
- Subjects
Scaffold ,Fabrication ,Materials science ,chemistry.chemical_element ,chemistry.chemical_compound ,Tissue engineering ,chemistry ,Aluminium ,Polycaprolactone ,General Earth and Planetary Sciences ,Composite material ,Porosity ,Neural cell ,Layer (electronics) ,Original Research - Abstract
In tissue engineering, the structure of nanofibrous scaffolds and optimization of their properties play important role in the enhancement of cell growth and proliferation. Therefore, the basic idea of the current study is to find a proper method for tuning the extent of porosity of the scaffold, study the effect of porosity on the cell growth, and optimize the extent of porosity with the aim of achieving the maximum cell growth. To tune the scaffold’s porosity, four types of metal mesh with different mesh sizes were employed as collectors. For this purpose, the structural properties of polycaprolactone nanofibrous layers which were electrospun on collectors, and the level of neural A-172 cell growth on layers were investigated, and the results were compared with the results attained for the fabricated nanofibrous layer on a flat aluminum collector. It was found that upon changing the porosity of the metal mesh as collector, the fibers’ diameter would be inevitably changed, albeit insignificantly, and following no specific trends. However, changing the mesh size has shown a significant effect on the thickness and porosity of nanofibrous layer. According to the MTT assay results, the optimum neural cell growth was observed for the electrospun nanofibrous scaffold with the porosity of 96% and pore size of (0.42–23 µm) which has been fabricated on the type-4 collector having a mesh size of 10. The fabricated scaffold using this mesh with the optimum extent of porosity (58%) resulted in 44% enhancement in the cell growth as compared with the fabricated layer on the flat collector.
- Published
- 2021
127. Amphioxus neuroglia: Molecular characterization and evidence for early compartmentalization of the developing nerve cord
- Author
-
Emanuela Marcenaro, Valentina Obino, Thurston C. Lacalli, Federico Caicci, Tiziana Bachetti, Mario Pestarino, Lucia Manni, Matteo Bozzo, Michael Schubert, Simona Candiani, Laboratoire de Biologie du Développement de Villefranche sur mer (LBDV), Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut de la Mer de Villefranche (IMEV), and Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Neurogenesis ,Central nervous system ,astroglia ,cephalochordates ,evolution ,glial and neural progenitors ,radial glia ,Context (language use) ,Chordate ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,biology.animal ,Precursor cell ,medicine ,Animals ,Progenitor cell ,Neural cell ,ComputingMilieux_MISCELLANEOUS ,Lancelets ,biology ,Vertebrate ,biology.organism_classification ,Biological Evolution ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,nervous system ,Neurology ,Vertebrates ,Neuroglia ,030217 neurology & neurosurgery - Abstract
Glial cells play important roles in the development and homeostasis of metazoan nervous systems. However, while their involvement in the development and function in the central nervous system (CNS) of vertebrates is increasingly well understood, much less is known about invertebrate glia and the evolutionary history of glial cells more generally. An investigation into amphioxus glia is therefore timely, as this organism is the best living proxy for the last common ancestor of all chordates, and hence provides a window into the role of glial cell development and function at the transition of invertebrates and vertebrates. We report here our findings on amphioxus glia as characterized by molecular probes correlated with anatomical data at the transmission electron microscopy (TEM) level. The results show that amphioxus glial lineages express genes typical of vertebrate astroglia and radial glia, and that they segregate early in development, forming what appears to be a spatially separate cell proliferation zone positioned laterally, between the dorsal and ventral zones of neural cell proliferation. Our study provides strong evidence for the presence of vertebrate-type glial cells in amphioxus, while highlighting the role played by segregated progenitor cell pools in CNS development. There are implications also for our understanding of glial cells in a broader evolutionary context, and insights into patterns of precursor cell deployment in the chordate nerve cord.
- Published
- 2021
128. Neural stem/precursor cells dynamically change their epigenetic landscape to differentially respond to BMP signaling for fate switching during brain development
- Author
-
Kinichi Nakashima, Sayako Katada, Takuya Imamura, Shoko Sato, Takumi Nakagawa, Katsuhide Igarashi, Hitoshi Kurumizaka, Mizuki Honda, Jun Takouda, and Yasuyuki Ohkawa
- Subjects
biology ,SOXE Transcription Factors ,Neurogenesis ,Brain ,Cell Differentiation ,SMAD ,Bone morphogenetic protein ,Cell biology ,Epigenesis, Genetic ,Histone ,Neural Stem Cells ,Pregnancy ,DNA methylation ,Genetics ,biology.protein ,Humans ,Female ,Epigenetics ,Neural cell ,Transcription factor ,Developmental Biology ,Genome-Wide Association Study ,Research Paper - Abstract
During neocortical development, tight regulation of neurogenesis-to-astrogenesis switching of neural precursor cells (NPCs) is critical to generate a balanced number of each neural cell type for proper brain functions. Accumulating evidence indicates that a complex array of epigenetic modifications and the availability of extracellular factors control the timing of neuronal and astrocytic differentiation. However, our understanding of NPC fate regulation is still far from complete. Bone morphogenetic proteins (BMPs) are renowned as cytokines that induce astrogenesis of gliogenic late-gestational NPCs. They also promote neurogenesis of mid-gestational NPCs, although the underlying mechanisms remain elusive. By performing multiple genome-wide analyses, we demonstrate that Smads, transcription factors that act downstream from BMP signaling, target dramatically different genomic regions in neurogenic and gliogenic NPCs. We found that histone H3K27 trimethylation and DNA methylation around Smad-binding sites change rapidly as gestation proceeds, strongly associated with the alteration of accessibility of Smads to their target binding sites. Furthermore, we identified two lineage-specific Smad-interacting partners—Sox11 for neurogenic and Sox8 for astrocytic differentiation—that further ensure Smad-regulated fate-specific gene induction. Our findings illuminate an exquisite regulation of NPC property change mediated by the interplay between cell-extrinsic cues and -intrinsic epigenetic programs during cortical development.
- Published
- 2021
129. Heterogeneous ‘cell types’ can improve performance of deep neural networks
- Author
-
Alex T. Piet, Doty B, Anton Arkhipov, and Stefan Mihalas
- Subjects
Normalization (statistics) ,Contextual image classification ,Computer science ,business.industry ,Activation function ,Biological neural network ,Synthetic biological circuit ,Artificial intelligence ,Layer (object-oriented design) ,business ,Convolutional neural network ,Neural cell - Abstract
Deep convolutional neural networks (CNNs) are powerful computational tools for a large variety of tasks (Goodfellow, 2016). Their architecture, composed of layers of repeated identical neural units, draws inspiration from visual neuroscience. However, biological circuits contain a myriad of additional details and complexity not translated to CNNs, including diverse neural cell types (Tasic, 2018). Many possible roles for neural cell types have been proposed, including: learning, stabilizing excitation and inhibition, and diverse normalization (Marblestone, 2016; Gouwens, 2019). Here we investigate whether neural cell types, instantiated as diverse activation functions in CNNs, can assist in the feed-forward computational abilities of neural circuits. Our heterogeneous cell type networks mix multiple activation functions within each activation layer. We assess the value of mixed activation functions by comparing image classification performance to that of homogeneous control networks with only one activation function per network. We observe that mixing activation functions can improve the image classification abilities of CNNs. Importantly, we find larger improvements when the activation functions are more diverse, and in more constrained networks. Our results suggest a feed-forward computational role for diverse cell types in biological circuits. Additionally, our results open new avenues for the development of more powerful CNNs.
- Published
- 2021
130. LINC00665 rescues bupivacaine induced neurotoxicity in human neural cell of SH-SY5Y through has-miR-34a-5p
- Author
-
Yang Zhoujing, Sheng Hu, Ji Ling, and He Yinbin
- Subjects
SH-SY5Y ,Chemistry ,Cell Survival ,General Neuroscience ,Neurotoxicity ,RNA ,Apoptosis ,medicine.disease ,Bupivacaine ,In vitro ,Cell biology ,MicroRNAs ,Cell Line, Tumor ,medicine ,Humans ,RNA, Long Noncoding ,Viability assay ,Epigenetics ,Neural cell - Abstract
Background Excessive application of local anesthetics, bupivacaine (BUP) may induce neurotoxicity and lead to neurologic dysfunctions in human brains. Yet, the exact molecular mechanisms underlying BUP-induced neurotoxicity was not fully understood. In this study, we utilized an in vitro SH‐SY5Y cell culture model to explore the functional mechanism of long intergenic non-protein coding RNA 665 (LINC00665) in regulating BUP-induced neurotoxicity. Methods SH‐SY5Y cells were induced with BUP in vitro, and their viability and apoptosis were monitored. BUP-induced LINC00665 expression was also monitored, by qRT-PCR. LINC00665 was then overexpressed in SH‐SY5Y cells, and its effects on BUP-induced neurotoxicity were investigated. The downstream target transcript of LINC00665, human mature microRNA-34a-5p (hsa-miR-34a-5p) was investigated in BUP-induced SH‐SY5Y cells. Co-regulation of LINC00665 / hsa-miR-132–3p epigenetic axis was further examined on BUP-induced apoptosis in SH‐SY5Y cells. Results BUP reduced cell viability, induced apoptosis and downregulated LINC00665 in SH‐SY5Y cells. LINC00665 overexpression rescued BUP-induced neurotoxicity in SH‐SY5Y cells. Hsa-miR-34a-5p expression was directly correlated with BUP treatment and LINC00665 overexpression in SH‐SY5Y cells. Upregulating hsa-miR-34a-5p reversed the rescuing effects of LINC00665 on BUP-induced SH‐SY5Y apoptosis. Conclusions BUP-induced neurotoxicity in human neural cells may be regulated by the epigenetic axis of LINC00665 / hsa-miR-34a-5p.
- Published
- 2021
131. Nanophase surface arrays on poly (lactic-co-glycolic acid) upregulate neural cell functions
- Author
-
Didem Mimiroglu, Tulin Yanik, and Batur Ercan
- Subjects
Nanostructure ,Materials science ,Neurite ,Surface Properties ,technology, industry, and agriculture ,Metals and Alloys ,Biomedical Engineering ,Surface finish ,Matrix (biology) ,Glycolates ,Biomaterials ,PLGA ,chemistry.chemical_compound ,Glycols ,chemistry ,Chemical engineering ,Polylactic Acid-Polyglycolic Acid Copolymer ,Ceramics and Composites ,Lactic Acid ,Nanoscopic scale ,Neural cell ,Glycolic acid ,Polyglycolic Acid - Abstract
Nerve guidance channels (NGCs) promote cell-extracellular matrix (ECM) interactions occurring within the nanoscale. However, studies focusing on the effects of nanophase topography on neural cell functions are limited, and mostly concentrated on the sub-micron level (>100 nm) surface topography. Therefore, the aim of this study was to fabricate
- Published
- 2021
132. Non-linear Model-based Control of Neural Cell Dynamics
- Author
-
Tomastik R and Abhishek Dutta
- Subjects
Text mining ,nervous system ,Computer science ,business.industry ,Dynamics (mechanics) ,Non linear model ,Control (linguistics) ,business ,Biological system ,Neural cell - Abstract
This paper aims to discuss the control of a neuron’s firing. It is desired to control the neuron by injecting a current, whichallows the voltage of the neuron membrane to reach what is called the threshold voltage. When a neuron reaches this voltage,the potassium and sodium ion gates open allowing for the neuron to fire an action potential, observed as a voltage spike.Controlling this effect is helpful for those with certain diseases or disabilities. Four types of controllers are designed andsimulated on both the nonlinear system of the neuron and its linearized form, and all are found to meet specifications.
- Published
- 2021
133. Neural Transcription Correlates of Multimodal Cortical Phenotypes during Development
- Author
-
Marielle V. Fortier, Anqi Qiu, Michael J. Meaney, Peter D. Gluckman, Diliana Pecheva, Lynette Pei-Chi Shek, Annie Lee, Yap Seng Chong, and Joann S. Poh
- Subjects
Male ,Cognitive Neuroscience ,Synaptogenesis ,Biology ,Cellular and Molecular Neuroscience ,Child Development ,Gene expression ,medicine ,Humans ,Child ,Neural cell ,Cell Proliferation ,Cerebral Cortex ,Neurons ,Gene Expression Regulation, Developmental ,Infant ,Cell migration ,Human brain ,Brain Cortical Thickness ,Magnetic Resonance Imaging ,Phenotype ,medicine.anatomical_structure ,Astrocytes ,Child, Preschool ,Female ,Synaptic signaling ,Neuroglia ,Neuroscience ,Astrocyte - Abstract
During development, cellular events such as cell proliferation, migration, and synaptogenesis determine the structural organization of the brain. These processes are driven in part by spatiotemporally regulated gene expression. We investigated how the genetic signatures of specific neural cell types shape cortical organization of the human brain throughout infancy and childhood. Using a transcriptional atlas and in vivo magnetic resonance imaging (MRI) data, we demonstrated time-dependent associations between the expression levels of neuronal and glial genes and cortical macro- and microstructure. Neonatal cortical phenotypes were associated with prenatal glial but not neuronal gene expression. These associations reflect cell migration and proliferation during fetal development. Childhood cortical phenotypes were associated with neuronal and astrocyte gene expression related to synaptic signaling processes, reflecting the refinement of cortical connections. These findings indicate that sequential developmental stages contribute to distinct MRI measures at different time points. This helps to bridge the gap between the genetic mechanisms driving cellular changes and widely used neuroimaging techniques.
- Published
- 2019
134. Magnetic Control of Axon Navigation in Reprogrammed Neurons
- Author
-
Ann Na Cho, Jong Seung Lee, Taekyu Oh, Jinwoo Cheon, Eunna Chung, Jin Kim, Jung Uk Lee, Yoonhee Jin, Jae Hyun Lee, Seung Woo Cho, Kisuk Yang, and Ji Wook Kim
- Subjects
Deleted in Colorectal Cancer ,Induced Pluripotent Stem Cells ,Bioengineering ,02 engineering and technology ,Host tissue ,Antibodies ,Neurites ,medicine ,Humans ,General Materials Science ,Axon ,Magnetite Nanoparticles ,Receptor ,Neural cell ,Chemistry ,Mechanical Engineering ,General Chemistry ,Cellular Reprogramming ,DCC Receptor ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,Axons ,Transplantation ,Magnetic Fields ,medicine.anatomical_structure ,nervous system ,Axon guidance ,Receptor clustering ,0210 nano-technology ,Neuroscience - Abstract
While neural cell transplantation represents a promising therapy for neurodegenerative diseases, the formation of functional networks of transplanted cells with host neurons constitutes one of the challenging steps. Here, we introduce a magnetic guidance methodology that controls neurite growth signaling via magnetic nanoparticles (MNPs) conjugated with antibodies targeting the deleted in colorectal cancer (DCC) receptor (DCC-MNPs). Activation of the DCC receptors by clusterization and subsequent axonal growth of the induced neuronal (iN) cells was performed in a spatially controlled manner. In addition to the directionality of the magnetically controlled axon projection, axonal growth of the iN cells assisted the formation of functional connections with pre-existing primary neurons. Our results suggest magnetic guidance as a strategy for improving neuronal connectivity by spatially guiding the axonal projections of transplanted neural cells for synaptic interactions with the host tissue.
- Published
- 2019
135. Polyaniline-polycaprolactone blended nanofibers for neural cell culture
- Author
-
Pauline R. Hoffman, Fábio F.F. Garrudo, Caitlyn A. Chapman, Jorge Morgado, Carlos A V Rodrigues, Paiyz E. Mikael, Frederico Castelo Ferreira, João C. Silva, Robert J. Linhardt, Ranodhi N. Udangawa, and Joaquim M. S. Cabral
- Subjects
Conductive polymer ,Materials science ,Polymers and Plastics ,Biocompatibility ,Organic Chemistry ,General Physics and Astronomy ,02 engineering and technology ,010402 general chemistry ,021001 nanoscience & nanotechnology ,01 natural sciences ,0104 chemical sciences ,Neural tissue engineering ,chemistry.chemical_compound ,Tissue engineering ,chemistry ,Nanofiber ,Polycaprolactone ,Polyaniline ,Materials Chemistry ,0210 nano-technology ,Neural cell ,Biomedical engineering - Abstract
Neurodegenerative diseases compromise the quality of life of increasing numbers of the world’s aging population. While diagnosis is possible, no effective treatments are available. Using both tissue engineering and nanomedicine approaches, it is possible to develop systems appropriated for cell transplantation. Culturing neural stem cells (NSCs) on conductive polymers promotes their differentiation yield. The study herein aims at optimizing and characterizing NSC-compatible, electrically conductive poly(capro-e-lactone) (PCL)-polyaniline (PANI) electrospun scaffolds for neural tissue engineering applications. Furthermore, the optimal PANI to PCL ratio required for ideal electroconductivity properties is still not well understood. The obtained fibers were characterized by FTIR, TGA and DSC, and their material’s mechanical properties and electroconductivity, were investigated. For the first time, PCL-PANI fiber’s biocompatibility was assessed in NSCs; cell adhesion, growth rate and morphology were evaluated and correlated with the material’s physico-chemical properties. All the samples tested were able to support neural stem cell growth without any major changes on the cell’s typical morphology. We were also successfully able to produce electrically conductive nanofibers with conductivities above of biological fluids (7.7 × 10−2 S/cm vs 1.0 × 10−2 S/cm), making these ideal candidates for in vitro neural differentiation studies under electrical stimulation. Overall, this study provides valuable knowledge to improve future, in vitro models for drug testing and tissue engineering applications.
- Published
- 2019
136. Small molecules re-establish neural cell fate of human fibroblasts via autophagy activation
- Author
-
Phongsakorn Kunhorm, Nipha Chaicharoenaudomrung, Nudjanad Heebkaew, Wilasinee Promjantuek, Areechun Sotthibundhu, Narawadee Rujanapun, and Parinya Noisa
- Subjects
0301 basic medicine ,Cell type ,Models, Biological ,Small Molecule Libraries ,ATG12 ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,SOX2 ,Autophagy ,Humans ,Cell Lineage ,Neural cell ,Neurons ,Chemistry ,Wnt signaling pathway ,Cell Biology ,General Medicine ,Fibroblasts ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Stem cell ,Reactive Oxygen Species ,Neural development ,Developmental Biology - Abstract
The generation of neural cells is of great interest in medical research because of its promising in neurodegenerative diseases. Small chemical molecules have been used for inducing specific cell types across lineage boundaries. Therefore, to direct neural cell fate, small molecule is a feasible approach for generating clinically relevant cell types without genetic alterations. Human fibroblasts have been directly induced into neural cells with different combinations of small molecules; however, the mechanism underlying neural induction is still not fully understood. In this study, human fibroblasts were induced into neural cells by using only 4 small molecules in a short time period, 5 d. Small molecules used in this study included WNT activator, DNMT inhibitor, Notch inhibitor, and retinoic acid. Neural-specific genes, including NESTIN, TUJ1, and SOX2, were upregulated upon the induction for 5 d. Noteworthy, this neural induction process by small molecules coincided with the activation of autophagy. Autophagy-related genes, such as LC3, ATG12, and LAMP1, were enhanced upon neural induction, and the number of induced-neural cells decreased when autophagy was suppressed by chloroquine. The activation of autophagy was found to reduce ROS generation within the induced-neural cells, and the inhibition of autophagy by chloroquine suppressed the expression of antioxidant genes, CATALASE, SOD, and GPX. This implied that autophagy maintained the optimal level of ROS for neural induction of human fibroblasts. Altogether, this study presented the effective and convenient condition to induce neural cells from human fibroblasts and revealed the positive roles of autophagy in controlling neural cell induction.
- Published
- 2019
137. Less is more: Investigating the influence of cellular nanoparticle load on transfection outcomes in neural cells
- Author
-
Jacqueline A. Tickle and Divya M. Chari
- Subjects
Cell Survival ,Green Fluorescent Proteins ,0206 medical engineering ,Cell ,Population ,Biomedical Engineering ,Medicine (miscellaneous) ,Nanoparticle ,02 engineering and technology ,Gene delivery ,Transfection ,Biomaterials ,Magnetics ,03 medical and health sciences ,Plasmid ,Glial Fibrillary Acidic Protein ,medicine ,Animals ,Transgenes ,Magnetite Nanoparticles ,education ,Neural cell ,030304 developmental biology ,Neurons ,0303 health sciences ,education.field_of_study ,Chemistry ,R1 ,020601 biomedical engineering ,Cell biology ,Luminescent Proteins ,medicine.anatomical_structure ,Intracellular - Abstract
Genetic engineering of cell transplant populations offers potential for delivery of neurotherapeutic factors to modify the regenerative microenvironment of the injured spinal cord. The use of magnetic nanoparticle (MNP) based vectors has reduced the traditional reliance on viral methods and their associated obstacles in terms of scale up and safety. Studies utilising magnetic assistive platforms for MNP‐mediated gene delivery have found transfection efficiency in astrocytes (a major transplant and homeostatic neural cell type) to be both frequency and amplitude‐dependent. It is widely assumed that increased intracellular particle load will enhance transfection efficiency in a cell population. Therefore, we tested repeat delivery of MNP:plasmid complexes in conjunction with oscillating magnetic field parameters‐ a process termed ‘magneto‐multifection’‐ in astrocytes of primary origin in an attempt to enhance transfection levels. We show i) levels of transfection using magneto‐multifection equal that seen with viral methods; ii) reporter protein expression using two reporter plasmids shows a diverse profile of single/dual transfected cells with implications for delivery of a ‘cocktail’ of neurotherapeutic proteins and, iii) contrary to expectation, an inverse relationship exists between particle load and reporter protein expression.
- Published
- 2019
138. Attentive neural cell instance segmentation
- Author
-
Dimitris N. Metaxas, Jingru Yi, Menglin Jiang, Qiaoying Huang, Daniel J. Hoeppner, and Pengxiang Wu
- Subjects
Computer science ,Health Informatics ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Deep Learning ,0302 clinical medicine ,Minimum bounding box ,Distortion ,Image Processing, Computer-Assisted ,Code (cryptography) ,Radiology, Nuclear Medicine and imaging ,Segmentation ,Neural cell ,Microscopy ,Models, Statistical ,Radiological and Ultrasound Technology ,business.industry ,Detector ,Pattern recognition ,Computer Graphics and Computer-Aided Design ,Task (computing) ,Computer Vision and Pattern Recognition ,Artificial intelligence ,business ,Focus (optics) ,Neuroglia ,030217 neurology & neurosurgery - Abstract
Neural cell instance segmentation, which aims at joint detection and segmentation of every neural cell in a microscopic image, is essential to many neuroscience applications. The challenge of this task involves cell adhesion, cell distortion, unclear cell contours, low-contrast cell protrusion structures, and background impurities. Consequently, current instance segmentation methods generally fall short of precision. In this paper, we propose an attentive instance segmentation method that accurately predicts the bounding box of each cell as well as its segmentation mask simultaneously. In particular, our method builds on a joint network that combines a single shot multi-box detector (SSD) and a U-net. Furthermore, we employ the attention mechanism in both detection and segmentation modules to focus the model on the useful features. The proposed method is validated on a dataset of neural cell microscopic images. Experimental results demonstrate that our approach can accurately detect and segment neural cell instances at a fast speed, comparing favorably with the state-of-the-art methods. Our code is released on GitHub. The link is https://github.com/yijingru/ANCIS-Pytorch.
- Published
- 2019
139. Fine Manual Dexterity Assessment After Autologous Neural Cell Ecosystem (ANCE) Transplantation in a Non-human Primate Model of Parkinson’s Disease
- Author
-
Pauline Chatagny, Jean-François Brunet, Eric M. Rouiller, Jérôme Cottet, Michela Fregosi, Laura Carrara, Véronique Moret, Jocelyne Bloch, Simon Borgognon, and Simon Badoud
- Subjects
Parkinson's disease ,Cell Transplantation ,Pilot Projects ,Transplantation, Autologous ,Motor symptoms ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Rating scale ,Animals ,Medicine ,Neural cell ,030304 developmental biology ,0303 health sciences ,Non human primate ,Behavior, Animal ,business.industry ,MPTP ,MPTP Poisoning ,Recovery of Function ,General Medicine ,medicine.disease ,Neostriatum ,Transplantation ,Disease Models, Animal ,Macaca fascicularis ,chemistry ,Motor Skills ,Female ,Motor recovery ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Background. Autologous neural cell ecosystem (ANCE) transplantation improves motor recovery in MPTP monkeys. These motor symptoms were assessed using semi-quantitative clinical rating scales, widely used in many studies. However, limitations in terms of sensitivity, combined with relatively subjective assessment of their different items, make inter-study comparisons difficult to achieve. Objective. The aim of this study was to quantify the impact of MPTP intoxication in macaque monkeys on manual dexterity and assess whether ANCE can contribute to functional recovery. Methods. Four animals were trained to perform 2 manual dexterity tasks. After reaching a motor performance plateau, the animals were subjected to an MPTP lesion. After the occurrence of a spontaneous functional recovery plateau, all 4 animals were subjected to ANCE transplantation. Results. Two of 4 animals underwent a full spontaneous recovery before the ANCE transplantation, whereas the 2 other animals (symptomatic) presented moderate to severe Parkinson’s disease (PD)-like symptoms affecting manual dexterity. The time to grasp small objects using the precision grip increased in these 2 animals. After ANCE transplantation, the 2 symptomatic animals underwent a significant functional recovery, reflected by a decrease in time to execute the different tasks, as compared with the post-lesion phase. Conclusions. Manual dexterity is affected in symptomatic MPTP monkeys. The 2 manual dexterity tasks reported here as pilot are pertinent to quantify PD symptoms and reliably assess a treatment in MPTP monkeys, such as the present ANCE transplantation, to be confirmed in a larger cohort of animals before future clinical applications.
- Published
- 2019
140. Use of human pluripotent stem cell-derived cells for neurodegenerative disease modeling and drug screening platform
- Author
-
Javier Vitorica, Antonia Gutierrez, Juan Antonio García-León, and Universidad de Sevilla. Departamento de Bioquímica y Biología Molecular
- Subjects
Pluripotent Stem Cells ,Drug ,amyotrophic lateral sclerosis ,Parkinson's disease ,Neurogenesis ,media_common.quotation_subject ,Drug Evaluation, Preclinical ,Computational biology ,Disease ,multiple sclerosis ,01 natural sciences ,03 medical and health sciences ,neurodegenerative disease ,Huntington's disease ,disease modeling ,Drug Discovery ,human-induced pluripotent stem cells ,medicine ,Animals ,Humans ,CRISPR ,drug screening ,Induced pluripotent stem cell ,Neural cell ,030304 developmental biology ,media_common ,Gene Editing ,Neurons ,Pharmacology ,hiPSCs ,0303 health sciences ,hiP ,business.industry ,Disease progression ,Neurodegenerative Diseases ,medicine.disease ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Parkinson’s disease ,Molecular Medicine ,CRISPR-Cas9 ,CRISPR-Cas Systems ,business ,Alzheimer’s disease ,Huntington’s disease - Abstract
Most neurodegenerative diseases are characterized by a complex and mostly still unresolved pathology. This fact, together with the lack of reliable disease models, has precluded the development of effective therapies counteracting the disease progression. The advent of human pluripotent stem cells has revolutionized the field allowing the generation of disease-relevant neural cell types that can be used for disease modeling, drug screening and, possibly, cell transplantation purposes. In this Review, we discuss the applications of human pluripotent stem cells, the development of efficient protocols for the derivation of the different neural cells and their applicability for robust in vitro disease modeling and drug screening platforms for most common neurodegenerative conditions.
- Published
- 2019
141. Inhibition of microRNA-375 ameliorated ketamine-induced neurotoxicity in human embryonic stem cell derived neurons
- Author
-
Fang Wang, Xiaoqiang Wang, Lu Li, Fang Shu, Haigang Lv, Xin Zhao, and Li Wu
- Subjects
0301 basic medicine ,Neurite ,Human Embryonic Stem Cells ,Biology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Neurotrophic factors ,medicine ,Humans ,Viability assay ,RNA, Small Interfering ,Neural cell ,Anesthetics ,Neurons ,Pharmacology ,Brain-Derived Neurotrophic Factor ,Neurotoxicity ,Cell Differentiation ,medicine.disease ,Embryonic stem cell ,Cell biology ,MicroRNAs ,030104 developmental biology ,Apoptosis ,embryonic structures ,Ketamine ,Neurotoxicity Syndromes ,Reactive Oxygen Species ,030217 neurology & neurosurgery - Abstract
Excessive exposure to commonly used anesthetic agents, such as ketamine, may induce permanent damage to immature human brains. In this work, we used a human embryonic stem cell (hESC)-derived neuron model to assess the expression and function of human microRNA 735 (hsa-miR-375) in regulating ketamine-induced neural cell death and neural toxicity in vitro. In the in vitro culture, hESC-derived neurons were incubated with ketamine for 72 h. After that, cell viability, reactive oxygen species activity, neural apoptosis, neurite degeneration, and hsa-miR-375 gene expression were assessed, respectively. We found ketamine induced neural death, reactive oxygen species augmentation, neural apoptosis, neurite degeneration and hsa-miR-375 upregulation, in hESC-derived neurons. In addition, we discovered that, lentivirus-mediated mR-375 downregulation protected ketamine-induced neural cell death and neural toxicity. Also, human brain derived neurotrophic factor (BDNF) was found to be directly and reversely regulated by hsa-miR-375. Moreover, BDNF downregulation was shown to functionally reverse the protective effect of miR-375 downregulation on ketamine-induced neural cell death and neural toxicity. Overall, this work provided strong evidence showing hsa-miR-375 is an active regulator in anesthesia-induced neural cell death and neural toxicity, possibly via inverse regulation on BDNF gene.
- Published
- 2019
142. Biodegradable Neural Cell Culture Sheet Made of Polyanhydride thin Film with Micro-Trench Structures
- Author
-
Satomitsu Imai and Yuki Nakamura
- Subjects
Materials science ,Trench ,Electrical and Electronic Engineering ,Thin film ,Composite material ,Neural cell ,Electronic, Optical and Magnetic Materials - Published
- 2019
143. Improved cellular response on functionalized polypyrrole interfaces
- Author
-
Elnaz Elahirad, Masoud Mozafari, Mahsa Khalili, Tarlan Eslami Arshaghi, Akbar Karkhaneh, Masumeh Dodel, Fathollah Moztarzadeh, and Sanaz Naghavi Alhosseini
- Subjects
0301 basic medicine ,food.ingredient ,Physiology ,Clinical Biochemistry ,technology, industry, and agriculture ,Cell Biology ,Polypyrrole ,Gelatin ,Electrospinning ,Neural tissue engineering ,Chitosan ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,food ,chemistry ,Tissue engineering ,030220 oncology & carcinogenesis ,Biophysics ,Surface modification ,Neural cell - Abstract
Neuroregeneration strategies involve multiple factors to stimulate nerve regeneration. Neural support with chemical and physical cues to optimize neural growth and replacing the lesion neuron and axons are crucial for designing neural scaffolds, which is a promising treatment approach. In this study, polypyrrole polymerization and its functionalization at the interface developed by glycine and gelatin for further optimization of cellular response. Nanofibrous scaffolds were fabricated by electrospinning of polyvinyl alcohol and chitosan solutions. The electrospun scaffolds were polymerized on the surface by pyrrole monomers to form an electroactive interface for further applications in neural tissue engineering. The polymerized polypyrrole showed a positive zeta potential value of 57.5 ± 5.46 mV. The in vitro and in vivo biocompatibility of the glycine and gelatin-functionalized polypyrrole-coated scaffolds were evaluated. No inflammatory cells were observed for the implanted scaffolds. Further, DAPI nucleus staining showed a superior cell attachment on the gelatin-functionalized polypyrrole-coated scaffolds. The topography and tuned positively charged polypyrrole interface with gelatin functionalization is expected to be particularly efficient physical and chemical simultaneous factors for promoting neural cell adhesion.
- Published
- 2019
144. The Antimicrobial Peptide CopA3 Inhibits Clostridium difficile Toxin A-Induced Viability Loss and Apoptosis in Neural Cells
- Author
-
Joon Ha Lee, Ho Kim, I Na Yoon, and Jae Sam Hwang
- Subjects
Chemistry ,Toxin ,Clostridium difficile toxin A ,Inflammation ,General Medicine ,Clostridium difficile ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Cell biology ,Neurotransmitter secretion ,Downregulation and upregulation ,Apoptosis ,medicine ,medicine.symptom ,Neural cell ,Biotechnology - Abstract
Numerous studies have reported that enteric neurons involved in controlling neurotransmitter secretion and motility in the gut critically contribute to the progression of gut inflammation. Clostridium difficile toxins, which cause severe colonic inflammation, are also known to affect enteric neurons. Our previous study showed that C. difficile toxin A directly induces neural cell toxicities, such as viability loss and apoptosis. In the current study, we attempted to identify a potent inhibitor of toxin A-induced neural cell toxicity that may aid in managing toxin A-induced gut inflammation. In our recent study, we found that the Korea dung beetle-derived antimicrobial peptide CopA3 completely blocked neural cell apoptosis caused by okadaic acid or 6-OHDA. Here, we examined whether the antimicrobial peptide CopA3 inhibited toxin A-induced neural cell damage. In neuroblastoma SH-SY5Y cells, CopA3 treatment protected against both apoptosis and viability loss caused by toxin A. CopA3 also completely inhibited activation of the pro-apoptotic factor, caspase-3. Additionally, CopA3 rescued toxin A-induced downregulation of neural cell proliferation. However, CopA3 had no effect on signaling through ROS/p38 MAPK/p27kip1, suggesting that CopA3 inhibits toxin A-induced neural cell toxicity independent of this well-characterized toxin A pathway. Our data further suggest that ability of CopA3 to rescue toxin A-induced neural cell damage may also ameliorate the gut inflammation caused by toxin A.
- Published
- 2019
145. Controlled cellular automata
- Author
-
Achilles A. Beros, Monique Chyba, and Oleksandr Markovichenko
- Subjects
Statistics and Probability ,Brain development ,Computer science ,Applied Mathematics ,General Engineering ,Complex system ,Morphogenesis ,01 natural sciences ,Cellular automaton ,Computer Science Applications ,010101 applied mathematics ,Control system ,Tumor growth ,0101 mathematics ,Control (linguistics) ,Biological system ,Neural cell - Abstract
Cellular Automata have been successfully used to model evolution of complex systems based on simples rules. In this paper we introduce controlled cellular automata to depict the dynamics of systems with controls that can affect their evolution. Using theory from discrete control systems, we derive results for the control of cellular automata in specific cases. The paper is mostly oriented toward two applications: fire spreading; morphogenesis and tumor growth. In both cases, we illustrate the impact of a control on the evolution of the system. For the fire, the control is assumed to be either firelines or firebreaks to prevent spreading or dumping of water, fire retardant and chemicals (foam) on the fire to neutralize it. In the case of cellular growth, the control describes mechanisms used to regulate growth factors and morphogenic events based on the existence of extracellular matrix structures called fractones. The hypothesis is that fractone distribution may coordinate the timing and location of neural cell proliferation, thereby guiding morphogenesis, at several stages of early brain development.
- Published
- 2019
146. Zinc finger E-box–binding homeobox 1 (ZEB1) is required for neural differentiation of human embryonic stem cells
- Author
-
Mingxia Du, Hongmei Wang, Longkuo Xia, Xiaoyin Lu, Yuan Jiang, Da Zhang, Long Yan, Baoyang Hu, Dewen Ding, and Wenliang Zhu
- Subjects
0301 basic medicine ,Zinc finger ,Cell Biology ,Biology ,Biochemistry ,Embryonic stem cell ,Regenerative medicine ,Transplantation ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,Neuron ,Stem cell ,Induced pluripotent stem cell ,Molecular Biology ,Neural cell ,Neuroscience - Abstract
Human pluripotent stem cells hold great promise for improving regenerative medicine. However, a risk for tumor formation and difficulties in generating large amounts of subtype derivatives remain the major obstacles for clinical applications of stem cells. Here, we discovered that zinc finger E-box–binding homeobox 1 (ZEB1) is highly expressed upon differentiation of human embryonic stem cells (hESCs) into neuronal precursors. CRISPR/Cas9-mediated ZEB1 depletion did not impede neural fate commitment, but prevented hESC-derived neural precursors from differentiating into neurons, indicating that ZEB1 is required for neuronal differentiation. ZEB1 overexpression not only expedited neural differentiation and neuronal maturation, which ensured safer neural cell transplantation, but also facilitated the generation of excitatory cortical neurons, which were valuable for managing certain neurological disorders, such as Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS). Our study provides useful information on how human neural cells are generated, which may help in forming strategies for developing and improving replacement therapies for treating patients with neurological diseases.
- Published
- 2018
147. Visualizing the Role of Lipid Dynamics during Infrared Neural Stimulation with Hyperspectral Stimulated Raman Scattering Microscopy
- Author
-
Craig L. Duvall, E. Duco Jansen, Anita Mahadevan-Jansen, Graham A. Throckmorton, Rekha Gautam, Bryan R. Dollinger, Wilson R. Adams, Andrea K. Locke, and Ana I. Borrachero-Conejo
- Subjects
Chemistry ,Dynamics (mechanics) ,Cell ,Membrane structure ,Stimulation ,symbols.namesake ,medicine.anatomical_structure ,Microscopy ,medicine ,symbols ,Biophysics ,Lipid bilayer ,Neural cell ,Raman scattering - Abstract
Infrared neural stimulation, or INS, is a method of using pulsed infrared light to yield label-free neural stimulation with broad experimental and translational utility. Despite its robust demonstration, the mechanistic and biophysical underpinnings of INS have been the subject of debate for more than a decade. The role of lipid membrane thermodynamics appears to play an important role in how fast IR-mediated heating nonspecifically drives action potential generation. Direct observation of lipid membrane dynamics during INS remains to be shown in a live neural model system. To directly test the involvement of lipid dynamics in INS, we used hyperspectral stimulated Raman scattering (hsSRS) microscopy to study biochemical signatures of high-speed vibrational dynamics underlying INS in a live neural cell culture model. Findings suggest that lipid bilayer structural changes are occurring during INS in vitro in NG108-15 neuroglioma cells. Lipid-specific signatures of cell SRS spectra were found to vary with stimulation energy and radiant exposure. Spectroscopic observations were verified against high-speed ratiometric fluorescence imaging of a conventional lipophilic membrane structure reporter, di-4-ANNEPS. Overall, the presented data supports the hypothesis that INS causes changes in the lipid membrane of neural cells by changing lipid membrane packing order – which coincides with likelihood of cell stimulation. Furthermore, this work highlights the potential of hsSRS as a method to study biophysical and biochemical dynamics safely in live cells.
- Published
- 2021
148. Analysis of Translation in the Developing Mouse Brain using Polysome Profiling
- Author
-
Shreeya Kedia, Sarah L Erickson, and Guang Yang
- Subjects
Messenger RNA ,Translational efficiency ,General Immunology and Microbiology ,General Chemical Engineering ,General Neuroscience ,Brain ,Translation (biology) ,Biology ,Neural stem cell ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Mice ,Neural Stem Cells ,Polyribosomes ,Protein Biosynthesis ,Polysome ,Translational regulation ,Gene expression ,Animals ,RNA, Messenger ,Neural cell - Abstract
The proper development of the mammalian brain relies on a fine balance of neural stem cell proliferation and differentiation into different neural cell types. This balance is tightly controlled by gene expression that is fine-tuned at multiple levels, including transcription, post-transcription and translation. In this regard, a growing body of evidence highlights a critical role of translational regulation in coordinating neural stem cell fate decisions. Polysome fractionation is a powerful tool for the assessment of mRNA translational status at both global and individual gene levels. Here, we present an in-house polysome profiling pipeline to assess translational efficiency in cells from the developing mouse cerebral cortex. We describe the protocols for sucrose gradient preparation, tissue lysis, ultracentrifugation and fractionation-based analysis of mRNA translational status.
- Published
- 2021
149. Differentiation of cortical brain organoids and optic nerve-like structures from retinal confluent cultures of pluripotent stem cells
- Author
-
Robin R. Ali, Erdahl Teber, Pengyi Yang, Grady C. Smith, Mark E. Graham, Di Xiao, Scott Lee, Anai Gonzalez-Cordero, Jesse R. Wark, Milan Fernando, Ted Wong, and Hani Jieun Kim
- Subjects
Retina ,Retinal ,Biology ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Cortex (anatomy) ,Organoid ,medicine ,Optic nerve ,Stem cell ,Induced pluripotent stem cell ,Neuroscience ,Neural cell - Abstract
Advances in the study of neurological conditions have been possible due to induced pluripotent stem cell technologies and the generation of neural cell types and organoids. Numerous studies have described the generation of neural ectoderm-derived retinal and brain structures from pluripotent stem cells. However, the field is still troubled by technical challenges, including high culture costs and organoid-to-organoid variability. Here, we describe a simple and economical protocol that reproducibly gives rise to the neural retina and cortical brain regions from confluent cultures of stem cells. The spontaneously generated cortical organoids were isolated and cultured in suspension conditions for maturation and are transcriptionally comparable to organoids generated by other methods and to human foetal cortex. Furthermore, these organoids show spontaneous functional network activity with proteomic analysis and electron microscopy demonstrating the presence of synaptic components and maturity. The generation of retinal and brain organoids in close proximity also enabled their mutual isolation. Further culture of this complex organoid system demonstrated the formation of optic nerve-like structures connecting retinal and brain organoids, which might facilitate the investigation of the mechanisms of neurological diseases of the eye and brain.
- Published
- 2021
150. Acorde: unraveling functionally-interpretable networks of isoform co-usage from single cell data
- Author
-
Ángeles Arzalluz-Luque, Ana Conesa, Pedro Salguero, and Sonia Tarazona
- Subjects
Gene isoform ,medicine.anatomical_structure ,Mechanism (biology) ,Computer science ,Cell ,Alternative splicing ,medicine ,Computational biology ,Gene ,Neural cell ,Function (biology) - Abstract
Alternative splicing (AS) is a highly-regulated post-transcriptional mechanism known to modulate isoform expression within genes and contribute to cell-type identity. However, the extent to which alternative isoforms establish co-expression networks that may relevant in cellular function has not been explored yet. Here, we present acorde, a pipeline that successfully leverages bulk long reads and single-cell data to confidently detect alternative isoform co-expression relationships. To achieve this, we developed and validated percentile correlations, a novel approach that overcomes data sparsity and yields accurate co-expression estimates from single-cell data. Next, acorde uses correlations to cluster co-expressed isoforms into a network, unraveling cell type-specific alternative isoform usage patterns. By selecting same-gene isoforms between these clusters, we subsequently detect and characterize genes with co-differential isoform usage (coDIU) across neural cell types. Finally, we predict functional elements from long read-defined isoforms and provide insight into biological processes, motifs and domains potentially controlled by the coordination of post-transcriptional regulation.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.