15 results on '"Martin W. Nicholson"'
Search Results
2. GABAA receptors and neuroligin 2 synergize to promote synaptic adhesion and inhibitory synaptogenesis
- Author
-
Yusheng Sui, Martin Mortensen, Banghao Yuan, Martin W. Nicholson, Trevor G. Smart, and Jasmina N. Jovanovic
- Subjects
inhibition ,GABAergic synapse ,synaptic ,extrasynaptic ,stable cell lines ,medium spiny neurons ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
GABAA receptors (γ-aminobutyric acid-gated receptors type A; GABAARs), the major structural and functional postsynaptic components of inhibitory synapses in the mammalian brain, belong to a family of GABA-gated Cl−/HCO3− ion channels. They are assembled as heteropentamers from a family of subunits including: α (1–6), β(1–3), γ(1–3), δ, ε, π, θ and ρ(1–3). GABAARs together with the postsynaptic adhesion protein Neuroligin 2 (NL2) and many other pre- and post-synaptic proteins guide the initiation and functional maturation of inhibitory GABAergic synapses. This study examined how GABAARs and NL2 interact with each other to initiate the formation of synapses. Two functionally distinct GABAAR subtypes, the synaptic type α2β2γ2-GABAARs versus extrasynaptic type α4β3δ-GABAARs were expressed in HEK293 cells alone or together with NL2 and co-cultured with striatal GABAergic medium spiny neurons to enable innervation of HEK293 cells by GABAergic axons. When expressed alone, only the synaptic α2β2γ2-GABAARs induced innervation of HEK293 cells. However, when GABAARs were co-expressed with NL2, the effect on synapse formation exceeded the individual effects of these proteins indicating a synergistic interaction, with α2β2γ2-GABAAR/NL2 showing a significantly greater synaptogenic activity than α4β3δ-GABAAR/NL2 or NL2 alone. To investigate the molecular basis of this interaction, different combinations of GABAAR subunits and NL2 were co-expressed, and the degree of innervation and synaptic activity assessed, revealing a key role of the γ2 subunit. In biochemical assays, the interaction between NL2 and α2β2γ2-GABAAR was established and mapped to the large intracellular domain of the γ2 subunit.
- Published
- 2024
- Full Text
- View/download PDF
3. Cardiac-specific microRNA-125b deficiency induces perinatal death and cardiac hypertrophy
- Author
-
Chen-Yun Chen, Desy S. Lee, Oi Kuan Choong, Sheng-Kai Chang, Tien Hsu, Martin W. Nicholson, Li-Wei Liu, Po-Ju Lin, Shu-Chian Ruan, Shu-Wha Lin, Chung-Yi Hu, and Patrick C. H. Hsieh
- Subjects
Medicine ,Science - Abstract
Abstract MicroRNA-125b, the first microRNA to be identified, is known to promote cardiomyocyte maturation from embryonic stem cells; however, its physiological role remains unclear. To investigate the role of miR-125b in cardiovascular biology, cardiac-specific miR-125b-1 knockout mice were generated. We found that cardiac-specific miR-125b-1 knockout mice displayed half the miR-125b expression of control mice resulting in a 60% perinatal death rate. However, the surviving mice developed hearts with cardiac hypertrophy. The cardiomyocytes in both neonatal and adult mice displayed abnormal mitochondrial morphology. In the deficient neonatal hearts, there was an increase in mitochondrial DNA, but total ATP production was reduced. In addition, both the respiratory complex proteins in mitochondria and mitochondrial transcription machinery were impaired. Mechanistically, using transcriptome and proteome analysis, we found that many proteins involved in fatty acid metabolism were significantly downregulated in miR-125b knockout mice which resulted in reduced fatty acid metabolism. Importantly, many of these proteins are expressed in the mitochondria. We conclude that miR-125b deficiency causes a high mortality rate in neonates and cardiac hypertrophy in adult mice. The dysregulation of fatty acid metabolism may be responsible for the cardiac defect in the miR-125b deficient mice.
- Published
- 2021
- Full Text
- View/download PDF
4. Copy number variant hotspots in Han Taiwanese population induced pluripotent stem cell lines - lessons from establishing the Taiwan human disease iPSC Consortium Bank
- Author
-
Ching-Ying Huang, Ling-Hui Li, Wan-Tseng Hsu, Yu-Che Cheng, Martin W. Nicholson, Chun-Lin Liu, Chien-Yu Ting, Hui-Wen Ko, Shih-Han Syu, Cheng-Hao Wen, Zhuge Yan, Hsiang-Po Huang, Hong-Lin Su, Po-Min Chiang, Chia-Ning Shen, Hsin-Fu Chen, B. Lin Ju Yen, Huai-En Lu, Shiaw-Min Hwang, Shih-Hwa Chiou, Hong-Nerng Ho, Jer-Yuarn Wu, Timothy J. Kamp, Joseph C. Wu, and Patrick C. H. Hsieh
- Subjects
Human induced pluripotent stem cell ,Cell differentiation ,Stem cell bank ,Drug screening ,Copy number variant ,Hotspot ,Medicine - Abstract
Abstract Background The Taiwan Human Disease iPSC Service Consortium was established to accelerate Taiwan’s growing stem cell research initiatives and provide a platform for researchers interested in utilizing induced pluripotent stem cell (iPSC) technology. The consortium has generated and characterized 83 iPSC lines: 11 normal and 72 disease iPSC lines covering 21 different diseases, several of which are of high incidence in Taiwan. Whether there are any reprogramming-induced recurrent copy number variant (CNV) hotspots in iPSCs is still largely unknown. Methods We performed genome-wide copy number variant screening of 83 Han Taiwanese iPSC lines and compared them with 1093 control subjects using an Affymetrix genome-wide human SNP array. Results In the iPSCs, we identified ten specific CNV loci and seven “polymorphic” CNV regions that are associated with the reprogramming process. Additionally, we established several differentiation protocols for our iPSC lines. We demonstrated that our iPSC-derived cardiomyocytes respond to pharmacological agents and were successfully engrafted into the mouse myocardium demonstrating their potential application in cell therapy. Conclusions The CNV hotspots induced by cell reprogramming have successfully been identified in the current study. This finding may be used as a reference index for evaluating iPSC quality for future clinical applications. Our aim was to establish a national iPSC resource center generating iPSCs, made available to researchers, to benefit the stem cell community in Taiwan and throughout the world.
- Published
- 2020
- Full Text
- View/download PDF
5. Population-based high-throughput toxicity screen of human iPSC-derived cardiomyocytes and neurons
- Author
-
Ching Ying Huang, Martin W. Nicholson, Jyun Yuan Wang, Chien Yu Ting, Ming Heng Tsai, Yu Che Cheng, Chun Lin Liu, Darien Z.H. Chan, Yi Chan Lee, Ching Chuan Hsu, Yu Hung Hsu, Chiou Fong Yang, Cindy M.C. Chang, Shu Chian Ruan, Po Ju Lin, Jen Hao Lin, Li Lun Chen, Marvin L. Hsieh, Yuan Yuan Cheng, Wan Tseng Hsu, Yi Ling Lin, Chien Hsiun Chen, Yu Hsiang Hsu, Ying Ta Wu, Timothy A. Hacker, Joseph C. Wu, Timothy J. Kamp, and Patrick C.H. Hsieh
- Subjects
CP: Stem cell research ,Biology (General) ,QH301-705.5 - Abstract
Summary: In this study, we establish a population-based human induced pluripotent stem cell (hiPSC) drug screening platform for toxicity assessment. After recruiting 1,000 healthy donors and screening for high-frequency human leukocyte antigen (HLA) haplotypes, we identify 13 HLA-homozygous “super donors” to represent the population. These “super donors” are also expected to represent at least 477,611,135 of the global population. By differentiating these representative hiPSCs into cardiomyocytes and neurons we show their utility in a high-throughput toxicity screen. To validate hit compounds, we demonstrate dose-dependent toxicity of the hit compounds and assess functional modulation. We also show reproducible in vivo drug toxicity results using mouse models with select hit compounds. This study shows the feasibility of using a population-based hiPSC drug screening platform to assess cytotoxicity, which can be used as an innovative tool to study inter-population differences in drug toxicity and adverse drug reactions in drug discovery applications.
- Published
- 2022
- Full Text
- View/download PDF
6. Human iPSC banking: barriers and opportunities
- Author
-
Ching-Ying Huang, Chun-Lin Liu, Chien-Yu Ting, Yueh-Ting Chiu, Yu-Che Cheng, Martin W. Nicholson, and Patrick C. H. Hsieh
- Subjects
Induced pluripotent stem cell (iPSC) ,Cell bank ,Personalized medicine ,Medicine - Abstract
Abstract The introduction of induced pluripotent stem cells (iPSCs) has opened up the potential for personalized cell therapies and ushered in new opportunities for regenerative medicine, disease modeling, iPSC-based drug discovery and toxicity assessment. Over the past 10 years, several initiatives have been established that aim to collect and generate a large amount of human iPSCs for scientific research purposes. In this review, we compare the construction and operation strategy of some iPSC banks as well as their ongoing development. We also introduce the technical challenges and offer future perspectives pertaining to the establishment and management of iPSC banks.
- Published
- 2019
- Full Text
- View/download PDF
7. Utility of iPSC-Derived Cells for Disease Modeling, Drug Development, and Cell Therapy
- Author
-
Martin W. Nicholson, Chien-Yu Ting, Darien Z. H. Chan, Yu-Che Cheng, Yi-Chan Lee, Ching-Chuan Hsu, Ching-Ying Huang, and Patrick C. H. Hsieh
- Subjects
induced pluripotent stem cells ,cell therapy ,cardiomyocytes ,neurons ,Cytology ,QH573-671 - Abstract
The advent of induced pluripotent stem cells (iPSCs) has advanced our understanding of the molecular mechanisms of human disease, drug discovery, and regenerative medicine. As such, the use of iPSCs in drug development and validation has shown a sharp increase in the past 15 years. Furthermore, many labs have been successful in reproducing many disease phenotypes, often difficult or impossible to capture, in commonly used cell lines or animal models. However, there still remain limitations such as the variability between iPSC lines as well as their maturity. Here, we aim to discuss the strategies in generating iPSC-derived cardiomyocytes and neurons for use in disease modeling, drug development and their use in cell therapy.
- Published
- 2022
- Full Text
- View/download PDF
8. Cardio- and Neurotoxicity of Selected Anti-COVID-19 Drugs
- Author
-
Martin W. Nicholson, Ching-Ying Huang, Jyun-Yuan Wang, Chien-Yu Ting, Yu-Che Cheng, Darien Z. H. Chan, Yi-Chan Lee, Ching-Chuan Hsu, Yu-Hung Hsu, Cindy M. C. Chang, Marvin L. Hsieh, Yuan-Yuan Cheng, Yi-Ling Lin, Chien-Hsiun Chen, Ying-Ta Wu, Timothy A. Hacker, Joseph C. Wu, Timothy J. Kamp, and Patrick C. H. Hsieh
- Subjects
stem cell research ,cardiomyocyte ,neuron ,drug screening ,human leukocyte antigen ,human-induced pluripotent stem cell ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Since December 2019, the novel coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has infected ~435 million people and caused ~6 million related deaths as of March 2022. To combat COVID-19, there have been many attempts to repurpose FDA-approved drugs or revive old drugs. However, many of the current treatment options have been known to cause adverse drug reactions. We employed a population-based drug screening platform using 13 human leukocyte antigen (HLA) homozygous human induced pluripotent cell (iPSC) lines to assess the cardiotoxicity and neurotoxicity of the first line of anti-COVID-19 drugs. We also infected iPSC-derived cells to understand the viral infection of cardiomyocytes and neurons. We found that iPSC-derived cardiomyocytes express the ACE2 receptor which correlated with a higher infection of the SARS-CoV-2 virus (r = 0.86). However, we were unable to detect ACE2 expression in neurons which correlated with a low infection rate. We then assessed the toxicity of anti-COVID-19 drugs and identified two cardiotoxic compounds (remdesivir and arbidol) and four neurotoxic compounds (arbidol, remdesivir, hydroxychloroquine, and chloroquine). These data show that this platform can quickly and easily be employed to further our understanding of cell-specific infection and identify drug toxicity of potential treatment options helping clinicians better decide on treatment options.
- Published
- 2022
- Full Text
- View/download PDF
9. Selective Modulation of α5 GABAA Receptors Exacerbates Aberrant Inhibition at Key Hippocampal Neuronal Circuits in APP Mouse Model of Alzheimer’s Disease
- Author
-
Alexandra L. Petrache, Archie A. Khan, Martin W. Nicholson, Alessandra Monaco, Martyna Kuta-Siejkowska, Shozeb Haider, Stephen Hilton, Jasmina N. Jovanovic, and Afia B. Ali
- Subjects
Alzheimer’s disease ,GABAA receptors ,synaptic ,interneurons ,hippocampus ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Selective negative allosteric modulators (NAMs), targeting α5 subunit-containing GABAA receptors (GABAARs) as potential therapeutic targets for disorders associated with cognitive deficits, including Alzheimer’s disease (AD), continually fail clinical trials. We investigated whether this was due to the change in the expression of α5 GABAARs, consequently altering synaptic function during AD pathogenesis. Using medicinal chemistry and computational modeling, we developed aqueous soluble hybrids of 6,6-dimethyl-3-(2-hydroxyethyl) thio-1-(thiazol-2-yl)-6,7-dihydro-2-benzothiophene-4(5H)-one, that demonstrated selective binding and high negative allosteric modulation, specifically for the α5 GABAAR subtypes in constructed HEK293 stable cell-lines. Using a knock-in mouse model of AD (APPNL−F/NL−F), which expresses a mutant form of human amyloid-β (Aβ), we performed immunofluorescence studies combined with electrophysiological whole-cell recordings to investigate the effects of our key molecule, α5-SOP002 in the hippocampal CA1 region. In aged APPNL−F/NL−F mice, selective preservation of α5 GABAARs was observed in, calretinin- (CR), cholecystokinin- (CCK), somatostatin- (SST) expressing interneurons, and pyramidal cells. Previously, we reported that CR dis-inhibitory interneurons, specialized in regulating other interneurons displayed abnormally high levels of synaptic inhibition in the APPNL−F/NL−F mouse model, here we show that this excessive inhibition was “normalized” to control values with bath-applied α5-SOP002 (1 μM). However, α5-SOP002, further impaired inhibition onto CCK and pyramidal cells that were already largely compromised by exhibiting a deficit of inhibition in the AD model. In summary, using a multi-disciplinary approach, we show that exposure to α5 GABAAR NAMs may further compromise aberrant synapses in AD. We, therefore, suggest that the α5 GABAAR is not a suitable therapeutic target for the treatment of AD or other cognitive deficits due to the widespread neuronal-networks that use α5 GABAARs.
- Published
- 2020
- Full Text
- View/download PDF
10. Commensal gut microbiota-derived acetate and propionate enhance heart adaptation in response to cardiac pressure overload in mice
- Author
-
Chen-Ju Lin, Yu-Che Cheng, Hung-Chih Chen, Yu-Kai Chao, Martin W. Nicholson, Eric C.L. Yen, Timothy J. Kamp, and Patrick C.H. Hsieh
- Subjects
Mice ,Animals ,Medicine (miscellaneous) ,Heart ,Propionates ,Acetates ,Fibrosis ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Gastrointestinal Microbiome - Published
- 2022
- Full Text
- View/download PDF
11. Population-based high-throughput toxicity screen of human iPSC-derived cardiomyocytes and neurons
- Author
-
Ching Ying Huang, Martin W. Nicholson, Jyun Yuan Wang, Chien Yu Ting, Ming Heng Tsai, Yu Che Cheng, Chun Lin Liu, Darien Z.H. Chan, Yi Chan Lee, Ching Chuan Hsu, Yu Hung Hsu, Chiou Fong Yang, Cindy M.C. Chang, Shu Chian Ruan, Po Ju Lin, Jen Hao Lin, Li Lun Chen, Marvin L. Hsieh, Yuan Yuan Cheng, Wan Tseng Hsu, Yi Ling Lin, Chien Hsiun Chen, Yu Hsiang Hsu, Ying Ta Wu, Timothy A. Hacker, Joseph C. Wu, Timothy J. Kamp, and Patrick C.H. Hsieh
- Subjects
Neurons ,Mice ,Drug-Related Side Effects and Adverse Reactions ,Induced Pluripotent Stem Cells ,Animals ,Humans ,Cell Differentiation ,Myocytes, Cardiac ,General Biochemistry, Genetics and Molecular Biology ,Cardiotoxicity ,Cells, Cultured - Abstract
In this study, we establish a population-based human induced pluripotent stem cell (hiPSC) drug screening platform for toxicity assessment. After recruiting 1,000 healthy donors and screening for high-frequency human leukocyte antigen (HLA) haplotypes, we identify 13 HLA-homozygous "super donors" to represent the population. These "super donors" are also expected to represent at least 477,611,135 of the global population. By differentiating these representative hiPSCs into cardiomyocytes and neurons we show their utility in a high-throughput toxicity screen. To validate hit compounds, we demonstrate dose-dependent toxicity of the hit compounds and assess functional modulation. We also show reproducible in vivo drug toxicity results using mouse models with select hit compounds. This study shows the feasibility of using a population-based hiPSC drug screening platform to assess cytotoxicity, which can be used as an innovative tool to study inter-population differences in drug toxicity and adverse drug reactions in drug discovery applications.
- Published
- 2021
12. Inhibitory synapse formation in a co-culture model incorporating GABAergic medium spiny neurons and HEK293 cells stably expressing GABAA receptors
- Author
-
Laura E, Brown, Celine, Fuchs, Martin W, Nicholson, F Anne, Stephenson, Alex M, Thomson, and Jasmina N, Jovanovic
- Subjects
stable cell lines ,medium spiny neurons ,Presynaptic Terminals ,Rats, Sprague-Dawley ,Mice ,Pregnancy ,Animals ,Humans ,HEK 293 cell line ,Issue 93 ,GABAergic Neurons ,gamma-Aminobutyric Acid ,Mice, Inbred BALB C ,synaptogenesis ,Synaptic Potentials ,Receptors, GABA-A ,synaptic inhibition ,co-culture ,Coculture Techniques ,Corpus Striatum ,Rats ,HEK293 Cells ,nervous system ,Synapses ,GABAergic ,Developmental neuroscience ,Female ,Neuroscience - Abstract
Inhibitory neurons act in the central nervous system to regulate the dynamics and spatio-temporal co-ordination of neuronal networks. GABA (γ-aminobutyric acid) is the predominant inhibitory neurotransmitter in the brain. It is released from the presynaptic terminals of inhibitory neurons within highly specialized intercellular junctions known as synapses, where it binds to GABAA receptors (GABAARs) present at the plasma membrane of the synapse-receiving, postsynaptic neurons. Activation of these GABA-gated ion channels leads to influx of chloride resulting in postsynaptic potential changes that decrease the probability that these neurons will generate action potentials. During development, diverse types of inhibitory neurons with distinct morphological, electrophysiological and neurochemical characteristics have the ability to recognize their target neurons and form synapses which incorporate specific GABAARs subtypes. This principle of selective innervation of neuronal targets raises the question as to how the appropriate synaptic partners identify each other. To elucidate the underlying molecular mechanisms, a novel in vitro co-culture model system was established, in which medium spiny GABAergic neurons, a highly homogenous population of neurons isolated from the embryonic striatum, were cultured with stably transfected HEK293 cell lines that express different GABAAR subtypes. Synapses form rapidly, efficiently and selectively in this system, and are easily accessible for quantification. Our results indicate that various GABAAR subtypes differ in their ability to promote synapse formation, suggesting that this reduced in vitro model system can be used to reproduce, at least in part, the in vivo conditions required for the recognition of the appropriate synaptic partners and formation of specific synapses. Here the protocols for culturing the medium spiny neurons and generating HEK293 cells lines expressing GABAARs are first described, followed by detailed instructions on how to combine these two cell types in co-culture and analyze the formation of synaptic contacts.
- Published
- 2014
13. Inhibitory Synapse Formation in a Co-culture Model Incorporating GABAergic Medium Spiny Neurons and HEK293 Cells Stably Expressing GABAA Receptors
- Author
-
Jasmina N. Jovanovic, Alex M. Thomson, F. Anne Stephenson, Martin W. Nicholson, Celine Fuchs, and Laura E. Brown
- Subjects
General Immunology and Microbiology ,General Chemical Engineering ,General Neuroscience ,General Biochemistry, Genetics and Molecular Biology - Published
- 2014
- Full Text
- View/download PDF
14. Affinity and kinetic analysis of L-selectin (CD62L) binding to glycosylation-dependent cell-adhesion molecule-1
- Author
-
Martin W. Nicholson, A. Neil Barclay, P. Anton van der Merwe, Steven D. Rosen, and Mark S. Singer
- Subjects
High endothelial venules ,Biochemistry ,Mice ,Animals ,L-Selectin ,Cell adhesion ,Molecular Biology ,chemistry.chemical_classification ,biology ,Cell adhesion molecule ,Mucins ,Antibodies, Monoclonal ,Cell Biology ,Adhesion ,Rats ,Dissociation constant ,Kinetics ,chemistry ,CD4 Antigens ,biology.protein ,L-selectin ,Glycoprotein ,Selectin ,Protein Binding ,Signal Transduction - Abstract
The selectin family of cell adhesion molecules mediates the tethering and rolling of leukocytes on blood vessel endothelium. It has been postulated that the molecular basis of this highly dynamic adhesion is the low affinity and rapid kinetics of selectin interactions. However, affinity and kinetic analyses of monomeric selectins binding their natural ligands have not previously been reported. Leukocyte selectin (L-selectin, CD62L) binds preferentially to O-linked carbohydrates present on a small number of mucin-like glycoproteins, such as glycosylation-dependent cell adhesion molecule-1 (GlyCAM-1), expressed in high endothelial venules. GlyCAM-1 is a soluble secreted protein which, following binding to CD62L, stimulates beta2-integrin-mediated adhesion of lymphocytes. Using surface plasmon resonance, we show that a soluble monomeric form of CD62L binds to purified immobilized GlyCAM-1 with a dissociation constant (Kd) of 108 microM. CD62L dissociates from GlyCAM-1 with a very fast dissociation rate constant (>/=10 s-1) which agrees well with the reported dissociation rate constant of CD62L-mediated leukocyte tethers. The calculated association rate constant is >/=10(5) M-1 s-1. At concentrations just above its mean serum level (approximately 1.5 microg/ml or approximately 30 nM), GlyCAM-1 binds multivalently to immobilized CD62L. It follows that soluble GlyCAM-1 may cross-link CD62L when it binds to cells, suggesting a mechanism for signal transduction.
- Published
- 1998
15. CD4+CD8- thymocytes that express L-selectin protect rats from diabetes upon adoptive transfer
- Author
-
Ben Seddon, Abdelhadi Saoudi, Martin W. Nicholson, and Don Mason
- Subjects
Adoptive cell transfer ,medicine.medical_specialty ,medicine.medical_treatment ,T cell ,CD8 Antigens ,Immunology ,Thymus Gland ,medicine.disease_cause ,Autoimmunity ,Graft vs Host Reaction ,T-Lymphocyte Subsets ,Internal medicine ,medicine ,Immunology and Allergy ,Animals ,L-Selectin ,biology ,Antibodies, Monoclonal ,Rats, Inbred Strains ,Th1 Cells ,Adoptive Transfer ,Cell biology ,Rats ,Thymectomy ,Thymocyte ,Endocrinology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 1 ,CD4 Antigens ,biology.protein ,Cytokines ,L-selectin ,Antibody ,CD8 - Abstract
Previous studies have shown that insulin-dependent diabetes can be induced in normal PVG.RT1u rats by a protocol of adult thymectomy and irradiation. The injection of CD4+ T cells from non-irradiated syngeneic donors prevents the onset of disease in approximately 50% of pre-diabetic recipients but all rats are protected if a particular subset of CD4+ cells is transferred. These protective cells express TCR alpha beta and have a memory phenotype, being CD45RClow RT6+. Further studies have demonstrated that the transfer of CD4+CD8- thymocytes, like that of unfractionated CD4+ peripheral T cells, also protects approximately half of recipients from diabetes suggesting that, as with the peripheral T cells, a functional heterogeneity may exist amongst CD4+CD8- thymocytes. In this study, we show that L-selectin is expressed by 50-60% of all CD4+CD8- thymocytes from 6-week-old rats. Adoptive transfer of these populations into thymectomized and irradiated rats revealed that the protection from diabetes observed by CD4+CD8- thymocytes was mediated almost entirely by the L-selectin+ subset. Cells with this phenotype were also able to mediate both humoral and cell mediated responses, providing primed B cells with help for secondary antibody responses and mediating local graft-versus-host reactions. L-selectin- CD4+CD8- thymocytes failed to mediate these responses. These data indicate that CD4+CD8- thymocytes must mature to the stage of L-selectin expression, before they can mediate normal T cell function. The implications of these results are discussed with respect to the possible role of murine NK1.1+ thymocytes in the control of autoimmunity.
- Published
- 1996
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.