Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.447
Filter
1.
Mol Ther ; 31(2): 331-343, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36575793

ABSTRACT

Leukemia inhibitory factor (LIF) is a pleiotropic cytokine of the interleukin-6 (IL-6) superfamily. LIF was initially discovered as a factor to induce the differentiation of myeloid leukemia cells and thus inhibit their proliferation. Subsequent studies have highlighted the multi-functions of LIF under a wide variety of physiological and pathological conditions in a highly cell-, tissue-, and context-dependent manner. Emerging evidence has demonstrated that LIF plays an essential role in the stem cell niche, where it maintains the homeostasis and regeneration of multiple somatic tissues, including intestine, neuron, and muscle. Further, LIF exerts a crucial regulatory role in immunity and functions as a protective factor against many immunopathological diseases, such as infection, inflammatory bowel disease (IBD), and graft-verse-host disease (GVHD). It is worth noting that while LIF displays a tumor-suppressive function in leukemia, recent studies have highlighted the oncogenic role of LIF in many types of solid tumors, further demonstrating the complexities and context-dependent effects of LIF. In this review, we summarize the recent insights into the roles and mechanisms of LIF in stem cell homeostasis and regeneration, immunity, and cancer, and discuss the potential therapeutic options for human diseases by modulating LIF levels and functions.


Subject(s)
Growth Inhibitors , Interleukin-6 , Humans , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Growth Inhibitors/pharmacology , Growth Inhibitors/physiology , Cell Differentiation , Leukemia Inhibitory Factor Receptor alpha Subunit , Lymphokines/pharmacology , Lymphokines/physiology
2.
PLoS Comput Biol ; 17(11): e1009576, 2021 11.
Article in English | MEDLINE | ID: mdl-34748539

ABSTRACT

Advances in genetic engineering technologies have allowed the construction of artificial genetic circuits, which have been used to generate spatial patterns of differential gene expression. However, the question of how cells can be programmed, and how complex the rules need to be, to achieve a desired tissue morphology has received less attention. Here, we address these questions by developing a mathematical model to study how cells can collectively grow into clusters with different structural morphologies by secreting diffusible signals that can influence cellular growth rates. We formulate how growth regulators can be used to control the formation of cellular protrusions and how the range of achievable structures scales with the number of distinct signals. We show that a single growth inhibitor is insufficient for the formation of multiple protrusions but may be achieved with multiple growth inhibitors, and that other types of signals can regulate the shape of protrusion tips. These examples illustrate how our approach could potentially be used to guide the design of regulatory circuits for achieving a desired target structure.


Subject(s)
Cell Proliferation/physiology , Cell Shape/physiology , Cellular Reprogramming Techniques/methods , Models, Biological , Animals , Cell Aggregation/physiology , Cell Communication/physiology , Cell Surface Extensions/physiology , Cellular Reprogramming Techniques/statistics & numerical data , Computational Biology , Computer Simulation , Gene Regulatory Networks , Genetic Engineering/methods , Genetic Engineering/statistics & numerical data , Growth Inhibitors/physiology , Humans , Morphogenesis/physiology , Synthetic Biology
3.
Gut ; 64(7): 1028-39, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25107557

ABSTRACT

OBJECTIVE: In this study, we investigated the role of Trefoil factor 1 (TFF1) in regulating cell proliferation and tumour development through ß-catenin signalling using in vivo and in vitro models of gastric tumorigenesis. DESIGN: Tff1-knockout (Tff1-KO) mice, immunohistochemistry, luciferase reporter, qRT-PCR, immunoblot, and phosphatase assays were used to examine the role of TFF1 on ß-catenin signalling pathway. RESULTS: Nuclear localisation of ß-catenin with transcriptional upregulation of its target genes, c-Myc and Ccnd1, was detected in hyperplastic tissue at an early age of 4-6 weeks and maintained during all stages of gastric tumorigenesis in the Tff1-KO mice. The reconstitution of TFF1 or TFF1 conditioned media significantly inhibited the ß-catenin/T-cell factor (TCF) transcription activity in MKN28 gastric cancer cells. In agreement with these results, we detected a reduction in the levels of nuclear ß-catenin with downregulation of c-MYC and CCND1 mRNA. Analysis of signalling molecules upstream of ß-catenin revealed a decrease in phosphorylated glycogen synthase kinase 3ß (p-GSK3ß) (Ser9) and p-AKT (Ser473) protein levels following the reconstitution of TFF1 expression; this was consistent with the increase of p-ß-catenin (Ser33/37/Thr41) and decrease of p-ß-catenin (Ser552). This TFF1-induced reduction in phosphorylation of GSK3ß, and AKT was dependent on protein phosphatase 2A (PP2A) activity. The treatment with okadaic acid or knockdown of PP2A abrogated these effects. Consistent with the mouse data, we observed loss of TFF1 and an increase in nuclear localisation of ß-catenin in stages of human gastric tumorigenesis. CONCLUSIONS: Our data indicate that loss of TFF1 promotes ß-catenin activation and gastric tumorigenesis through regulation of PP2A, a major regulator of AKT-GSK3ß signalling.


Subject(s)
Growth Inhibitors/physiology , Peptides/physiology , Protein Phosphatase 2/physiology , Proto-Oncogene Proteins c-akt/physiology , Stomach Neoplasms/pathology , Stomach Neoplasms/physiopathology , beta Catenin/physiology , Animals , Cell Line, Tumor , Cell Proliferation , Down-Regulation/physiology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Immunohistochemistry , Mice , Mice, Knockout , Transcriptional Activation/physiology , Trefoil Factor-1
4.
Curr Alzheimer Res ; 10(7): 706-13, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23905992

ABSTRACT

Sequential cleavages of APP by ß-secretase and γ-secretase release ß-amyloid (Aß) and one secreted form of APP (sAPP-ß) in Alzheimer' s disease (AD). Alternatively, in non-pathological situations, APP is predominantly cleaved by α-secretase within the amyloid sequence, to release the other soluble form of APP, sAPP-α. However, the functions of the two types of sAPP are still unclear. We performed this study to compare the function of sAPP-α and sAPP-ß in differentiation of the glioma cell line U251. We found that sAPP-α suppressed astrocytic differentiation and promoted neuronal differentiation in U251 cells. Additionally, sAPP-α enhanced U251 terminal differentiation into a cholinergic-like neuronal phenotype. In contrast, sAPP-ß suppressed neuronal differentiation and promoted the astrocytic differentiation of U251 cells. These findings could not only enrich the knowledge of the potential physiological function of sAPP-α and sAPP-ß, but also indicate that they may be connected to the pathological mechanism of AD. Furthermore, these findings suggest that new strategies, such as increasing the level of sAPP-α and/or decreasing the level of sAPP-ß in brain, or transplanting stem cells with increased sAPP-α and/or decreased sAPP-ß, may have potential value for AD treatment.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Astrocytes/physiology , Cell Differentiation/physiology , Neurons/physiology , Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Cell Line, Tumor , Glioma/chemistry , Glioma/pathology , Growth Inhibitors/physiology , Humans , Neurons/metabolism , Neurons/pathology
5.
J Neurosci ; 33(28): 11479-93, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23843519

ABSTRACT

Toll-like receptors (TLRs) recognize both pathogen- and danger-associated molecular patterns and induce innate immune responses. Some TLRs are expressed in neurons and regulate neurodevelopment and neurodegeneration. However, the downstream signaling pathways and effectors for TLRs in neurons are still controversial. In this report, we provide evidence that TLR7 negatively regulates dendrite growth through the canonical myeloid differentiation primary response gene 88 (Myd88)-c-Fos-interleukin (IL)-6 pathway. Although both TLR7 and TLR8 recognize single-stranded RNA (ssRNA), the results of quantitative reverse transcription-PCR suggested that TLR7 is the major TLR recognizing ssRNA in brains. In both in vitro cultures and in utero electroporation experiments, manipulation of TLR7 expression levels was sufficient to alter neuronal morphology, indicating the presence of intrinsic TLR7 ligands. Besides, the RNase A treatment that removed ssRNA in cultures promoted dendrite growth. We also found that the addition of ssRNA and synthetic TLR7 agonists CL075 and loxoribine, but not R837 (imiquimod), to cultured neurons specifically restricted dendrite growth via TLR7. These results all suggest that TLR7 negatively regulates neuronal differentiation. In cultured neurons, TLR7 activation induced IL-6 and TNF-α expression through Myd88. Using Myd88-, IL-6-, and TNF-α-deficient neurons, we then demonstrated the essential roles of Myd88 and IL-6, but not TNF-α, in the TLR7 pathway to restrict dendrite growth. In addition to neuronal morphology, TLR7 knockout also affects mouse behaviors, because young mutant mice ∼2 weeks of age exhibited noticeably lower exploratory activity in an open field. In conclusion, our study suggests that TLR7 negatively regulates dendrite growth and influences cognition in mice.


Subject(s)
Dendrites/physiology , Down-Regulation/physiology , Growth Inhibitors/physiology , Interleukin-6/physiology , Membrane Glycoproteins/physiology , Myeloid Differentiation Factor 88/physiology , Proto-Oncogene Proteins c-fos/physiology , Signal Transduction/physiology , Toll-Like Receptor 7/physiology , Animals , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy
6.
Anticancer Drugs ; 24(7): 667-76, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23695011

ABSTRACT

Malignant melanoma is the most aggressive form of skin cancer. Although various antimelanoma approaches have been used in the clinics to treat the disease over the last three decades, none of the drugs significantly prolonged the survival of metastatic melanoma patients; hence, effective drugs against metastatic melanoma are highly desired. In this study, we explored an antimetastatic melanoma agent derived from traditional Chinese medicinal herbs and found that jatrorrhizine hydrochloride (JH), an active component of the traditional Chinese medicinal herb Coptis chinensis, inhibited the proliferation and neovascularization of C8161 human metastatic melanoma cells. JH suppressed C8161 cell proliferation in a dose-dependent manner, with a half-maximal inhibitory concentration of 47.4±1.6 µmol/l; however, it did not induce significant cellular apoptosis at doses up to 320 µmol/l. Mechanistic studies showed that JH-induced C8161 cell cycle arrest at the G0/G1 transition, which was accompanied by overexpression of the cell cycle-suppressive genes p21 and p27 at higher doses. Moreover, JH reduced C8161 cell-mediated neovascularization in vitro and in vivo and impeded the expression of the gene for VE-cadherin, a key protein in tumor vasculogenic mimicry and angiogenesis. Taken together, the effective inhibitory effects of JH on metastatic melanoma cell proliferation and neovascularization with low toxicity suggest that JH is a potential new antimelanoma drug candidate.


Subject(s)
Berberine/analogs & derivatives , Growth Inhibitors/physiology , Melanoma/drug therapy , Melanoma/prevention & control , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/prevention & control , Animals , Berberine/pharmacology , Berberine/therapeutic use , Cell Line, Tumor , Growth Inhibitors/pharmacology , Humans , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/pathology
7.
J Immunol ; 191(1): 164-70, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23720815

ABSTRACT

NO is a free radical with pleiotropic functions. We have shown earlier that NO induces a population of CD4(+)CD25(+)Foxp3(-) regulatory T cells (NO-Tregs) that suppress the functions of CD4(+)CD25(-) effector T cells in vitro and in vivo. We report in this study an unexpected finding that NO-Tregs suppressed Th17 but not Th1 cell differentiation and function. In contrast, natural Tregs (nTregs), which suppressed Th1 cells, failed to suppress Th17 cells. Consistent with this observation, NO-Tregs inhibited the expression of retinoic acid-related orphan receptor γt but not T-bet, whereas nTregs suppressed T-bet but not retinoic acid-related orphan receptor γt expression. The NO-Treg-mediated suppression of Th17 was partially cell contact-dependent and was associated with IL-10. In vivo, adoptively transferred NO-Tregs potently attenuated experimental autoimmune encephalomyelitis. The disease suppression was accompanied by a reduction of Th17, but not Th1 cells in the draining lymph nodes, and a decrease in the production of IL-17, but an increase in IL-10 synthesis. Our results therefore demonstrate the differential suppressive function between NO-Tregs and nTregs and indicate specialization of the regulatory mechanism of the immune system.


Subject(s)
Cell Differentiation/immunology , Growth Inhibitors/physiology , Nitric Oxide/physiology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Cell Communication/immunology , Cells, Cultured , Female , Growth Inhibitors/pharmacology , Immune Tolerance/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/pharmacology , Primary Cell Culture , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology , Th17 Cells/cytology
8.
Brain ; 136(Pt 5): 1488-507, 2013 May.
Article in English | MEDLINE | ID: mdl-23587805

ABSTRACT

Tripeptidyl peptidase 1 (TPP1) deficiency causes CLN2 disease, late infantile (or classic late infantile neuronal ceroid lipofuscinosis), a paediatric neurodegenerative disease of autosomal recessive inheritance. Patients suffer from blindness, ataxia, epilepsy and cognitive defects, with MRI indicating widespread brain atrophy, and profound neuron loss is evident within the retina and brain. Currently there are no effective therapies for this disease, which causes premature death in adolescence. Zebrafish have been successfully used to model a range of neurological and behavioural abnormalities. The aim of this study was to characterize the pathological and functional consequences of Tpp1 deficiency in zebrafish and to correlate these with human CLN2 disease, thereby providing a platform for drug discovery. Our data show that homozygous tpp1(sa0011) mutant (tpp1(sa0011)(-/-)) zebrafish display a severe, progressive, early onset neurodegenerative phenotype, characterized by a significantly small retina, a small head and curved body. The mutant zebrafish have significantly reduced median survival with death occurring 5 days post-fertilization. As in human patients with CLN2 disease, mutant zebrafish display storage of subunit c of mitochondrial ATP-synthase, hypertrophic lysosomes as well as localized apoptotic cell death in the retina, optic tectum and cerebellum. Further neuropathological phenotypes of these mutants provide novel insights into mechanisms of pathogenesis in CLN2 disease. Secondary neurogenesis in the retina, optic tectum and cerebellum is impaired and axon tracts within the spinal cord, optic nerve and the posterior commissure are disorganized, with the optic nerve failing to reach its target. This severe neurodegenerative phenotype eventually results in functional motor impairment, but this is preceded by a phase of hyperactivity that is consistent with seizures. Importantly, both of these locomotion phenotypes can be assayed in an automated manner suitable for high-throughput studies. Our study provides proof-of-principle that tpp1(sa0011)(-/-) mutants can utilize the advantages of zebrafish for understanding pathogenesis and drug discovery in CLN2 disease and other epilepsies.


Subject(s)
Aminopeptidases/deficiency , Cell Proliferation , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/deficiency , Disease Progression , Neuronal Ceroid-Lipofuscinoses/enzymology , Neuronal Ceroid-Lipofuscinoses/pathology , Serine Proteases/deficiency , Aminopeptidases/genetics , Aminopeptidases/physiology , Animals , Animals, Genetically Modified , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/physiology , Disease Models, Animal , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Humans , Motor Activity/physiology , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neuronal Ceroid-Lipofuscinoses/genetics , Serine Proteases/genetics , Serine Proteases/physiology , Tripeptidyl-Peptidase 1 , Zebrafish
9.
Glia ; 61(4): 539-49, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23355222

ABSTRACT

Microglial cells constitute the first line of defense of the central nervous system (CNS) against microbial invasion. Pathogens are detected thanks to an array of innate immune receptors termed pattern recognition receptors (PRRs). PRRs have been thoroughly characterized in bone marrow-derived macrophages, but the PRRs repertoire and functionality in microglial cells remain largely unknown. Microglial cells express various Toll-like Receptors and the Nod1/2 receptors. Recently, a novel innate immune signalling pathway, the inflammasome pathway has been uncovered. Inflammasome activation leads to caspase-1 activation, release of the proinflammatory cytokines, IL-1ß and IL-18 and cell death in a process termed pyroptosis. One inflammasome receptor, NLRP3, has been characterized in microglial cells and associated with response to infections and in the initiation of neuro-degeneration in an Alzheimer's disease model. Legionella pneumophila (L.pneumophila) is a flagellated bacterium replicating within macrophages. In bone marrow-derived macrophages, L. pneumophila is detected in a flagellin-dependent manner by the Naip5-NLRC4 (Ipaf) inflammasome pathway. In this study, we decided to use L. pneumophila to investigate the presence and the functionality of this inflammasome in primary murine microglial cells. We show that microglial cells detect L. pneumophila infection in a flagellin-dependent manner leading to caspase-1-mediated bacterial growth restriction, infected cell death and secretion of the proinflammatory cytokines IL-1ß and IL18. Overall, our data demonstrate that microglial cells have a functional Naip5-NLRC4 inflammasome likely to be important to monitor and clear CNS infections by flagellated bacteria.


Subject(s)
Flagellin/metabolism , Growth Inhibitors/metabolism , Inflammasomes/metabolism , Legionella pneumophila/pathogenicity , Legionnaires' Disease/metabolism , Legionnaires' Disease/microbiology , Microglia/cytology , Microglia/microbiology , Animals , Animals, Newborn , Apoptosis , Apoptosis Regulatory Proteins/physiology , Calcium-Binding Proteins/physiology , Cells, Cultured , Flagellin/pharmacology , Growth Inhibitors/physiology , Inflammasomes/physiology , Legionella pneumophila/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Neuronal Apoptosis-Inhibitory Protein/physiology
10.
Toxicol Appl Pharmacol ; 267(3): 228-37, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23352504

ABSTRACT

Accumulated evidence has revealed a tight link between arsenic trioxide (ATO)-induced apoptosis and mitotic arrest in cancer cells. AKT, a serine/threonine kinase frequently over-activated in diverse tumors, plays critical roles in stimulating cell cycle progression, abrogating cell cycle checkpoints, suppressing apoptosis, and regulating mitotic spindle assembly. Inhibition of AKT may therefore enhance ATO cytotoxicity and thus its clinical utility. We show that AKT was activated by ATO in HeLa-S3 cells. Inhibition of AKT by inhibitors of the phosphatidyl inositol 3-kinase/AKT pathway significantly enhanced cell sensitivity to ATO by elevating mitotic cell apoptosis. Ectopic expression of the constitutively active AKT1 had no effect on ATO-induced spindle abnormalities but reduced kinetochore localization of BUBR1 and MAD2 and accelerated mitosis exit, prevented mitotic cell apoptosis, and enhanced the formation of micro- or multi-nuclei in ATO-treated cells. These results indicate that AKT1 activation may prevent apoptosis of ATO-arrested mitotic cells by attenuating the function of the spindle checkpoint and therefore allowing the formation of micro- or multi-nuclei in surviving daughter cells. In addition, AKT1 activation upregulated the expression of aurora kinase B (AURKB) and survivin, and depletion of AURKB or survivin reversed the resistance of AKT1-activated cells to ATO-induced apoptosis. Thus, AKT1 activation suppresses ATO-induced mitotic cell apoptosis, despite the presence of numerous spindle abnormalities, probably by upregulating AURKB and survivin and attenuating spindle checkpoint function. Inhibition of AKT therefore effectively sensitizes cancer cells to ATO by enhancing mitotic cell apoptosis.


Subject(s)
Apoptosis/drug effects , Mitosis/drug effects , Oxides/toxicity , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/physiology , Up-Regulation/drug effects , Apoptosis/physiology , Arsenic Trioxide , Arsenicals , Growth Inhibitors/antagonists & inhibitors , Growth Inhibitors/physiology , Growth Inhibitors/toxicity , HeLa Cells , Humans , Mitosis/physiology , Up-Regulation/physiology
11.
J Immunol ; 190(4): 1447-56, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23296704

ABSTRACT

Nucleic acid-reactive B cells frequently arise in the bone marrow but are tolerized by mechanisms including receptor editing, functional anergy, and/or deletion. TLR9, a sensor of endosomal dsDNA, both promotes and regulates systemic autoimmunity in vivo, but the precise nature of its apparently contradictory roles in autoimmunity remained unclear. In this study, using the 3H9 anti-DNA BCR transgene in the autoimmune-prone MRL.Fas(lpr) mouse model of systemic lupus erythematosus, we identify the stages at which TLR9 contributes to establishing and breaking B cell tolerance. Although TLR9 is dispensable for L chain editing during B cell development in the bone marrow, TLR9 limits anti-DNA B cell life span in the periphery and is thus tolerogenic. In the absence of TLR9, anti-DNA B cells have much longer life spans and accumulate in the follicle, neither activated nor deleted. These cells retain some characteristics of anergic cells, in that they have elevated basal BCR signaling but impaired induced responses and downregulate their cell-surface BCR expression. In contrast, whereas TLR9-intact anergic B cells accumulate near the T/B border, TLR9-deficient anti-DNA B cells are somewhat more dispersed throughout the follicle. Nonetheless, in older autoimmune-prone animals, TLR9 expression specifically within the B cell compartment is required for spontaneous peripheral activation of anti-DNA B cells and their differentiation into Ab-forming cells via an extrafollicular pathway. Thus, TLR9 has paradoxical roles in regulating anti-DNA B cells: it helps purge the peripheral repertoire of autoreactive cells, yet is also required for their activation.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Clonal Anergy/immunology , DNA/immunology , Growth Inhibitors/physiology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Toll-Like Receptor 9/physiology , Animals , B-Lymphocyte Subsets/cytology , Clonal Anergy/genetics , Growth Inhibitors/genetics , Humans , Mice , Mice, Inbred MRL lpr , Mice, Knockout , Mice, Transgenic , Radiation Chimera/immunology , Toll-Like Receptor 9/genetics
12.
PLoS One ; 8(1): e52143, 2013.
Article in English | MEDLINE | ID: mdl-23320069

ABSTRACT

BACKGROUND: Recent studies show the importance of interactions between CD47 expressed on acute myeloid leukemia (AML) cells and the inhibitory immunoreceptor, signal regulatory protein-alpha (SIRPα) on macrophages. Although AML cells express SIRPα, its function has not been investigated in these cells. In this study we aimed to determine the role of the SIRPα in acute myeloid leukemia. DESIGN AND METHODS: We analyzed the expression of SIRPα, both on mRNA and protein level in AML patients and we further investigated whether the expression of SIRPα on two low SIRPα expressing AML cell lines could be upregulated upon differentiation of the cells. We determined the effect of chimeric SIRPα expression on tumor cell growth and programmed cell death by its triggering with an agonistic antibody in these cells. Moreover, we examined the efficacy of agonistic antibody in combination with established antileukemic drugs. RESULTS: By microarray analysis of an extensive cohort of primary AML samples, we demonstrated that SIRPα is differentially expressed in AML subgroups and its expression level is dependent on differentiation stage, with high levels in FAB M4/M5 AML and low levels in FAB M0-M3. Interestingly, AML patients with high SIRPα expression had a poor prognosis. Our results also showed that SIRPα is upregulated upon differentiation of NB4 and Kasumi cells. In addition, triggering of SIRPα with an agonistic antibody in the cells stably expressing chimeric SIRPα, led to inhibition of growth and induction of programmed cell death. Finally, the SIRPα-derived signaling synergized with the activity of established antileukemic drugs. CONCLUSIONS: Our data indicate that triggering of SIRPα has antileukemic effect and may function as a potential therapeutic target in AML.


Subject(s)
Antigens, Differentiation/metabolism , Apoptosis , Growth Inhibitors/physiology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, Immunologic/metabolism , Adult , Antibodies, Monoclonal/administration & dosage , Antigens, Differentiation/genetics , Antineoplastic Agents/administration & dosage , Apoptosis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Child , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/therapy , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Leukemia, Promyelocytic, Acute/therapy , Molecular Targeted Therapy , Prognosis , Receptors, Immunologic/genetics , Signal Transduction/genetics
13.
Circ Res ; 112(2): 257-66, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23168335

ABSTRACT

RATIONALE: Nkx2.5 is a transcription factor that regulates cardiomyogenesis in vivo and in embryonic stem cells. It is also a common target in congenital heart disease. Although Nkx2.5 has been implicated in the regulation of many cellular processes that ultimately contribute to cardiomyogenesis and morphogenesis of the mature heart, relatively little is known about how it is regulated at a functional level. OBJECTIVE: We have undertaken a proteomic screen to identify novel binding partners of Nkx2.5 during cardiomyogenic differentiation in an effort to better understand the regulation of its transcriptional activity. METHODS AND RESULTS: Purification of Nkx2.5 from differentiating cells identified the myosin phosphatase subunits protein phosphatase 1ß and myosin phosphatase targeting subunit 1 (Mypt1) as novel binding partners. The interaction with protein phosphatase 1 ß/Mypt1 resulted in exclusion of Nkx2.5 from the nucleus and, consequently, inhibition of its transcriptional activity. Exclusion of Nkx2.5 was inhibited by treatment with leptomycin B and was dependent on an Mypt1 nuclear export signal. Furthermore, in transient transfection experiments, Nkx2.5 colocalized outside the nucleus with phosphorylated Mypt1 in a manner dependent on Wnt signaling and Rho-associated protein kinase. Treatment of differentiating mouse embryonic stem cells with Wnt3a resulted in enhanced phosphorylation of endogenous Mypt1, increased nuclear exclusion of endogenous Nkx2.5, and a failure to undergo terminal cardiomyogenesis. Finally, knockdown of Mypt1 resulted in rescue of Wnt3a-mediated inhibition of cardiomyogenesis, indicating that Mypt1 is required for this process. CONCLUSIONS: We have identified a novel interaction between Nkx2.5 and myosin phosphatase. Promoting this interaction represents a novel mechanism whereby Wnt3a regulates Nkx2.5 and inhibits cardiomyogenesis.


Subject(s)
Growth Inhibitors/physiology , Homeodomain Proteins/metabolism , Myocytes, Cardiac/physiology , Myosin-Light-Chain Phosphatase/physiology , Signal Transduction/physiology , Transcription Factors/metabolism , Wnt Signaling Pathway/physiology , Wnt3A Protein/physiology , rho-Associated Kinases/physiology , Animals , Embryonic Stem Cells/enzymology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , HEK293 Cells , Homeobox Protein Nkx-2.5 , Humans , Mice , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Protein Phosphatase 1/metabolism , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism
14.
Development ; 140(1): 66-75, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23222438

ABSTRACT

There is a gradient of ß-catenin expression along the colonic crypt axis with the highest levels at the crypt bottom. In addition, colorectal cancers show a heterogeneous subcellular pattern of ß-catenin accumulation. However, it remains unclear whether different levels of Wnt signalling exert distinct roles in the colonic epithelium. Here, we investigated the dose-dependent effect of canonical Wnt activation on colonic epithelial differentiation by controlling the expression levels of stabilised ß-catenin using a doxycycline-inducible transgenic system in mice. We show that elevated levels of Wnt signalling induce the amplification of Lgr5+ cells, which is accompanied by crypt fission and a reduction in cell proliferation among progenitor cells. By contrast, lower levels of ß-catenin induction enhance cell proliferation rates of epithelial progenitors without affecting crypt fission rates. Notably, slow-cycling cells produced by ß-catenin activation exhibit activation of Notch signalling. Consistent with the interpretation that the combination of Notch and Wnt signalling maintains crypt cells in a low proliferative state, the treatment of ß-catenin-expressing mice with a Notch inhibitor turned such slow-cycling cells into actively proliferating cells. Our results indicate that the activation of the canonical Wnt signalling pathway is sufficient for de novo crypt formation, and suggest that different levels of canonical Wnt activations, in cooperation with Notch signalling, establish a hierarchy of slower-cycling stem cells and faster-cycling progenitor cells characteristic for the colonic epithelium.


Subject(s)
Cell Cycle/physiology , Colon/cytology , Intestinal Mucosa/cytology , Signal Transduction/physiology , Wnt Signaling Pathway/physiology , Animals , Cell Line , Cell Proliferation , Colon/pathology , Colon/physiology , Gene Knock-In Techniques , Growth Inhibitors/physiology , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Stem Cells/cytology , Stem Cells/metabolism , beta Catenin/biosynthesis , beta Catenin/physiology
15.
J Immunol ; 190(1): 159-67, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23225884

ABSTRACT

MEK1 phosphorylates ERK1/2 and regulates T cell generation, differentiation, and function. MEK1 has recently been shown to translocate to the nucleus. Its nuclear function is largely unknown. By studying human CD4 T cells, we demonstrate that a low level of MEK1 is present in the nucleus of CD4 T cells under basal conditions. T cell activation further increases the nuclear translocation of MEK1. MEK1 interacts with the nuclear receptor corepressor silencing mediator of retinoid and thyroid hormone receptor (SMRT). MEK1 reduces the nuclear level of SMRT in an activation-dependent manner. MEK1 is recruited to the promoter of c-Fos upon TCR stimulation. Conversely, SMRT is bound to the c-Fos promoter under basal conditions and is removed upon TCR stimulation. We examined the role of SMRT in regulation of T cell function. Small interfering RNA-mediated knockdown of SMRT results in a biphasic effect on cytokine production. The production of the cytokines IL-2, IL-4, IL-10, and IFN-γ increases in the early phase (8 h) and then decreases in the late phase (48 h). The late-phase decrease is associated with inhibition of T cell proliferation. The late-phase inhibition of T cell activation is, in part, mediated by IL-10 that is produced in the early phase and, in part, by ß-catenin signaling. Thus, we have identified a novel nuclear function of MEK1. MEK1 triggers a complex pattern of early T cell activation, followed by a late inhibition through its interaction with SMRT. This biphasic dual effect most likely reflects a homeostatic regulation of T cell function by MEK1.


Subject(s)
Active Transport, Cell Nucleus/immunology , CD4-Positive T-Lymphocytes/immunology , MAP Kinase Kinase 1/physiology , Nuclear Receptor Co-Repressor 1/physiology , Nuclear Receptor Co-Repressor 2/antagonists & inhibitors , Nuclear Receptor Co-Repressor 2/physiology , Active Transport, Cell Nucleus/genetics , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/metabolism , Down-Regulation/genetics , Down-Regulation/immunology , Gene Silencing/immunology , Growth Inhibitors/genetics , Growth Inhibitors/metabolism , Growth Inhibitors/physiology , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Promoter Regions, Genetic/immunology , Protein Binding/genetics , Protein Binding/immunology , Proto-Oncogene Proteins c-fos/metabolism
16.
J Immunol ; 190(1): 138-46, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23225885

ABSTRACT

Adenosine is a key endogenous signaling molecule that regulates immune responses. A(2B) adenosine receptor (AR) is a relatively low-affinity receptor for adenosine, and the activation of A(2B)AR is believed to require pathological level of adenosine that is associated with ischemia, inflammation, trauma, or other types of stress. The role of A(2B)AR in the pathogenesis of multiple sclerosis (MS) is still unclear. In this study, we discovered that A(2B)AR was upregulated both in the peripheral blood leukocytes of MS patients and the peripheral lymphoid tissues of experimental autoimmune encephalomyelitis (EAE) mice. A(2B)AR-specific antagonists, CVT-6883 and MRS-1754, alleviated the clinical symptoms of EAE and protected the CNS from immune damage. A(2B)AR-knockout mice also developed less severe EAE. Further study indicated that blocking or deleting A(2B)AR inhibited Th17 cell differentiation by blocking IL-6 production from APCs such as dendritic cells. In dendritic cells, A(2B)AR was also upregulated during the development of EAE. CVT-6883 and genetic deletion of A(2B)AR significantly reduced adenosine-mediated IL-6 production. The phospholipase Cß-protein kinase C and p38 MAPK pathways were found to be involved in the A(2B)AR-mediated IL-6 production. Our findings not only revealed the pathological role of A(2B)AR in EAE, but also suggested that this receptor might be a new therapeutic target for the development of anti-MS drugs.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-6/antagonists & inhibitors , Receptor, Adenosine A2B/metabolism , Th17 Cells/immunology , Adult , Animals , Cells, Cultured , Coculture Techniques , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Growth Inhibitors/antagonists & inhibitors , Growth Inhibitors/physiology , Humans , Interleukin-6/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptor, Adenosine A2B/deficiency , Th17 Cells/metabolism , Th17 Cells/pathology , Up-Regulation/immunology
17.
J Immunol ; 190(1): 21-6, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23225888

ABSTRACT

Signaling lymphocytic activation molecule family receptors and the specific adapter signaling lymphocytic activation molecule-associated protein modulate the development of innate-like lymphocytes. In this study, we show that the thymus of Ly9-deficient mice contains an expanded population of CD8 single-positive cells with the characteristic phenotype of innate memory-like CD8(+) T cells. Moreover, the proportion of these innate CD8(+) T cells increased dramatically postinfection with mouse CMV. Gene expression profiling of Ly9-deficient mice thymi showed a significant upregulation of IL-4 and promyelocytic leukemia zinc finger. Analyses of Ly9(-/-)IL4ra(-/-) double-deficient mice revealed that IL-4 was needed to generate the thymic innate CD8(+) T cell subset. Furthermore, increased numbers of invariant NKT cells were detected in Ly9-deficient thymi. In wild-type mice, IL-4 levels induced by α-galactosylceramide injection could be inhibited by a mAb against Ly9. Thus, Ly9 plays a unique role as an inhibitory cell surface receptor regulating the size of the thymic innate CD8(+) T cell pool and the development of invariant NKT cells.


Subject(s)
Antigens, CD/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Down-Regulation/immunology , Immunologic Memory , Natural Killer T-Cells/immunology , Receptors, Cell Surface/physiology , Thymus Gland/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Down-Regulation/genetics , Female , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Immunity, Innate/genetics , Immunologic Memory/genetics , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Natural Killer T-Cells/cytology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family , Signaling Lymphocytic Activation Molecule Family Member 1 , Thymus Gland/cytology
18.
Invest Ophthalmol Vis Sci ; 53(13): 8433-46, 2012 Dec 19.
Article in English | MEDLINE | ID: mdl-23139280

ABSTRACT

PURPOSE: The secreted Ly6/uPAR-related protein-1 (Slurp1), associated with the hyperkeratotic disorder mal de Meleda, is abundantly expressed in corneas. Here, we examine its corneal expression and functions. METHODS: Gene expression was quantified by quantitative PCR (qPCR), immunoblots, and immunofluorescent staining. Effect of Kruppel-like factor 4 (Klf4) on Slurp1 promoter was evaluated by chromatin immunoprecipitation (ChIP) and transient transfections. Adenoviral vectors were used to express Slurp1 in corneas. Leukocytic infiltration in bacterial lipopolysaccharide (LPS)-, herpes simplex virus type 1 (HSV-1)-, or adenovirus (serotype 5)-treated mouse corneas was characterized by flow cytometry. RESULTS: Corneal expression of Slurp1 increased sharply upon mouse eyelid opening, concurrent with the elevated expression of Klf4. Slurp1 was significantly decreased in Klf4 conditional null (Klf4CN) corneas that displayed elevated expression of cytokines and cytokine receptors, as well as neutrophil influx consistent with a proinflammatory environment. In additional models of corneal inflammation, Slurp1 expression was abrogated within 24 hours of LPS injection or HSV-1 or adenoviral infection, accompanied by a predominantly neutrophilic infiltrate. Neutrophilic infiltration was enhanced in HSV-1-infected Klf4CN corneas lacking Slurp1. SLURP1 promoter activity was stimulated by KLF4, suppressed by IL-4, IL-13, and TNFα, and unperturbed by IFN-γ. Slurp1 downregulation and neutrophil influx were comparable in HSV-1-infected wild-type (WT) and Ifng-/- mouse corneas. Mouse corneas infected with Slurp1-expressing adenoviral vectors displayed reduced signs of inflammation and restricted neutrophilic infiltration compared with those infected with control vectors. CONCLUSIONS: Klf4 regulates the expression of Slurp1, a key immunomodulatory peptide that is abundantly expressed in healthy corneas and is downregulated in proinflammatory conditions.


Subject(s)
Antigens, Ly/genetics , Cornea/metabolism , Gene Expression Regulation/physiology , Growth Inhibitors/physiology , Kruppel-Like Transcription Factors/physiology , Urokinase-Type Plasminogen Activator/genetics , Adenoviridae/genetics , Animals , Antigens, Ly/metabolism , Base Sequence , Chromatin Immunoprecipitation , Cytokines/pharmacology , Female , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Genetic Vectors , Herpesvirus 1, Human/physiology , Immune System Diseases , Immunoblotting , Immunologic Factors/genetics , Immunologic Factors/metabolism , Keratitis, Herpetic/genetics , Keratitis, Herpetic/metabolism , Kruppel-Like Factor 4 , Leukocyte Disorders , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Sequence Data , Polymerase Chain Reaction , Transfection , Urokinase-Type Plasminogen Activator/metabolism
19.
J Neurosci ; 32(41): 14442-7, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23055514

ABSTRACT

Chondroitin sulfate proteoglycans (CSPGs) are a major component of the glial scar that contributes to the limited regeneration of the CNS after axonal injury. However, the intracellular mechanisms that mediate the effects of CSPGs are not fully understood. Here we show that axonal growth inhibition mediated by CSPGs requires intra-axonal protein synthesis. Application of CSPGs to postnatal rat dorsal root ganglia axons results in an increase in the axonal levels of phosphorylated 4E-BP1, a marker of increased protein translation. Axons grown in media containing CSPGs exhibit markedly reduced growth rates, which can be restored by the selective application of protein synthesis inhibitors to distal axons. We show that these axons contain transcripts encoding RhoA, a regulator of the cytoskeleton that is commonly used by the signaling pathways activated by many inhibitors of axon growth. We also show that selective application of CSPGs to axons results in increased intra-axonal synthesis of RhoA and that depletion of RhoA transcripts from axons results in enhanced growth of axons in the presence of CSPGs. These data identify local translation as an effector pathway of CSPGs and demonstrate that local translation of RhoA contributes to the axon growth inhibitory effect of CSPGs.


Subject(s)
Axons/physiology , Chondroitin Sulfate Proteoglycans/physiology , Growth Inhibitors/physiology , Protein Biosynthesis/genetics , rhoA GTP-Binding Protein/genetics , Animals , Cells, Cultured , Female , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Male , Rats , rhoA GTP-Binding Protein/biosynthesis
20.
J Immunol ; 189(9): 4379-86, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23002443

ABSTRACT

B lymphopoiesis declines with age, and in rabbits this occurs by 8 wk of age. We found that CFU fibroblasts (CFU-Fs) in the bone marrow (BM) decrease 10-fold by a few weeks of age and that the CFU-Fs preferentially differentiate into adipocytes instead of osteoblasts. BM becomes filled with fat spaces during this time, making rabbit a unique model to study the effects of accelerated fat accumulation on B lymphopoiesis. We show that adipocytes of both rabbit and human secrete a soluble factor(s) that inhibits B lymphopoiesis, and we tested if this inhibition was due to effects on the BM stroma or hematopoietic progenitors. Pretreatment of BM mononuclear cells with adipocyte conditioned medium dramatically inhibited their differentiation into proB cells in cocultures with OP9 stromal cells. In contrast, pretreatment of OP9 stromal cells with adipocyte conditioned medium had no effect on B lymphopoiesis. Using human hematopoietic stem cells, we show that inhibition by the adipocyte-derived factor occurred at the common lymphoid progenitor to preproB cell stage. We propose that the age-related decline in B lymphopoiesis is due to a decrease in CFU-Fs, an increase in adipocytes, and an adipocyte-derived factor that blocks B lymphopoiesis at the common lymphoid progenitor to preproB cell stage.


Subject(s)
Adipocytes/physiology , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Fatty Acid-Binding Proteins/physiology , Growth Inhibitors/physiology , Lymphopoiesis/immunology , Animals , B-Lymphocyte Subsets/metabolism , Cell Line , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Osteoblasts/cytology , Osteoblasts/immunology , Osteoblasts/metabolism , Rabbits , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...