Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Biosensors (Basel) ; 13(7)2023 Jul 17.
Article in English | MEDLINE | ID: mdl-37504139

ABSTRACT

Bacterial infections represent a serious and global threat in modern medicine; thus, it is very important to rapidly detect pathogenic bacteria, such as Escherichia coli (E. coli) O157:H7. Once treatments are delayed after the commencement of symptoms, the patient's health quickly deteriorates. Hence, real-time detection and monitoring of infectious agents are highly critical in early diagnosis for correct treatment and safeguarding public health. To detect these pathogenic bacteria, many approaches have been applied by the biosensors community, for example, widely-used polymerase chain reaction (PCR), enzyme-linked immunosorbent assay (ELISA), culture-based method, and adenosine triphosphate (ATP) bioluminescence. However, these approaches have drawbacks, such as time-consumption, expensive equipment, and being labor-intensive, making it critical to develop ultra-sensitive and highly selective detection. The microfluidic platform based on surface plasmon resonance (SPR), electrochemical sensing, and rolling circle amplification (RCA) offers proper alternatives capable of supplementing the technological gap for pathogen detection. Note that the microfluidic biochip allows to develop rapid, sensitive, portable, and point-of-care (POC) diagnostic tools. This review focuses on recent studies regarding accurate and rapid detection of E. coli O157:H7, with an emphasis on POC methods and devices that complement microfluidic systems. We also examine the efficient whole-body detection by employing antimicrobial peptides (AMPs), which has attracted growing attention in many applications.


Subject(s)
Biosensing Techniques , Escherichia coli O157 , Humans , Microfluidics , Point-of-Care Systems , Biosensing Techniques/methods , Point-of-Care Testing
2.
Exp Mol Med ; 54(11): 1955-1966, 2022 11.
Article in English | MEDLINE | ID: mdl-36376495

ABSTRACT

NANOG plays a key role in cellular plasticity and the acquisition of the stem cell state during reprogramming, but its role in the regenerative process remains unclear. Here, we show that the induction of NANOG in neuronal cells is necessary for the physiological initiation of neuronal regeneration in response to ischemic stress. Specifically, we found that NANOG was preferentially expressed in undifferentiated neuronal cells, and forced expression of Nanog in neural progenitor cells (NPCs) promoted their self-renewing expansion both in ex-vivo slice cultures and in vitro limiting dilution analysis. Notably, the upstream region of the Nanog gene contains sequence motifs for hypoxia-inducible factor-1 alpha (HIF-1α). Therefore, cerebral neurons exposed to hypoxia significantly upregulated NANOG expression selectively in primitive (CD133+) cells, but not in mature cells, leading to the expansion of NPCs. Notably, up to 80% of the neuronal expansion induced by hypoxia was attributed to NANOG-expressing neuronal cells, whereas knockdown during hypoxia abolished this expansion and was accompanied by the downregulation of other pluripotency-related genes. Moreover, the number of NANOG-expressing neuronal cells were transiently increased in response to ischemic insult, predominantly in the infarct area of brain regions undergoing neurogenesis, but not in non-neurogenic loci. Together, these findings reveal a functional effect of NANOG-induction for the initiation of adaptive neuronal regeneration among heterogeneous NPC subsets, pointing to cellular plasticity as a potential link between regeneration and reprogramming processes.


Subject(s)
Nanog Homeobox Protein , Neural Stem Cells , Brain/metabolism , Hypoxia/metabolism , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neural Stem Cells/metabolism , Neurogenesis/genetics , Neurons/metabolism , Animals
3.
Schizophr Res ; 246: 225-234, 2022 08.
Article in English | MEDLINE | ID: mdl-35810486

ABSTRACT

Stem cell technologies have presented explicit evidence of the neurodevelopmental hypothesis of schizophrenia. However, few studies investigated relevance of the schizophrenia genetic liability and the use of genetic reprogramming on pluripotent stem cells to the impaired neurodevelopment shown by stem cells. Therefore, this study sought to investigate the cellular phenotypes of induced neural stem cells (iNSCs) derived without genetic modification from patients with schizophrenia and from genetic high risk (GHR) individuals. Three patients with a diagnosis of schizophrenia, 3 GHR individuals who had two or more relatives with schizophrenia, and 3 healthy volunteers participated. iNSCs were derived using a small molecule-based lineage switch method, and their gene expression levels and migration capabilities were examined. Demographic characteristics were not different among the groups (age, χ2 = 5.637, P = .060; education, χ2 = 2.111, P = .348). All participants stayed well during the follow-up except one GHR individual who developed psychosis 1.5 years later. Migration capacity was impaired in iNSCs from patients with schizophrenia (SZ-iNSCs) compared to iNSCs from GHR individuals or controls (P < .001). iNSCs from a GHR individual who later developed schizophrenia showed migratory impairment that was similar to SZ-iNSCs. Gene expression levels of Sox2 in SZ-iNSCs were significantly lower than those in controls (P = .028). Defective migration in genetically unmodified SZ-iNSCs is the first direct demonstration of neurodevelopmental abnormalities in schizophrenia. Additionally, alterations in gene expression in SZ-iNSCs suggest mechanisms by which genetic liability leads to aberrant neurodevelopment.


Subject(s)
Neural Stem Cells , Psychotic Disorders , Schizophrenia , Humans , Neural Stem Cells/metabolism , Psychotic Disorders/genetics , Psychotic Disorders/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism
4.
Cells ; 11(5)2022 03 04.
Article in English | MEDLINE | ID: mdl-35269514

ABSTRACT

Astrocytes display regenerative potential in pathophysiologic conditions. In our previous study, heme oxygenase-1 (HO-1) promoted astrocytic mitochondrial functions in mice via the peroxisome-proliferator-activating receptor-γ coactivator-1α (PGC-1α) pathway on administering Korean red ginseng extract (KRGE) after traumatic brain injury (TBI). In this study, KRGE promoted astrocytic mitochondrial functions, assessed with oxygen consumption and adenosine triphosphate (ATP) production, which could be regulated by the translocase of the outer membrane of mitochondria 20 (Tom20) pathway with a PGC-1α-independent pathway. The HO-1-Tom20 axis induced an increase in mitochondrial functions, detected with cytochrome c oxidase subunit 2 and cytochrome c. HO-1 crosstalk with nicotinamide phosphoribosyltransferase was concomitant with the upregulated nicotinamide adenine dinucleotide (NAD)/NADH ratio, thereby upregulating NAD-dependent class I sirtuins. In adult neural stem cells (NSCs), KRGE-treated, astrocyte-conditioned media increased oxygen consumption and Tom20 levels through astrocyte-derived HO-1. HO inactivation by Sn(IV) protoporphyrin IX dichloride in TBI mice administered KRGE decreased neuronal markers, together with Tom20. Thus, astrocytic HO-1 induced astrocytic mitochondrial functions. HO-1-related, astrocyte-derived factors may also induce neuronal differentiation and mitochondrial functions of adult NSCs after TBI. KRGE-mediated astrocytic HO-1 induction may have a key role in repairing neurovascular function post-TBI in peri-injured regions by boosting astrocytic and NSC mitochondrial functions.


Subject(s)
Brain Injuries, Traumatic , Neural Stem Cells , Panax , Animals , Astrocytes/metabolism , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Heme Oxygenase-1/metabolism , Mice , Mitochondria/metabolism , NAD/metabolism , Neural Stem Cells/metabolism , Panax/metabolism
5.
Int J Mol Sci ; 22(16)2021 Aug 09.
Article in English | MEDLINE | ID: mdl-34445248

ABSTRACT

The functional neural circuits are partially repaired after an ischemic stroke in the central nervous system (CNS). In the CNS, neurovascular units, including neurons, endothelial cells, astrocytes, pericytes, microglia, and oligodendrocytes maintain homeostasis; however, these cellular networks are damaged after an ischemic stroke. The present review discusses the repair potential of stem cells (i.e., mesenchymal stem cells, endothelial precursor cells, and neural stem cells) and gaseous molecules (i.e., nitric oxide and carbon monoxide) with respect to neuroprotection in the acute phase and regeneration in the late phase after an ischemic stroke. Commonly shared molecular mechanisms in the neurovascular unit are associated with the vascular endothelial growth factor (VEGF) and its related factors. Stem cells and gaseous molecules may exert therapeutic effects by diminishing VEGF-mediated vascular leakage and facilitating VEGF-mediated regenerative capacity. This review presents an in-depth discussion of the regeneration ability by which endogenous neural stem cells and endothelial cells produce neurons and vessels capable of replacing injured neurons and vessels in the CNS.


Subject(s)
Endothelial Cells/metabolism , Ischemic Stroke/metabolism , Neuroglia/metabolism , Neurons/metabolism , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Endothelial Cells/pathology , Humans , Ischemic Stroke/pathology , Neuroglia/pathology , Neurons/pathology , Stem Cells/pathology
6.
Biomaterials ; 275: 120980, 2021 08.
Article in English | MEDLINE | ID: mdl-34198163

ABSTRACT

We expanded the application of endothelin-1 (EDN1) by treating human mesenchymal stem cell (hMSC) organotypic spinal cord slice cultures with EDN1. EDN1-treated hMSCs significantly enhanced neuronal outgrowth. The underlying mechanism of this effect was evaluated via whole-genome methylation. EDN1 increased whole-genome demethylation and euchromatin. To observe demethylation downstream of EDN1, deaminases and glycosylases were screened, and APOBEC1 was found to cause global demethylation and OCT4 gene activation. The sequence of methyl-CpG-binding domain showed similar patterns between EDN1- and APOBEC1-induced demethylation. SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin subfamily A member 4 (SMARC A4) and SMARC subfamily D, member 2 (SMARC D2) were screened via methyl-CpG-binding domain sequencing as a modulator in response to EDN1. Chromatin immunoprecipitation of the H3K9me3, H3K27me3, and H3K4me4 binding sequences on the APOBEC1 promoter was analyzed following treatment with or without siSMARC A4 or siSMARC D2. The results suggested that SMARC A4 and SMARC D2 induced a transition from H3K9me3 to H3K4me3 in the APOBEC1 promoter region following EDN1 treatment. Correlations between EDN1 pathways and therapeutic efficacy in hBM-MSCs were determined in a sciatic nerve injury mouse model. Thus, EDN1 may be a useful novel-concept bioactive peptide and biomaterial component for improving hMSC regenerative capability.


Subject(s)
Mesenchymal Stem Cells , Sciatic Neuropathy , Animals , Bone Marrow , Endothelin-1 , Humans , Mice , Sciatic Nerve
7.
Dement Neurocogn Disord ; 20(1): 1-8, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33552214

ABSTRACT

BACKGROUND AND PURPOSE: Neural stem cells (NSCs) have the ability to regenerate, proliferate, and differentiate, enabling them to play important roles in the recovery of the damaged nervous system. However, in neurodegenerative diseases such as Alzheimer's disease (AD), the NSCs are damaged as well. Glia-like cells from human mesenchymal stem cells (ghMSCs) are functionally enhanced adult stem cells. In the present study, we investigated whether ghMSCs could protect NSCs from amyloid beta (Aß)-mediated toxicity. METHODS: Rat NSCs were obtained from E13-14 fetal rat cortices. NSCs were seeded in pre-coated plates, and the next day, cells were simultaneously treated with 20 µM Aß and 0.4 µm pore insert well-seeded ghMSCs. After 48 hours of co-treatment, cell viability and proliferation were evaluated. After 2 hours of co-treatment, western blotting was performed to measure inflammasome-related factors, such as NOD-like receptor family pyrin domain containing 3, caspase-1, and interleukin-1ß. RESULTS: The results showed that ghMSCs increased viability and proliferation and reduced the toxicity of NSCs injured by Aß by reducing the NRLP3 inflammasome activation of NSCs induced by Aß. CONCLUSIONS: In this study, we confirmed that ghMSCs could protect NSCs in an in vitro model of AD through the regulation of inflammatory response.

8.
Mol Neurobiol ; 56(11): 7617-7630, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31081524

ABSTRACT

Stem cell therapy is considered to be a promising future treatment for intractable neurological diseases, although all the clinical trials using stem cells have not yet shown any good results. Early passage mesenchymal stem cells (MSCs) have been used in most clinical trials because of the issues on safety and efficacy. However, it is not easy to get plenty of cells enough for the treatment and it costs too much. Lots of late passage MSCs can be obtained at lower cost but their efficacy would be a big hurdle for clinical trials. If late passage MSCs with better efficacy could be used in clinical trials, it could be a new and revolutionary solution to reduce cost and enhance easier clinical trials. In the present study, it was investigated whether late passage MSCs could be induced into glia-like cells (ghMSCs); ghMSCs had better efficacy and they protected neurons and the brain from ischemia, and insulin-like growth factor binding protein-4 (IGFBP-4) played a critical role in beneficial effect of ghMSCs. ghMSCs were induced from MSCs and treated in in vitro and in vivo models of ischemia. They effectively protected neurons from ischemia and restored the brain damaged by cerebral infarction. These beneficial effects were significantly blocked by IGFBP-4 antibody. The current study demontsrated that late passage hMSCs can be efficiently induced into ghMSCs with better neuroprotective effect on ischemic stroke. Moreover, the results indicate that IGFBP-4 released from ghMSCs may serve as one of the key neuronal survival factors secreted from ghMSCs.


Subject(s)
Brain Ischemia/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Mesenchymal Stem Cells/metabolism , Neuroglia/metabolism , Neuroprotection , Stroke/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Cerebral Infarction/pathology , Culture Media, Conditioned/pharmacology , Enzyme Activation , Glucose/deficiency , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , Male , Models, Biological , Neurons/metabolism , Oxygen , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Receptor, IGF Type 1/metabolism , bcl-2-Associated X Protein/metabolism
9.
Int J Stem Cells ; 11(2): 177-186, 2018 Nov 30.
Article in English | MEDLINE | ID: mdl-30408408

ABSTRACT

BACKGROUND AND OBJECTIVES: Glial scarring and inflammation after spinal cord injury (SCI) interfere with neural regeneration and functional recovery due to the inhibitory microenvironment of the injured spinal cord. Stem cell transplantation can improve functional recovery in experimental models of SCI, but many obstacles to clinical application remain due to concerns regarding the effectiveness and safety of stem cell transplantation for SCI patients. In this study, we investigated the effects of transplantation of human mesenchymal stem cells (hMSCs) that were genetically modified to express Olig2 in a rat model of SCI. METHODS: Bone marrow-derived hMSCs were genetically modified to express Olig2 and transplanted one week after the induction of contusive SCI in a rat model. Spinal cords were harvested 7 weeks after transplantation. RESULTS: Transplantation of Olig2-expressing hMSCs significantly improved functional recovery in a rat model of contusive SCI model compared to the control hMSC-transplanted group. Transplantation of Olig2-expressing hMSCs also attenuated glial scar formation in spinal cord lesions. Immunohistochemical analysis showed that transplanted Olig2-expressing hMSCs were partially differentiated into Olig1-positive oligodendrocyte-like cells in spinal cords. Furthermore, NF-M-positive axons were more abundant in the Olig2-expressing hMSC-transplanted group than in the control hMSC-transplanted group. CONCLUSIONS: We suggest that Olig2-expressing hMSCs are a safe and optimal cell source for treating SCI.

10.
Int J Mol Med ; 40(6): 1860-1868, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039467

ABSTRACT

Human bone marrow­derived mesenchymal stem cells (hMSCs) are a desirable cell source for cell­based therapy to treat nervous system injuries due to their ability to differentiate into specific cell types. In addition to their multipotency, hMSCs render the tissue microenvironment more favorable for tissue repair by secreting various growth factors. Our previous study demonstrated that hMSCs secrete several growth factors, including several insulin­like growth factor binding proteins (IGFBPs). Among these, IGFBP­6 binds with high affinity and inhibits insulin growth factor­2 (IGF­2) to inhibit the growth of IGF­2­dependent tumors. However, the function of IGFBP­6 in the nervous system remains to be fully elucidated. The present study investigated the protective effects of IGFBP­6 secreted by hMSCs on H2O2­injured primary cortical neuron cultures and lysolecithin­injured organotypic spinal cord slice cultures. Treatment of the H2O2­injured cortical neurons with conditioned media from hMSCs (hMSC­CM) increased the phosphorylation of Akt, reduced cell death and mitochondrial translocation of Bax, and regulated extracellular levels of IGF­1 and IGF­2. MTT assay, western blot analysis and ELISA were used to detect the cell viability and protein expression levels, respectively. An inhibitory antibody against IGFBP­6 eliminated this hMSC­CM­mediated neuroprotective effect in the injured cortical neuron cultures and spinal cord slice cultures. In addition, treatment with cyclolignan picropodophyllin, an inhibitor of IGF­1 receptor (IGF­1R), significantly inhibited neuronal protection by hMSC­CM. These findings demonstrated that hMSC­CM­mediated neuroprotection was attributed to IGF­1R­mediated signaling, potentiated via the inhibition of IGF­2 by IGFBP­6. The results of the present study provide insight into the mechanism by which hMSC administration may promote recovery from nerve injury.


Subject(s)
Insulin-Like Growth Factor Binding Protein 6/genetics , Insulin-Like Growth Factor II/genetics , Mesenchymal Stem Cells/metabolism , Neuroprotection/drug effects , Receptor, IGF Type 1/genetics , Culture Media, Conditioned/pharmacology , Gene Expression/genetics , Humans , Hydrogen Peroxide/toxicity , Lysophosphatidylcholines/toxicity , Neurons/drug effects , Organ Culture Techniques , Podophyllotoxin/administration & dosage , Podophyllotoxin/analogs & derivatives , Primary Cell Culture , Receptor, IGF Type 1/antagonists & inhibitors , Signal Transduction/drug effects , Spinal Cord/metabolism
11.
Sci Rep ; 7(1): 10166, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28860504

ABSTRACT

Cellular reprogramming using small molecules (SMs) without genetic modification provides a promising strategy for generating target cells for cell-based therapy. Human adipose-derived stem cells (hADSCs) are a desirable cell source for clinical application due to their self-renewal capacity, easy obtainability and the lack of safety concerns, such as tumor formation. However, methods to convert hADSCs into neural cells, such as neural stem cells (NSCs), are inefficient, and few if any studies have achieved efficient reprogramming of hADSCs into functional neurons. Here, we developed highly efficient induction protocols to generate NSC-like cells (iNSCs), neuron-like cells (iNs) and GABAergic neuron-like cells (iGNs) from hADSCs via SM-mediated inhibition of SMAD signaling without genetic manipulation. All induced cells adopted morphological, molecular and functional features of their bona fide counterparts. Electrophysiological data demonstrated that iNs and iGNs exhibited electrophysiological properties of neurons and formed neural networks in vitro. Microarray analysis further confirmed that iNSCs and iGNs underwent lineage switch toward a neural fate. Together, these studies provide rapid, reproducible and robust protocols for efficient generation of functional iNSCs, iNs and iGNs from hADSCs, which have utility for modeling disease pathophysiology and providing cell-therapy sources of neurological disorders.


Subject(s)
Adipose Tissue/cytology , GABAergic Neurons/cytology , Neural Stem Cells/cytology , Small Molecule Libraries/pharmacology , Adult , Cell Differentiation , Cell Lineage/drug effects , Cells, Cultured , Cellular Reprogramming/drug effects , Gene Expression Regulation/drug effects , Healthy Volunteers , Humans , Neural Stem Cells/drug effects , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Young Adult
12.
Mol Cancer Ther ; 16(9): 1866-1876, 2017 09.
Article in English | MEDLINE | ID: mdl-28522588

ABSTRACT

Neutropenia is a common adverse event in cancer patients treated with antibody-drug conjugates (ADC) and we aimed to elucidate the potential mechanism of this toxicity. To investigate whether ADCs affect neutrophil production from bone marrow, an in vitro assay was developed in which hematopoietic stem cells (HSC) were differentiated to neutrophils. Several antibodies against targets absent in HSCs and neutrophils were conjugated to MMAE via a cleavable valine-citrulline linker (vcMMAE-ADC) or MMAF via a noncleavable maleimidocaproyl linker (mcMMAF-ADC), and their cytotoxicity was tested in the neutrophil differentiation assay. Results showed that HSCs had similar sensitivity to vcMMAE-ADCs and mcMMAF-ADCs; however, vcMMAE-ADCs were more cytotoxic to differentiating neutrophils than the same antibody conjugated to mcMMAF. This inhibitory effect was not mediated by internalization of ADC either by macropinocytosis or FcγRs. Our results suggested that extracellular proteolysis of the cleavable valine-citrulline linker is responsible for the cytotoxicity to differentiating neutrophils. Mass spectrometry analyses indicated that free MMAE was released from vcMMAE-ADCs in the extracellular compartment when they were incubated with differentiating neutrophils or neutrophil conditioned medium, but not with HSC-conditioned medium. Using different protease inhibitors, our data suggested that serine, but not cysteine proteases, were responsible for the cleavage. In vitro experiments demonstrated that the purified serine protease, elastase, was capable of releasing free MMAE from a vcMMAE-ADC. Here we propose that ADCs containing protease cleavable linkers can contribute to neutropenia via extracellular cleavage mediated by serine proteases secreted by differentiating neutrophils in bone marrow. Mol Cancer Ther; 16(9); 1866-76. ©2017 AACRSee related article by Zhao et al., p. 1877.


Subject(s)
Antineoplastic Agents/adverse effects , Immunoconjugates/adverse effects , Myelopoiesis/drug effects , Neutropenia/blood , Neutropenia/etiology , Neutrophils/drug effects , Animals , Biomarkers , Cell Differentiation/drug effects , Cell Proliferation , Cell Survival/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Mice , Neutrophils/metabolism , Pinocytosis , Receptors, IgG/metabolism , Serine Proteases/metabolism
13.
Int J Mol Med ; 38(4): 1075-82, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27498709

ABSTRACT

Human mesenchymal stem cells (hMSCs) have great therapeutic potential due to their high plasticity, immune privileged status and ease of preparation, as well as a lack of ethical barriers to their use. However, their ultimate usefulness is limited by cellular senescence occurring secondary to increased cellular levels of reactive oxygen species (ROS) during their propagation in culture. The underlying molecular mechanisms responsible for this process in hMSCs remain unclear. An antioxidant polyphenol epigallocatechin-3-gallate (EGCG) found in green tea, is known to activate nuclear factor-erythroid 2-related factor 2 (Nrf2), a master transcriptional regulator of antioxidant genes. Herein, we examined the EGCG-mediated antioxidant mechanism in hMSCs exposed to ROS which involves Nrf2 activation. The H2O2-exposed hMSCs showed cellular senescence with significantly increased protein levels of acetyl-p53 and p21 in comparison with the untreated hMSCs, and these effects were prevented by pre-treatment with EGCG. By contrast, in Nrf2-knockdown hMSCs, EGCG lost its antioxidant effect, exhibiting high levels of acetyl-p53 and p21 following EGCG pre-treatment and H2O2 exposure. This indicates that Nrf2 and p53/p21 may be involved in the anti­senescent effect of EGCG in hMSCs. Taken together, these findings indicate the important role of EGCG in preventing oxidative stress-induced cellular senescence in hMSCs through Nrf2 activation, which has applications for the massive production of more suitable hMSCs for cell-based therapy.


Subject(s)
Catechin/analogs & derivatives , Cellular Senescence/drug effects , Mesenchymal Stem Cells/pathology , Oxidative Stress/drug effects , Acetylation/drug effects , Adult , Catechin/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Hydrogen Peroxide/toxicity , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Models, Biological , NF-E2-Related Factor 2/metabolism , Protein Transport/drug effects , Tumor Suppressor Protein p53/metabolism
14.
Tissue Eng Regen Med ; 13(5): 601-609, 2016 Oct.
Article in English | MEDLINE | ID: mdl-30603441

ABSTRACT

Enhancing adult nerve regeneration is a potential therapeutic strategy for treating spinal cord injury. Vascular endothelial growth factor (VEGF) is a major contributor to angiogenesis, which can reduce the spinal cord injury by inhibiting the inflammation and improve recovery after spinal cord injury. We have previously demonstrated that exogenous VEGF has neurotrophic effects on injured spinal nerves in organotypic spinal cord slice cultures. However, the mechanisms underlying the neurite growth by exogenous VEGF remain to be explored in spinal cord. In this study, we found out that exogenous VEGF mediated axonal outgrowth through VEGF receptor 1 (VEGFR1) and VEGFR2, both of which were expressed on organotypic spinal cord slices. Although VEGFR1 and VEGFR2 were constitutively expressed in some cells of control spinal cord slices, VEGF treatment upregulated expression of VEGFR1 and VEGFR2. Both VEGFR1 and VEGFR2 were expressed in neuronal cells as well as glial cells of organotypic spinal cord slices. We also observed that VEGF-induced axonal outgrowth was attenuated by a specific mitogen-activated protein kinase (MAPK) inhibitor PD98059 and a specific phosphoinositide 3-kinase (PI3K) inhibitor wortmannin. Thus, these findings suggest that these MAPK and PI3K pathways have important roles in regulating VEGF-induced axonal outgrowth in the postnatal spinal cord.

15.
Analyst ; 140(23): 7997-8006, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26524182

ABSTRACT

The ability of antimicrobial peptides (AMPs) for effective binding to multiple target microbes has drawn lots of attention as an alternative to antibodies for detecting whole bacteria. We investigated pathogenic Escherichia coli (E. coli) detection by applying a microfluidic based biosensing device embedded with AMP-labeled beads. According to a new channel design, our device is reusable by the repeated operation of detection and regeneration modes, and the binding rate is more enhanced due to even distribution of the bacterial suspension inside the chamber by implementing influx side channels. We observed higher binding affinity of pathogenic E. coli O157:H7 for AMP-labeled beads than nonpathogenic E. coli DH5α, and the fluorescence intensity of pathogenic E. coli was about 3.4 times higher than the nonpathogenic one. The flow rate of bacterial suspension should be applied above a certain level for stronger binding and rapid detection by attaining a saturation level of detection within a short time of less than 20 min. A possible improvement in the limit of detection in the level of 10 cells per mL for E. coli O157:H7 implies that the AMP-labeled beads have high potential for the sensitive detection of pathogenic E. coli at an appropriate flow rate.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Bacteriological Techniques/methods , Escherichia coli O157/isolation & purification , Lab-On-A-Chip Devices , Antimicrobial Cationic Peptides/chemistry , Limit of Detection , Microspheres
16.
Analyst ; 140(4): 1265-74, 2015 Feb 21.
Article in English | MEDLINE | ID: mdl-25555081

ABSTRACT

Human bone marrow-derived mesenchymal stem cells (hMSCs) consist of heterogeneous subpopulations with different multipotent properties: small and large cells with high and low multipotency, respectively. Accordingly, sorting out a target subpopulation from the others is very important to increase the effectiveness of cell-based therapy. We performed flow-based sorting of hMSCs by using optimally designed microfluidic chips based on the hydrodynamic filtration (HDF) principle. The chip was designed with the parameters rigorously determined by the complete analysis of laminar flow for flow fraction and complicated networks of main and multi-branched channels for hMSCs sorting into three subpopulations: small (<25 µm), medium (25-40 µm), and large (>40 µm) cells. By focusing with a proper ratio between main and side flows, cells migrate toward the sidewall due to a virtual boundary of fluid layers and enter the branch channels. This opens the possibility of sorting stem cells rapidly without damage. Over 86% recovery was achieved for each population of cells with complete purity in small cells, but the sorting efficiency of cells is slightly lower than that of rigid model particles, due to the effect of cell deformation. Finally, we confirmed that our method could successfully fractionate the three subpopulations of hMSCs by analyzing the surface marker expressions of cells from each outlet.


Subject(s)
Cell Separation/instrumentation , Mesenchymal Stem Cells/cytology , Microfluidic Analytical Techniques/instrumentation , Adult , Cell Line , Equipment Design , Filtration/instrumentation , Humans , Hydrodynamics
17.
Mol Cells ; 37(12): 881-7, 2014 Dec 31.
Article in English | MEDLINE | ID: mdl-25410904

ABSTRACT

Cell proliferation is tightly controlled by the cell-cycle regulatory proteins, primarily by cyclins and cyclin-dependent kinases (CDKs) in the G1 phase. The ankyrin repeat-rich membrane spanning (ARMS) scaffold protein, also known as kinase D-interacting substrate of 220 kDa (Kidins 220), has been previously identified as a prominent downstream target of neurotrophin and ephrin receptors. Many studies have reported that ARMS/Kidins220 acts as a major signaling platform in organizing the signaling complex to regulate various cellular responses in the nervous and vascular systems. However, the role of ARMS/Kidins220 in cell proliferation and cell-cycle progression has never been investigated. Here we report that knockdown of ARMS/Kidins220 inhibits mouse neuroblastoma cell proliferation by inducing slowdown of cell cycle in the G1 phase. This effect is mediated by the upregulation of a CDK inhibitor p21, which causes the decrease in cyclin D1 and CDK4 protein levels and subsequent reduction of pRb hyperphosphorylation. Our results suggest a new role of ARMS/Kidins220 as a signaling platform to regulate tumor cell proliferation in response to the extracellular stimuli.


Subject(s)
Cell Proliferation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Animals , Cell Line, Tumor , Cyclin D1 , Cyclin-Dependent Kinase 4 , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Mice , Phosphorylation , Signal Transduction
18.
PLoS One ; 9(11): e111515, 2014.
Article in English | MEDLINE | ID: mdl-25369402

ABSTRACT

Discoidin domain receptor 1 (DDR1) is a member of the receptor tyrosine kinase family. The receptor is activated upon binding to its ligand, collagen, and plays a crucial role in many fundamental processes such as cell differentiation, adhesion, migration and invasion. Although DDR1 is expressed in many normal tissues, upregulated expression of DDR1 in a variety of human cancers such as lung, colon and brain cancers is known to be associated with poor prognosis. Using shRNA silencing, we assessed the oncogenic potential of DDR1. DDR1 knockdown impaired tumor cell proliferation and migration in vitro and tumor growth in vivo. Microarray analysis of tumor cells demonstrated upregulation of TGFBI expression upon DDR1 knockdown, which was subsequently confirmed at the protein level. TGFBI is a TGFß-induced extracellular matrix protein secreted by the tumor cells and is known to act either as a tumor promoter or tumor suppressor, depending on the tumor environment. Here, we show that exogenous addition of recombinant TGFBI to BXPC3 tumor cells inhibited clonogenic growth and migration, thus recapitulating the phenotypic effect observed from DDR1 silencing. BXPC3 tumor xenografts demonstrated reduced growth with DDR1 knockdown, and the same xenograft tumors exhibited an increase in TGFBI expression level. Together, these data suggest that DDR1 expression level influences tumor growth in part via modulation of TGFBI expression. The reciprocal expression of DDR1 and TGFBI may help to elucidate the contribution of DDR1 in tumorigenesis and TGFBI may also be used as a biomarker for the therapeutic development of DDR1 specific inhibitors.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Transforming Growth Factor beta/genetics , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Discoidin Domain Receptors , Humans , Mice, SCID , RNA Interference , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Signal Transduction
19.
Proc Natl Acad Sci U S A ; 111(21): E2219-28, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24821775

ABSTRACT

The unfolded protein response (UPR) pathway, a stress-induced signaling cascade emanating from the endoplasmic reticulum (ER), regulates the expression and activity of molecules including BiP (HSPA5), IRE1 (ERN1), Blimp-1 (PRDM1), and X-box binding protein 1 (XBP1). These molecules are required for terminal differentiation of B cells into plasma cells and expressed at high levels in plasma cell-derived multiple myeloma. Although these molecules have no known role at early stages of B-cell development, here we show that their expression transiently peaks at the pre-B-cell receptor checkpoint. Inducible, Cre-mediated deletion of Hspa5, Prdm1, and Xbp1 consistently induces cellular stress and cell death in normal pre-B cells and in pre-B-cell acute lymphoblastic leukemia (ALL) driven by BCR-ABL1- and NRAS(G12D) oncogenes. Mechanistically, expression and activity of the UPR downstream effector XBP1 is regulated positively by STAT5 and negatively by the B-cell-specific transcriptional repressors BACH2 and BCL6. In two clinical trials for children and adults with ALL, high XBP1 mRNA levels at the time of diagnosis predicted poor outcome. A small molecule inhibitor of ERN1-mediated XBP1 activation induced selective cell death of patient-derived pre-B ALL cells in vitro and significantly prolonged survival of transplant recipient mice in vivo. Collectively, these studies reveal that pre-B ALL cells are uniquely vulnerable to ER stress and identify the UPR pathway and its downstream effector XBP1 as novel therapeutic targets to overcome drug resistance in pre-B ALL.


Subject(s)
B-Lymphocytes/physiology , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/metabolism , Gene Expression Regulation/drug effects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Unfolded Protein Response/drug effects , Adult , Animals , B-Lymphocytes/metabolism , Base Sequence , Basic-Leucine Zipper Transcription Factors/pharmacology , Blotting, Western , Cell Differentiation/physiology , Child , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/pharmacology , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/genetics , Flow Cytometry , Gene Deletion , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heterografts , Humans , Kaplan-Meier Estimate , Mice , Microarray Analysis , Molecular Sequence Data , Positive Regulatory Domain I-Binding Factor 1 , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-bcl-6 , Real-Time Polymerase Chain Reaction , Regulatory Factor X Transcription Factors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Analysis, RNA , Transcription Factors/genetics , Unfolded Protein Response/physiology , X-Box Binding Protein 1 , beta-Galactosidase
20.
Neoplasia ; 15(10): 1138-50, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204193

ABSTRACT

Preclinical evaluation of novel cancer agents requires models that accurately reflect the biology and molecular characteristics of human tumors. Molecular profiles of eight pancreatic ductal adenocarcinoma patient tumors were compared to corresponding passages of xenografts obtained by grafting tumor fragments into immunocompromised mice. Molecular characterization was performed by copy number analysis, gene expression and microRNA microarrays, mutation analysis, short tandem repeat (STR) profiling, and immunohistochemistry. Xenografts were found to be highly representative of their respective tumors, with a high degree of genetic stability observed by STR profiling and mutation analysis. Copy number variation (CNV) profiles of early and late xenograft passages were similar, with recurrent losses on chromosomes 1p, 3p, 4q, 6, 8p, 9, 10, 11q, 12p, 15q, 17, 18, 20p, and 21 and gains on 1q, 5p, 8q, 11q, 12q, 13q, 19q, and 20q. Pearson correlations of gene expression profiles of tumors and xenograft passages were above 0.88 for all models. Gene expression patterns between early and late passage xenografts were highly stable for each individual model. Changes observed in xenograft passages largely corresponded to human stromal compartment genes and inflammatory processes. While some differences exist between the primary tumors and corresponding xenografts, the molecular profiles remain stable after extensive passaging. Evidence for stability in molecular characteristics after several rounds of passaging lends confidence to clinical relevance and allows for expansion of models to generate the requisite number of animals required for cohorts used in drug screening and development studies.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Neoplasms, Experimental/metabolism , Pancreatic Neoplasms/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , DNA Copy Number Variations , Drug Evaluation, Preclinical , Gene Expression Profiling , Heterografts , Humans , Mice, SCID , MicroRNAs/genetics , Mutation , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Pancreatic Neoplasms/genetics , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL