Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
World J Clin Cases ; 10(15): 4911-4916, 2022 May 26.
Article in English | MEDLINE | ID: mdl-35801030

ABSTRACT

BACKGROUND: The indwelling nasogastric tube is commonly used for supplying enteral nutrition to patients who are unable to feed themselves, and accurate positioning is essential in the indwelling nasogastric tube in the body of the aforementioned patients. In clinical practice, abdominal radiography, auscultation, and clinical determination of the pH of the gastric juice are routinely used by medical personnel to determine the position of the tube; however, those treatments have proved limitations in specific cases. There are few case reports on the precise positioning of the nasogastric tube in patients with coronavirus disease 2019 (COVID-19), for whom a supply of necessary nutrition support is significant throughout the process of treatment. CASE SUMMARY: A 79-year-old patient, diagnosed with COVID-19 at the stage of combined syndromes with severe bacterial lung infection, respiratory failure, multiple co-morbidities, and a poor nutritional status, was presented to us and required an indwelling nasogastric tube for enteral nutrition support. After pre-treatment assessments including observation of the patient's nasal feeding status and examination of the nasal septal deviation, inflammation, obstruction, nasal leakage of cerebrospinal fluid, and other disorders that might render intubation inappropriate, we measured and marked the length of the nasogastric tube to be placed and delivered the tube to the intended length in the standard manner. Then further scrutiny was conducted to ensure that the tube was not coiled in the mouth, and gentle movements were made to avoid damage to the esophageal mucosa. However, back draw of the gastric juice using an empty needle failed, and the stethoscope could not be used for auscultation due to the specific condition presented by the internal organs of the patient, and the end of the tube was placed in saline with no bubbles spilling out. Therefore, it was not possible to determine whether the nasogastric tube was placed exactly in the stomach and no nutrient infusion was performed for the time being. Subsequently, the ultrasound probe was utilized to view the condition of the patient's stomach, where the nasogastric tube was found to be translucent and running parallel to the esophagus shaped as "=". The pre-conditions were achieved and 100 mL nutritional fluid was fed to the patient, who did not experience any discomfort throughout the procedure. His vital signs were stable with no adverse effects. CONCLUSION: We achieved successfully used ultrasound to position the nasogastric tube in a 79-year-old patient with COVID-19. The repeatable ultrasound application does not involve radiation and causes less disturbance in the neck, making it advantageous for rapid positioning of the nasogastric tube and worthy of clinical promotion and application.

2.
Invest Ophthalmol Vis Sci ; 61(12): 21, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33085740

ABSTRACT

Purpose: Yap1 encodes an evolutionarily conserved transcriptional coactivator and functions as a down-stream effector of the Hippo signaling pathway that controls tissue size and cell growth. Yap1 contributes to lens epithelial development. However, the effect of Yap1 haplodeficiency on the lens epithelium and its role in the development of cataracts has not been reported. The aim of the current study is to investigate Yap1 function and its regulatory mechanisms in lens epithelial cells (LECs). Methods: Lens phenotypes were investigated in Yap1 heterozygous mutant mice by visual observation and histological and biochemical methods. Primary LEC cultures were used to study regulatory molecular mechanism. Results: The heterozygous inactivation of Yap1 in mice caused cataracts during adulthood with defective LEC phenotypes. Despite a normal early development of the eye including the lens, the majority of Yap1 heterozygotes developed cataracts in the first six months of age. Cataract was preceded by multiple morphological defects in the lens epithelium, including decreased cell density and abnormal cell junctions. The low LEC density was coincident with reduced LEC proliferation. In addition, expression of the Yap1 target gene Crim1 was reduced in the Yap1+/- LEC, and overexpression of Crim1 restored Yap1+/- LEC cell proliferation in vitro. Conclusions: Homozygosity of the Yap1 gene was critical for adequate Crim1 expression needed to maintain the constant proliferation of LEC and to maintain a normal-sized lens. Yap1 haplodeficiency leads to cataracts.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cataract/physiopathology , Cell Cycle Proteins/physiology , Epithelial Cells/metabolism , Animals , Blotting, Western , Bone Morphogenetic Protein Receptors/metabolism , Bromodeoxyuridine/metabolism , Cataract/metabolism , Cell Count , Cell Proliferation/physiology , Cells, Cultured , Disease Progression , Epithelial Cells/pathology , Gene Expression Regulation, Developmental/physiology , Heterozygote , In Situ Nick-End Labeling , Lens, Crystalline/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Organogenesis , Real-Time Polymerase Chain Reaction , YAP-Signaling Proteins
3.
FASEB J ; 34(5): 6757-6768, 2020 05.
Article in English | MEDLINE | ID: mdl-32223016

ABSTRACT

Nuclear YAP1 plays a critical role in regulation of stem cell proliferation, tissue regeneration, and organ size in many types of epithelia. Due to rapid turnover of most epithelial cell types, the cytoplasmic function of YAP1 in epithelial cells has not been well studied. The retinal pigment epithelium (RPE) is a highly polarized epithelial cell type maintained at a senescence state, and offers an ideal cell model to study the active role of YAP1 in maintenance of the adult epithelial phenotype. Here, we show that the cytoplasmic function of YAP1 is essential to maintain adult RPE differentiation. Knockout of Yap1 in the adult mouse RPE caused cell depolarization and tight junction breakdown, and led to inhibition of RPE65 expression, diminishment of RPE pigments, and retraction of microvilli and basal infoldings. These changes in RPE further prompted the loss of adjacent photoreceptor outer segments and photoreceptor death, which eventually led to decline of visual function in older mice between 6 and 12 months of age. Furthermore, nuclear ß-catenin and its activity were significantly increased in mutant RPE. These results suggest that YAP1 plays an important role in active inhibition of Wnt/ß-catenin signaling, and is essential for downregulation of ß-catenin nuclear activity and prevention of dedifferentiation of adult RPE.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bestrophins/physiology , Cell Cycle Proteins/metabolism , Cell Differentiation , Retinal Pigment Epithelium/cytology , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cell Proliferation , Mice , Mice, Knockout , Retinal Pigment Epithelium/metabolism , YAP-Signaling Proteins
4.
Exp Eye Res ; 191: 107899, 2020 02.
Article in English | MEDLINE | ID: mdl-31866431

ABSTRACT

Retinal detachment (RD) induces ischemia and oxygen deficiency in the retina and results in multiple pathological events; photoreceptor cell degeneration and death is the eventual cause of vision decline. In this study, we investigated the therapeutic effects of mesenchymal stem cell-derived exosomes (MSC-Exos) in a rat retinal detachment (RD) model. The model was developed using a subretinal injection of 1% hyaluronic acid in male Sprague-Dawley rats. MSC-Exos were sub-retinally injected at the time of retinal separation to study their therapeutic function. The retinal expression levels of inflammatory cytokines TNF-α, IL-1ß, and MCP-1 were detected by RT-PCR, the autophagy-related protein 5 (Atg5) and microtubule-associated protein 1 light chain 3 beta (LC3) were detected by Western blot, and apoptosis was examined using TUNEL assays at 3 days following RD. Retinal structure was observed at 7 days post-RD. Proteomic analysis was also performed to detect proteins carried by MSC-Exos using iTRAQ-based technology combined with one-dimensional nano LC-nano-ESI- MS/MS. We found that expression of TNF-α and IL-1ß were significantly reduced, the LC3-II to LC3-I ratio was enhanced and cleavage of Atg5 was decreased after MSC-Exo treatment. Treatment with MSC-Exos also suppressed photoreceptor cell apoptosis and maintained normal retinal structure when compared to control groups. Proteomic analysis revealed that MSC-Exos contained proteins with anti-inflammatory, neuroprotective and anti-apoptotic effects. These results suggest that MSC-Exos have therapeutic effects on RD-induced retinal injury and can be used to reduce effects of retinal detachment on photoreceptor cell degeneration in patients.


Subject(s)
Exosomes/physiology , Mesenchymal Stem Cells/cytology , Retinal Detachment/therapy , Animals , Apoptosis/physiology , Autophagy-Related Protein 5/metabolism , Blotting, Western , Chemokine CCL2/genetics , Disease Models, Animal , Gene Expression Regulation/physiology , In Situ Nick-End Labeling , Injections, Intraocular , Interleukin-1beta/genetics , Male , Microtubule-Associated Proteins/metabolism , Photoreceptor Cells/pathology , Proteomics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Retinal Detachment/metabolism , Retinal Detachment/pathology , Tandem Mass Spectrometry , Tumor Necrosis Factor-alpha/genetics
5.
Sci Rep ; 6: 35202, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27734924

ABSTRACT

Key issues in corneal epithelium biology are the mechanism for corneal epithelium stem cells to maintain the corneal epithelial homeostasis and wound healing responses, and what are the regulatory molecular pathways involved. There are apparent discrepancies about the locations of the progenitor populations responsible for corneal epithelial self-renewal. We have developed a genetic mouse model to trace the corneal epithelial progenitor lineages during adult corneal epithelial homeostasis and wound healing response. Our data revealed that the early corneal epithelial progenitor cells expressing keratin-12 originated from limbus, and gave rise to the transit amplifying cells that migrated centripetally to differentiate into corneal epithelial cells. Our results support a model that both corneal epithelial homeostasis and wound healing are mainly maintained by the activated limbal stem cells originating form limbus, but not from the corneal basal epithelial layer. In the present study, we further demonstrated the nuclear expression of transcriptional coactivator YAP1 in the limbal and corneal basal epithelial cells and its essential role for maintaining the high proliferative potential of those corneal epithelial progenitor cells in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Lineage , Epithelium, Corneal/cytology , Keratin-12/metabolism , Phosphoproteins/metabolism , Stem Cells/cytology , Animals , Cell Cycle Proteins , Epithelium, Corneal/metabolism , Green Fluorescent Proteins/genetics , Mice , Mice, Transgenic , Stem Cells/metabolism , Wound Healing , YAP-Signaling Proteins
6.
Sci Rep ; 6: 34562, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-27686625

ABSTRACT

Although accumulated evidence supports the notion that mesenchymal stem cells (MSCs) act in a paracrine manner, the mechanisms are still not fully understood. Recently, MSC-derived exosomes (MSC-Exos), a type of microvesicle released from MSCs, were thought to carry functional proteins and RNAs to recipient cells and play therapeutic roles. In the present study, we intravitreally injected MSCs derived from either mouse adipose tissue or human umbilical cord, and their exosomes to observe and compare their functions in a mouse model of laser-induced retinal injury. We found that both MSCs and their exosomes reduced damage, inhibited apoptosis, and suppressed inflammatory responses to obtain better visual function to nearly the same extent in vivo. Obvious down-regulation of monocyte chemotactic protein (MCP)-1 in the retina was found after MSC-Exos injection. In vitro, MSC-Exos also down-regulated MCP-1 mRNA expression in primarily cultured retinal cells after thermal injury. It was further demonstrated that intravitreal injection of an MCP-1-neutralizing antibody promoted the recovery of retinal laser injury, whereas the therapeutic effect of exosomes was abolished when MSC-Exos and MCP-1 were administrated simultaneously. Collectively, these results suggest that MSC-Exos ameliorate laser-induced retinal injury partially through down-regulation of MCP-1.

7.
PLoS One ; 10(3): e0121185, 2015.
Article in English | MEDLINE | ID: mdl-25803615

ABSTRACT

The inhibition of NF-κB by genetic deletion or pharmacological inhibition of IKK2 significantly reduces laser-induced choroid neovascularization (CNV). To achieve a sustained and controlled intraocular release of a selective and potent IKK2 inhibitor, 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA-1) (MW: 279.29), we developed a biodegradable poly-lactide-co-glycolide (PLGA) polymer-delivery system to further investigate the anti-neovascularization effects of IKK2 inhibition and in vivo biosafety using laser-induced CNV mouse model. The solvent-evaporation method produced spherical TPCA-1-loaded PLGA microparticles characterized with a mean diameter of 2.4 »m and loading efficiency of 80%. Retrobulbar administration of the TPCA-1-loaded PLGA microparticles maintained a sustained drug level in the retina during the study period. No detectable TPCA-1 level was observed in the untreated contralateral eye. The anti-CNV effect of retrobulbarly administrated TPCA-1-loaded PLGA microparticles was assessed by retinal fluorescein leakage and isolectin staining methods, showing significantly reduced CNV development on day 7 after laser injury. Macrophage infiltration into the laser lesion was attenuated as assayed by choroid/RPE flat-mount staining with anti-F4/80 antibody. Consistently, laser induced expressions of Vegfa and Ccl2 were inhibited by the TPCA-1-loaded PLGA treatment. This TPCA-1 delivery system did not cause any noticeable cellular or functional toxicity to the treated eyes as evaluated by histology and optokinetic reflex (OKR) tests; and no systemic toxicity was observed. We conclude that retrobulbar injection of the small-molecule IKK2 inhibitor TPCA-1, delivered by biodegradable PLGA microparticles, can achieve a sustained and controllable drug release into choroid/retina and attenuate laser-induced CNV development without causing apparent systemic toxicity. Our results suggest a potential clinical application of TPCA-1 delivered by microparticles in treatment of CNV in the patients with age-related macular degeneration and other retinal neovascularization diseases.


Subject(s)
Amides/administration & dosage , Choroidal Neovascularization/drug therapy , I-kappa B Kinase/antagonists & inhibitors , Lactic Acid/administration & dosage , Macrophages/cytology , Macrophages/drug effects , Polyglycolic Acid/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Thiophenes/administration & dosage , Amides/chemistry , Animals , Choroidal Neovascularization/pathology , Disease Models, Animal , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Female , Lactic Acid/chemistry , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Protein Kinase Inhibitors/chemistry , Thiophenes/chemistry
8.
Cell Tissue Res ; 360(2): 233-43, 2015 May.
Article in English | MEDLINE | ID: mdl-25638408

ABSTRACT

Pathological retinal neovascularization, including retinopathy of prematurity and age-related macular degeneration, is the most common cause of blindness worldwide. Insulin-like growth factor-1 (IGF-1) has a direct mitogenic effect on endothelial cells, which is the basis of angiogenesis. Vascular endothelial growth factor (VEGF) activation in response to IGF-1 is well documented; however, the molecular mechanisms responsible for the termination of IGF-1 signaling are still not completely elucidated. Here, we show that the polypyrimidine tract-binding protein-associated splicing factor (PSF) is a potential negative regulator of VEGF expression induced by IGF stimulation. Functional analysis demonstrated that ectopic expression of PSF inhibits IGF-1-stimulated transcriptional activation and mRNA expression of the VEGF gene, whereas knockdown of PSF increased IGF-1-stimulated responses. PSF recruited Hakai to the VEGF transcription complex, resulting in inhibition of IGF-1-mediated transcription. Transfection with Hakai siRNA reversed the PSF-mediated transcriptional repression of VEGF gene transcription. In summary, these results show that PSF can repress the transcriptional activation of VEGF stimulated by IGF-1 via recruitment of the Hakai complex and delineate a novel regulatory mechanism of IGF-1/VEGF signaling that may have implications in the pathogenesis of neovascularization in ocular diseases.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Oxygen/adverse effects , RNA-Binding Proteins/metabolism , Retinal Neovascularization/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Animals , Disease Models, Animal , Female , Insulin-Like Growth Factor I/genetics , Male , Mice , Oxygen/pharmacology , PTB-Associated Splicing Factor , RNA-Binding Proteins/genetics , Retinal Neovascularization/chemically induced , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics
9.
J Invest Dermatol ; 135(6): 1621-1628, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25668240

ABSTRACT

The homozygous repeated epilation (Er/Er) mouse mutant of the gene encoding 14-3-3σ displays an epidermal phenotype characterized by hyperproliferative keratinocytes and undifferentiated epidermis. Heterozygous Er/+ mice develop spontaneous skin tumors and are highly sensitive to tumor-promoting 7,12-dimethylbenzanthracene/12-O-tetradecanoyl-phorbol-13-acetate induction. The molecular mechanisms underlying 14-3-3σ regulation of epidermal proliferation, differentiation, and tumor formation have not been well elucidated. In this study, we found that Er/Er keratinocytes failed to sequester Yap1 in the cytoplasm, leading to its nuclear localization during epidermal development in vivo and under differentiation-inducing culture conditions in vitro. In addition, enhanced Yap1 nuclear localization was also evident in 7,12-dimethylbenzanthracene/12-O-tetradecanoyl-phorbol-13-acetate-induced tumors from Er/+ skin. Furthermore, short hairpin RNA (shRNA) knockdown of Yap1 expression in Er/Er keratinocytes inhibited their proliferation, suggesting that YAP1 functions as a downstream effector of 14-3-3σ controlling epidermal proliferation. We then demonstrated that keratinocytes express all seven 14-3-3 protein isoforms, some of which form heterodimers with 14-3-3σ, either full-length wild type (WT) or the mutant form found in Er/Er mice. However, Er 14-3-3σ does not interact with Yap1, as demonstrated by coimmunoprecipitation. We conclude that Er 14-3-3σ disrupts the interaction between 14-3-3 and Yap1, and thus fails to block Yap1 nuclear transcriptional function, causing continued progenitor expansion and inhibition of differentiation in the Er/Er epidermis.


Subject(s)
14-3-3 Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Keratinocytes/metabolism , Phosphoproteins/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Active Transport, Cell Nucleus , Animals , Cell Cycle Proteins , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Cytoplasm/metabolism , Epidermis/metabolism , Gene Expression Regulation , Heterozygote , Homozygote , Keratinocytes/cytology , Lentivirus/genetics , Mice , Phenotype , Protein Isoforms/metabolism , RNA, Small Interfering/metabolism , Skin/metabolism , Tetradecanoylphorbol Acetate , YAP-Signaling Proteins
10.
Proc Natl Acad Sci U S A ; 112(4): 1065-70, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25583514

ABSTRACT

The Hippo and c-Jun N-terminal kinase (JNK) pathway both regulate growth and contribute to tumorigenesis when dysregulated. Whereas the Hippo pathway acts via the transcription coactivator Yki/YAP to regulate target gene expression, JNK signaling, triggered by various modulators including Rho GTPases, activates the transcription factors Jun and Fos. Here, we show that impaired Hippo signaling induces JNK activation through Rho1. Blocking Rho1-JNK signaling suppresses Yki-induced overgrowth in the wing disk, whereas ectopic Rho1 expression promotes tissue growth when apoptosis is prohibited. Furthermore, Yki directly regulates Rho1 transcription via the transcription factor Sd. Thus, our results have identified a novel molecular link between the Hippo and JNK pathways and implicated the essential role of the JNK pathway in Hippo signaling-related tumorigenesis.


Subject(s)
Drosophila Proteins/metabolism , Imaginal Discs/embryology , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/physiology , Protein Serine-Threonine Kinases/metabolism , Wings, Animal/embryology , rho GTP-Binding Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Imaginal Discs/cytology , Intracellular Signaling Peptides and Proteins/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic/physiology , Wings, Animal/cytology , YAP-Signaling Proteins , rho GTP-Binding Proteins/genetics
11.
Metab Brain Dis ; 30(4): 943-50, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25604732

ABSTRACT

Phagocytic clearance of the spent photoreceptor outer segments (OS) by RPE cells is regulated by circadian rhythm cycle and is essential for photoreceptor integrity and function. Mertk regulates RPE phagocytosis and a deficiency in Mertk causes photoreceptor degeneration and visual loss. This study aimed to investigate Mertk regulation of the microRNAs (miRNA), potentially regulating expression of their target genes, which affect phagocytosis. The differentially expressed miRNAs were identified using miRCURY(TM) microRNA Arrays from total RNA isolated at 0900 h and 1900 h from the mechanically dissociated RPE sheets of the WT and Mertk (-/-) mice, which were housed in a 12-h light-dark cycle with the lighting onset at 0700 h (7:00am). Validation of the differentially expressed miRNAs and assessment of the putative miRNA target gene expression were performed by real-time PCR. Among the differentially expressed miRNAs in the Mertk (-/-) RPE, seven miRNAs were up-regulated and 13 were down-regulated in the morning groups. Similarly, 24 miRNAs were found to be up-regulated and 13 were down-regulated in the evening groups. To search for those that may participate in regulating expression of cytoskeletal proteins, we examined the predicted target genes that might participate in phagocytosis were examined by real-time PCR. Of nine potential altered targets, four deregulated genes were myosin subunits. Notably, multiple members of the 21 up-regulated miRNAs can theoretically recognize these down-regulated mRNAs, particularly MyH14 and Myl3. This study shows that loss of Mertk alters miRNA expression, which in turn affects expression of the downstream target genes, potentially affecting phagocytosis.


Subject(s)
MicroRNAs/biosynthesis , Proto-Oncogene Proteins/deficiency , Receptor Protein-Tyrosine Kinases/deficiency , Retinal Pigment Epithelium/metabolism , Animals , Cells, Cultured , Gene Expression Regulation , Mice , Mice, Knockout , MicroRNAs/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , c-Mer Tyrosine Kinase
12.
Metab Brain Dis ; 30(3): 633-44, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25487541

ABSTRACT

The Tyro3, Axl and Mertk (TAM) subfamily of receptor protein tyrosine kinases functions in cell growth, differentiation, survival, and most recently found, in the regulation of immune responses and phagocytosis. All three receptors and their ligands, Gas6 (growth arrest-specific gene 6) and protein S, are expressed in the central nervous system (CNS). TAM receptors play pivotal roles in adult hippocampal neurogenesis. Loss of these receptors causes a comprised neurogenesis in the dentate gyrus of adult hippocampus. TAM receptors have a negative regulatory effect on microglia and peripheral antigen-presenting cells, and play a critical role in preventing overproduction of pro-inflammatory cytokines detrimental to the proliferation, differentiation, and survival of adult neuronal stem cells (NSCs). Besides, these receptors also play an intrinsic trophic function in supporting NSC survival, proliferation, and differentiation into immature neurons. All these events collectively ensure a sustained neurogenesis in adult hippocampus.


Subject(s)
Hippocampus/cytology , Hippocampus/metabolism , Neurogenesis/physiology , Receptor Protein-Tyrosine Kinases/deficiency , Age Factors , Animals , Humans , Signal Transduction/physiology
13.
PLoS One ; 9(12): e115140, 2014.
Article in English | MEDLINE | ID: mdl-25514676

ABSTRACT

Tyro3, Axl and Mertk (TAM) receptor tyrosine kinases play multiple functional roles by either providing intrinsic trophic support for cell growth or regulating the expression of target genes that are important in the homeostatic regulation of immune responses. TAM receptors have been shown to regulate adult hippocampal neurogenesis by negatively regulation of glial cell activation in central nervous system (CNS). In the present study, we further demonstrated that all three TAM receptors were expressed by cultured primary neural stem cells (NSCs) and played a direct growth trophic role in NSCs proliferation, neuronal differentiation and survival. The cultured primary NSCs lacking TAM receptors exhibited slower growth, reduced proliferation and increased apoptosis as shown by decreased BrdU incorporation and increased TUNEL labeling, than those from the WT NSCs. In addition, the neuronal differentiation and maturation of the mutant NSCs were impeded, as characterized by less neuronal differentiation (ß-tubulin III+) and neurite outgrowth than their WT counterparts. To elucidate the underlying mechanism that the TAM receptors play on the differentiating NSCs, we examined the expression profile of neurotrophins and their receptors by real-time qPCR on the total RNAs from hippocampus and primary NSCs; and found that the TKO NSC showed a significant reduction in the expression of both nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF), but accompanied by compensational increases in the expression of the TrkA, TrkB, TrkC and p75 receptors. These results suggest that TAM receptors support NSCs survival, proliferation and differentiation by regulating expression of neurotrophins, especially the NGF.


Subject(s)
Neural Stem Cells/metabolism , Neurogenesis/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis/genetics , Brain-Derived Neurotrophic Factor/biosynthesis , Cell Proliferation , Cell Survival , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Knockout , Nerve Growth Factors/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptor, trkA/biosynthesis , Receptor, trkB/biosynthesis , Receptor, trkC/biosynthesis , Receptors, Nerve Growth Factor/biosynthesis , Recombinant Proteins , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
14.
PLoS One ; 9(1): e87530, 2014.
Article in English | MEDLINE | ID: mdl-24489934

ABSTRACT

Choroidal neovascularization (CNV) is aberrant angiogenesis associated with exudative age-related macular degeneration (AMD), a leading cause of blindness in the elderly. Inflammation has been suggested as a risk factor for AMD. The IKK2/NF-κB pathway plays a key role in the inflammatory response through regulation of the transcription of cytokines, chemokines, growth factors and angiogenic factors. We investigated the functional role of IKK2 in development of the laser-induced CNV using either Ikk2 conditional knockout mice or an IKK2 inhibitor. The retinal neuronal tissue and RPE deletion of IKK2 was generated by breeding Ikk2(-/flox) mice with Nestin-Cre mice. Deletion of Ikk2 in the retina caused no obvious defect in retinal development or function, but resulted in a significant reduction in laser-induced CNV. In addition, intravitreal or retrobulbar injection of an IKK2 specific chemical inhibitor, TPCA-1, also showed similar inhibition of CNV. Furthermore, in vitro inhibition of IKK2 in ARPE-19 cells significantly reduced heat shock-induced expression of NFKBIA, IL1B, CCL2, VEGFA, PDGFA, HIF1A, and MMP-2, suggesting that IKK2 may regulate multiple molecular pathways involved in laser-induced CNV. The in vivo laser-induced expression of VEGFA, and HIF1A in RPE and choroidal tissue was also blocked by TPCA-1 treatment. Thus, IKK2/NF-κB signaling appears responsible for production of pro-inflammatory and pro-angiogenic factors in laser-induced CNV, suggesting that this intracellular pathway may serve as an important therapeutic target for aberrant angiogenesis in exudative AMD.


Subject(s)
Choroidal Neovascularization/therapy , I-kappa B Kinase/antagonists & inhibitors , Animals , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Lasers , Mice , Mice, Knockout , NF-kappa B/metabolism , NF-kappa B/physiology , Signal Transduction
15.
J Immunol ; 191(12): 6165-77, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24244024

ABSTRACT

TAM tyrosine kinases play multiple functional roles, including regulation of the target genes important in homeostatic regulation of cytokine receptors or TLR-mediated signal transduction pathways. In this study, we show that TAM receptors affect adult hippocampal neurogenesis and loss of TAM receptors impairs hippocampal neurogenesis, largely attributed to exaggerated inflammatory responses by microglia characterized by increased MAPK and NF-κB activation and elevated production of proinflammatory cytokines that are detrimental to neuron stem cell proliferation and neuronal differentiation. Injection of LPS causes even more severe inhibition of BrdU incorporation in the Tyro3(-/-)Axl(-/-)Mertk(-/-) triple-knockout (TKO) brains, consistent with the LPS-elicited enhanced expression of proinflammatory mediators, for example, IL-1ß, IL-6, TNF-α, and inducible NO synthase, and this effect is antagonized by coinjection of the anti-inflammatory drug indomethacin in wild-type but not TKO brains. Conditioned medium from TKO microglia cultures inhibits neuron stem cell proliferation and neuronal differentiation. IL-6 knockout in Axl(-/-)Mertk(-/-) double-knockout mice overcomes the inflammatory inhibition of neurogenesis, suggesting that IL-6 is a major downstream neurotoxic mediator under homeostatic regulation by TAM receptors in microglia. Additionally, autonomous trophic function of the TAM receptors on the proliferating neuronal progenitors may also promote progenitor differentiation into immature neurons.


Subject(s)
Dentate Gyrus/pathology , Microglia/physiology , Neurogenesis/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Astrocytes/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cytokines/biosynthesis , Cytokines/genetics , DNA Replication , Encephalitis/immunology , Encephalitis/pathology , Gene Expression Regulation , Indomethacin/pharmacology , Interleukin-6/antagonists & inhibitors , Interleukin-6/deficiency , Interleukin-6/genetics , Interleukin-6/physiology , Lipopolysaccharides/toxicity , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , NF-kappa B/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/genetics , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Toll-Like Receptors/immunology , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
16.
PLoS One ; 8(6): e64812, 2013.
Article in English | MEDLINE | ID: mdl-23840307

ABSTRACT

The Tyro3, Axl and Mertk (TAM) triply knockout (TKO) mice exhibit systemic autoimmune diseases, with characteristics of increased proinflammatory cytokine production, autoantibody deposition and autoreactive lymphocyte infiltration into a variety of tissues. Here we show that TKO mice produce high level of serum TNF-α and specific autoantibodies deposited onto brain blood vessels. The brain-blood barrier (BBB) in mutant brains exhibited increased permeability for Evans blue and fluorescent-dextran, suggesting a breakdown of the BBB in the mutant brains. Impaired BBB integrity facilitated autoreactive T cells infiltrating into all regions of the mutant brains. Brain autoimmune disorder caused accumulation of the ubiquitin-reactive aggregates in the mutant hippocampus, and early formation of autofluorescent lipofuscins in the neurons throughout the entire brains. Chronic neuroinflammation caused damage of the hippocampal mossy fibers and neuronal apoptotic death. This study shows that chronic systemic inflammation and autoimmune disorders in the TKO mice cause neuronal damage and death.


Subject(s)
Apoptosis , Autoimmune Diseases/genetics , Brain Damage, Chronic/immunology , CA3 Region, Hippocampal/pathology , Neurons/physiology , Animals , Autoantibodies/blood , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Blood-Brain Barrier/metabolism , Brain Damage, Chronic/genetics , Brain Damage, Chronic/pathology , CA3 Region, Hippocampal/blood supply , CA3 Region, Hippocampal/immunology , Capillary Permeability/immunology , Cells, Cultured , Cytokines/metabolism , Dentate Gyrus/blood supply , Dentate Gyrus/immunology , Dentate Gyrus/pathology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Gene Knockdown Techniques , Inclusion Bodies/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microvessels/immunology , Microvessels/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/blood , Ubiquitinated Proteins/metabolism , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
17.
Mol Vis ; 18: 403-11, 2012.
Article in English | MEDLINE | ID: mdl-22355251

ABSTRACT

PURPOSE: The Notch signaling pathway plays crucial roles in regulation of cell proliferation, differentiation and cell fate decision in multiple tissues and cell types. This study was designed to test the effects of enhanced Notch activity on corneal epithelium homeostasis and wound healing using the transgenic mice that overexpressed an activated Notch1 (NICD) in cornea epithelium. METHODS: The studies were performed on R26(fN1-ICD) transgenic mice that carry a NICD cDNA (cDNA) whose expression is prevented by a "Lox-STOP-Lox" cassette. When this transgenic mouse is bred to a mouse strain carrying a Cre recombinase expression cassette driven by a tissue-specific keratin 14 (K14) promoter, the floxed "STOP" cassette is excised and NICD is expressed in the cornea epithelium. The expression level of NICD and its downstream target genes, hairy and enhancer of split 1 (Hes1) and hairy/enhancer-of-split related with YRPW motif 1 (Hey1), in the transgenic corneal epithelium was examined by quantitative PCR (qPCR). The phenotypes and morphology of the transgenic corneal epithelium were compared with that of wild type (WT) controls. The proliferation rate of the epithelial cells was assessed by 5-bromo-2'-deoxyuridine (BrdU) incorporation and the differentiation statues were examined by K14, tumor protein p63 (p63), K12, and zona occludens 1 (ZO-1) immunoreactivity at either normal developmental condition or after corneal epithelial debridement. The corneal epithelial response to wound healing was studied by fluorescent staining and Richardson's staining macroscopically and by H&E staining at microscope level at 0, 6, 12, 18, and 24 h post injury. RESULTS: Although overexpression of NICD in cornea epithelium led to upregulation of its downstream targets, i.e., Hes1 and Hey1, this did not alter corneal epithelial cell proliferation and differentiation. However, wound healing induced Notch activity and overexpression of NICD promoted corneal epithelial wound healing, which was in agreement with more rapid early proliferation response in NICD transgenic mice than in the wild type control mice. CONCLUSIONS: These findings further demonstrate the functional role of Notch signaling in corneal epithelium wound healing response.


Subject(s)
Epithelium, Corneal/injuries , Epithelium, Corneal/physiopathology , Receptor, Notch1/physiology , Wound Healing/physiology , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , DNA Primers/genetics , Epithelium, Corneal/pathology , Gene Expression , Homeodomain Proteins/genetics , Homeostasis , Mice , Mice, Transgenic , Receptor, Notch1/genetics , Signal Transduction , Transcription Factor HES-1 , Wound Healing/genetics
18.
J Cell Biol ; 196(4): 497-511, 2012 Feb 20.
Article in English | MEDLINE | ID: mdl-22351927

ABSTRACT

Although the physiological basis of canonical or classical IκB kinase ß (IKKß)-nuclear factor κB (NF-κB) signaling pathway is well established, how alternative NF-κB signaling functions beyond its role in lymphoid development remains unclear. In particular, alternative NF-κB signaling has been linked with cellular metabolism, but this relationship is poorly understood. In this study, we show that mice deleted for the alternative NF-κB components IKKα or RelB have reduced mitochondrial content and function. Conversely, expressing alternative, but not classical, NF-κB pathway components in skeletal muscle stimulates mitochondrial biogenesis and specifies slow twitch fibers, suggesting that oxidative metabolism in muscle is selectively controlled by the alternative pathway. The alternative NF-κB pathway mediates this specificity by direct transcriptional activation of the mitochondrial regulator PPAR-γ coactivator 1ß (PGC-1ß) but not PGC-1α. Regulation of PGC-1ß by IKKα/RelB also is mammalian target of rapamycin (mTOR) dependent, highlighting a cross talk between mTOR and NF-κB in muscle metabolism. Together, these data provide insight on PGC-1ß regulation during skeletal myogenesis and reveal a unique function of alternative NF-κB signaling in promoting an oxidative metabolic phenotype.


Subject(s)
Cell Respiration , I-kappa B Kinase/metabolism , Muscle Development/physiology , Muscle, Skeletal/metabolism , Myoblasts/metabolism , NF-kappa B/metabolism , Animals , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Gene Expression Regulation , Immunoenzyme Techniques , Luciferases/metabolism , Mice , Mitochondria/metabolism , Muscle, Skeletal/cytology , Myoblasts/cytology , NF-kappa B/genetics , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors
19.
Carcinogenesis ; 32(12): 1782-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21926108

ABSTRACT

14-3-3σ plays a regulatory role in epidermal epithelial differentiation and loss of 14-3-3σ leads to increased proliferation and impaired differentiation. A tumor suppressor function for 14-3-3σ has been proposed based on the fact that some epithelial-derived tumors lose 14-3-3σ expression. p63, a p53 family member, is a master regulator of epidermal epithelial proliferation and differentiation and is necessary for the epidermal development. The function of p63 in tumorigenesis is still controversial and poorly defined as multiple isoforms have been found to play either collaborative or opposing roles. By using 'repeated epilation' heterozygous (Er/+) mice containing a dominant-negative 14-3-3σ mutation, the functional relationship of p63 with 14-3-3σ in epidermal proliferation, differentiation and tumorigenesis was investigated. It was found that p63, particularly the ΔNp63α isoform, was strongly expressed in 14-3-3σ-deficient keratinocytes and knockdown of p63 remarkably inhibited proliferation in these cells. To study the functional roles of 14-3-3σ and p63 in epidermal tumorigenesis, we adopted a 7,12-dimethylbenzanthracene/12-O-tetradecanoyl-phorbol-13-acetate (DMBA/TPA) two-stage tumorigenesis procedure to induce formation of skin papillomas and squamous cell carcinomas in Er/+ mice and identified strong p63 expression in resultant tumors. The loss of one allele of p63 caused by the generation of Er/+/p63(+/-) double compound mice decreased the sensitivity to DMBA-/TPA-induced tumorigenesis as compared with Er/+ mice. This study shows that p63 and 14-3-3σ play opposing roles in the development of skin tumors and that the accumulation of p63 is essential for Ras/14-3-3σ mutation-induced papilloma formation and squamous cell carcinoma carcinogenesis.


Subject(s)
14-3-3 Proteins/physiology , Cell Transformation, Neoplastic , Phosphoproteins/physiology , Skin Neoplasms/physiopathology , Trans-Activators/physiology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Blotting, Western , Carcinogens/toxicity , Cell Differentiation , Cell Proliferation , Gene Silencing , Homeostasis , Keratinocytes/cytology , Mice , Mice, Mutant Strains , Phosphoproteins/genetics , Polymerase Chain Reaction , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/toxicity , Trans-Activators/genetics
20.
J Immunol ; 187(8): 4178-86, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21918185

ABSTRACT

The TAM family of receptors (Tyro3, Axl, and Mertk) plays an important role in the negative regulation of response of dendritic cells (DCs) and macrophages to pathogenic stimuli, and mice lacking this receptor family develop spontaneous lupus-like systemic autoimmunity against a variety of tissues, including retina. To study the molecular mechanism underlying the TAM regulation of APC functions and subsequent effects on the induction of an autoimmune response against the eye, we examined CD4 T cell differentiation following retinal self-antigen immunization. CD4 T cells prepared from naive or interphotoreceptor retinoid-binding protein (IRBP)1-20-immunized Axl and Mertk double-knockout (dko) mice reacted to activation using anti-CD3 and anti-CD28 Abs or to bolster by self-antigen in vitro with a predominantly Th1 effector response, as characterized by increased IFN-γ production and higher frequency of IFN-γ-positive CD4 T cells. The Th17 effector response to IRBP immunization was similar in dko mice to that in wild-type controls, as shown by ELISA measurement of IL-17A in the culture medium and flow cytometric analysis of IL-17A-secreting CD4 T cells. Interestingly, APCs or DCs isolated from IRBP-immunized dko mice exhibited a greater ability to drive the Th1 response. The production of two driving cytokines for Th1 differentiation, IL-12 and IL-18, was dramatically increased in dko DCs and macrophages, and LPS stimulation bolstered their production. The preferential development into the Th1 subset in dko mice suggests that the cytokine milieu produced by the mutant mice in vivo or by mutant APCs in vitro selectively creates a differentiation environment favoring the Th1 effector response.


Subject(s)
Autoantigens/immunology , Autoimmunity/immunology , Cell Differentiation/immunology , Retina/immunology , Th1 Cells/immunology , Animals , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/immunology , Reverse Transcriptase Polymerase Chain Reaction , Th1 Cells/cytology , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...