Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 5(5): 101567, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38744277

ABSTRACT

Bispecific T cell engagers (TCEs) show promising clinical efficacy in blood tumors, but their application to solid tumors remains challenging. Here, we show that Fc-fused IL-7 (rhIL-7-hyFc) changes the intratumoral CD8 T cell landscape, enhancing the efficacy of TCE immunotherapy. rhIL-7-hyFc induces a dramatic increase in CD8 tumor-infiltrating lymphocytes (TILs) in various solid tumors, but the majority of these cells are PD-1-negative tumor non-responsive bystander T cells. However, they are non-exhausted and central memory-phenotype CD8 T cells with high T cell receptor (TCR)-recall capacity that can be triggered by tumor antigen-specific TCEs to acquire tumoricidal activity. Single-cell transcriptome analysis reveals that rhIL-7-hyFc-induced bystander CD8 TILs transform into cycling transitional T cells by TCE redirection with decreased memory markers and increased cytotoxic molecules. Notably, TCE treatment has no major effect on tumor-reactive CD8 TILs. Our results suggest that rhIL-7-hyFc treatment promotes the antitumor efficacy of TCE immunotherapy by increasing TCE-sensitive bystander CD8 TILs in solid tumors.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy , Interleukin-7 , Lymphocytes, Tumor-Infiltrating , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , CD8-Positive T-Lymphocytes/immunology , Interleukin-7/immunology , Interleukin-7/metabolism , Humans , Animals , Immunotherapy/methods , Mice , Neoplasms/immunology , Neoplasms/therapy , Cell Line, Tumor , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Bystander Effect/immunology
2.
J Immunother Cancer ; 12(3)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38471713

ABSTRACT

BACKGROUND: Recombinant human interleukin (rhIL)-7-hyFc (efineptakin alfa; NT-I7) is a potent T-cell amplifier, with two IL-7 molecules fused to IgD/IgG4 elements. rhIL-7-hyFc promotes extensive infiltration of CD8+ T cells into the tumor, concurrently increasing the numbers of intratumoral PD-1+CD8+ T cells. The hIL-2/TCB2 complex (SLC-3010) inhibits tumor growth by preferential activation of CD122 (IL-2Rß)high CD8+ T cells and natural killer cells, over regulatory T cells (Tregs). We investigated the underlying mechanisms of rhIL-7-hyFc and hIL-2/TCB2c antitumor activity and the potential synergistic efficacy, specifically focusing on tumor-specific CD8+ cells within the tumor and the tumor-draining lymph nodes (tdLN). METHODS: MC38 and CT26 tumor-bearing mice were administered with 10 mg/kg rhIL-7-hyFc intramuscularly and 0.9 mg/kg hIL-2/TCB2c intravenously. Anti-PD-1 monoclonal antibody was administered intraperitoneally three times at 3-day intervals at a dose of 5 mg/kg. Tumor volume was measured to assess efficacy. To compare the composition of immune cells between each monotherapy and the combination therapy, we analyzed tumors and tdLNs by flow cytometry. RESULTS: Our data demonstrate that the combination of rhIL-7-hyFc and hIL-2/TCB2c increases efficacy and generates an immune-stimulatory tumor microenvironment (TME). The TME is characterized by an increased infiltration of tumor-specific CD8+ T cells, and a decreased frequency of CD39highTIM-3+ Treg cells. Most importantly, rhIL-7-hyFc increases infiltration of a CD62L+Ly108+ early progenitor population of exhausted CD8+ T cells (TPEX), which may retain long-term proliferation capacity and replenish functional effector CD8+ T cells. hIL-2/TCB2c induces differentiation of CD62L+Ly108+ TPEX rapidly into CD101+ terminally differentiated subsets (terminally exhausted T cell (TEX term)). Our study also demonstrates that rhIL-7-hyFc significantly enhances the proliferation rate of TPEX in the tdLNs, positively correlating with their abundance within the tumor. Moreover, rhIL-7-hyFc and hIL-2/TCB2c can overcome the limited therapeutic effectiveness of PD-1 blockade, culminating in the complete regression of tumors. CONCLUSIONS: rhIL-7-hyFc can expand and maintain the progenitor pool of exhausted CD8+ T cells, whereas hIL-2/TCB2c promotes their differentiation into TEX term. Together, this induces an immune-stimulatory TME that improves the efficacy of checkpoint blockade.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-7 , Neoplasms , Recombinant Fusion Proteins , Humans , Animals , Mice , Tumor Microenvironment , Programmed Cell Death 1 Receptor , Immunologic Factors
3.
Cell Rep Med ; 5(1): 101362, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38232693

ABSTRACT

Repeated pandemics caused by the influenza virus and severe acute respiratory syndrome coronavirus (SARS-CoV) have resulted in serious problems in global public health, emphasizing the need for broad-spectrum antiviral therapeutics against respiratory virus infections. Here, we show the protective effects of long-acting recombinant human interleukin-7 fused with hybrid Fc (rhIL-7-hyFc) against major respiratory viruses, including influenza virus, SARS-CoV-2, and respiratory syncytial virus. Administration of rhIL-7-hyFc in a therapeutic or prophylactic regimen induces substantial antiviral effects. During an influenza A virus (IAV) infection, rhIL-7-hyFc treatment increases pulmonary T cells composed of blood-derived interferon γ (IFNγ)+ conventional T cells and locally expanded IL-17A+ innate-like T cells. Single-cell RNA transcriptomics reveals that rhIL-7-hyFc upregulates antiviral genes in pulmonary T cells and induces clonal expansion of type 17 innate-like T cells. rhIL-7-hyFc-mediated disease prevention is dependent on IL-17A in both IAV- and SARS-CoV-2-infected mice. Collectively, we suggest that rhIL-7-hyFc can be used as a broadly active therapeutic for future respiratory virus pandemic.


Subject(s)
Influenza, Human , Interleukin-17 , Animals , Mice , Humans , Interleukin-17/genetics , Interleukin-7 , T-Lymphocytes , SARS-CoV-2 , Influenza, Human/drug therapy , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
4.
Microbiome ; 10(1): 188, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333752

ABSTRACT

BACKGROUND: Comparisons of the gut microbiome of lean and obese humans have revealed that obesity is associated with the gut microbiome plus changes in numerous environmental factors, including high-fat diet (HFD). Here, we report that two species of Bifidobacterium are crucial to controlling metabolic parameters in the Korean population. RESULTS: Based on gut microbial analysis from 99 Korean individuals, we observed the abundance of Bifidobacterium longum and Bifidobacterium bifidum was markedly reduced in individuals with increased visceral adipose tissue (VAT), body mass index (BMI), blood triglyceride (TG), and fatty liver. Bacterial transcriptomic analysis revealed that carbohydrate/nucleoside metabolic processes of Bifidobacterium longum and Bifidobacterium bifidum were associated with protecting against diet-induced obesity. Oral treatment of specific commercial Bifidobacterium longum and Bifidobacterium bifidum enhanced bile acid signaling contributing to potentiate oxidative phosphorylation (OXPHOS) in adipose tissues, leading to reduction of body weight gain and improvement in hepatic steatosis and glucose homeostasis. Bifidobacterium longum or Bifidobacterium bifidum manipulated intestinal sterol biosynthetic processes to protect against diet-induced obesity in germ-free mice. CONCLUSIONS: Our findings support the notion that treatment of carbohydrate/nucleoside metabolic processes-enriched Bifidobacterium longum and Bifidobacterium bifidum would be a novel therapeutic strategy for reprograming the host metabolic homeostasis to protect against metabolic syndromes, including diet-induced obesity. Video Abstract.


Subject(s)
Bifidobacterium longum , Bifidobacterium , Humans , Mice , Animals , Bifidobacterium/metabolism , Nucleosides/metabolism , Nucleosides/therapeutic use , Oxidative Phosphorylation , Obesity/microbiology , Diet, High-Fat/adverse effects , Adipose Tissue, White/metabolism
5.
Proc Natl Acad Sci U S A ; 116(42): 21140-21149, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31570593

ABSTRACT

Retinoic acid-related orphan receptor α (RORα) functions as a transcription factor for various biological processes, including circadian rhythm, cancer, and metabolism. Here, we generate intestinal epithelial cell (IEC)-specific RORα-deficient (RORαΔIEC) mice and find that RORα is crucial for maintaining intestinal homeostasis by attenuating nuclear factor κB (NF-κB) transcriptional activity. RORαΔIEC mice exhibit excessive intestinal inflammation and highly activated inflammatory responses in the dextran sulfate sodium (DSS) mouse colitis model. Transcriptome analysis reveals that deletion of RORα leads to up-regulation of NF-κB target genes in IECs. Chromatin immunoprecipitation analysis reveals corecruitment of RORα and histone deacetylase 3 (HDAC3) on NF-κB target promoters and subsequent dismissal of CREB binding protein (CBP) and bromodomain-containing protein 4 (BRD4) for transcriptional repression. Together, we demonstrate that RORα/HDAC3-mediated attenuation of NF-κB signaling controls the balance of inflammatory responses, and therapeutic strategies targeting this epigenetic regulation could be beneficial to the treatment of chronic inflammatory diseases, including inflammatory bowel disease (IBD).


Subject(s)
Homeostasis/physiology , Inflammation/metabolism , Intestines/physiology , Orphan Nuclear Receptors/metabolism , Animals , Epigenesis, Genetic/physiology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Female , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Transcription Factors/metabolism , Transcription, Genetic/physiology , Transcriptome/physiology
6.
Cell Rep ; 26(5): 1357-1367.e5, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30699360

ABSTRACT

Katanin was the first microtubule (MT)-severing enzyme discovered, but how katanin executes MT severing remains poorly understood. Here, we report X-ray crystal structures of the apo and ATPγS-bound states of the catalytic AAA domain of human katanin p60 at 3.0 and 2.9 Å resolution, respectively. Comparison of the two structures reveals conformational changes induced by ATP binding and how such changes ensure hexamer stability. Moreover, we uncover structural details of pore loops (PLs) and show that Arg283, a residue unique to katanin among MT-severing enzymes, protrudes from PL1 and lines the entry of the catalytic pore. Functional studies suggest that PL1 and Arg283 play essential roles in the recognition and remodeling of the glutamylated, C-terminal tubulin tail and regulation of axon growth. In addition, domain-swapping experiments in katanin and spastin suggest that the non-homologous N-terminal region, which contains the MT-interacting and trafficking domain and a linker, confers specificity to the severing process.


Subject(s)
Glutamates/metabolism , Katanin/chemistry , Katanin/metabolism , Microtubules/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Arginine/metabolism , Axons/metabolism , HeLa Cells , Humans , Mice, Inbred ICR , Models, Molecular , Mutation/genetics , Protein Domains , Protein Multimerization , Sensory Receptor Cells/metabolism , Spastin/metabolism
7.
Glia ; 67(2): 360-375, 2019 02.
Article in English | MEDLINE | ID: mdl-30444070

ABSTRACT

Schwann cells (SCs), the primary glia in the peripheral nervous system (PNS), display remarkable plasticity in that fully mature SCs undergo dedifferentiation and convert to repair SCs upon nerve injury. Dedifferentiated SCs provide essential support for PNS regeneration by producing signals that enhance the survival and axon regrowth of damaged neurons, but the identities of neurotrophic factors remain incompletely understood. Here we show that SCs express and secrete progranulin (PGRN), depending on the differentiation status of SCs. PGRN expression and secretion markedly increased as primary SCs underwent dedifferentiation, while PGRN secretion was prevented by administration of cAMP, which induced SC differentiation. We also found that sciatic nerve injury, a physiological trigger of SC dedifferentiation, induced PGRN expression in SCs in vivo. These results suggest that dedifferentiated SCs express and secrete PGRN that functions as a paracrine factor to support the survival and axon growth of neighboring neurons after injury.


Subject(s)
Axons/pathology , Cell Proliferation/drug effects , Motor Neurons/pathology , Progranulins/metabolism , Schwann Cells/metabolism , Sciatic Neuropathy/pathology , Animals , Axons/drug effects , Bucladesine/pharmacology , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Fluoresceins/metabolism , Mass Spectrometry , Mice , Mice, Inbred ICR , Motor Neurons/drug effects , Progranulins/pharmacology , RNA, Messenger/metabolism , Schwann Cells/chemistry , Spinal Cord/cytology
8.
Cell Death Dis ; 9(11): 1118, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389914

ABSTRACT

Although additional sex combs-like 1 (ASXL1) has been extensively described in hematologic malignancies, little is known about the molecular role of ASXL1 in organ development. Here, we show that Asxl1 ablation in mice results in postnatal lethality due to cyanosis, a respiratory failure. This lung defect is likely caused by higher proliferative potential and reduced expression of surfactant proteins, leading to reduced air space and defective lung maturation. By microarray analysis, we identified E2F1-responsive genes, including Nmyc, as targets repressed by Asxl1. Nmyc and Asxl1 are reciprocally expressed during the fetal development of normal mouse lungs, whereas Nmyc downregulation is impaired in Asxl1-deficient lungs. Together with E2F1 and ASXL1, host cell factor 1 (HCF-1), purified as an Asxl1-bound protein, is recruited to the E2F1-binding site of the Nmyc promoter. The interaction occurs between the C-terminal region of Asxl1 and the N-terminal Kelch domain of HCF-1. Trimethylation (me3) of histone H3 lysine 27 (H3K27) is enriched in the Nmyc promoter upon Asxl1 overexpression, whereas it is downregulated in Asxl1-deleted lung and -depleted A549 cells, similar to H3K9me3, another repressive histone marker. Overall, these findings suggest that Asxl1 modulates proliferation of lung epithelial cells via the epigenetic repression of Nmyc expression, deficiency of which may cause hyperplasia, leading to dyspnea.


Subject(s)
E2F1 Transcription Factor/genetics , Epigenetic Repression , Epithelial Cells/metabolism , Lung/metabolism , N-Myc Proto-Oncogene Protein/genetics , Repressor Proteins/genetics , Respiratory Insufficiency/genetics , A549 Cells , Animals , E2F1 Transcription Factor/metabolism , Embryo, Mammalian , Epithelial Cells/pathology , Fetus , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Lethal , HEK293 Cells , Histones/genetics , Histones/metabolism , Host Cell Factor C1/genetics , Host Cell Factor C1/metabolism , Humans , Lung/growth & development , Lung/pathology , Mice , Mice, Knockout , N-Myc Proto-Oncogene Protein/metabolism , Organogenesis/genetics , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/deficiency , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/pathology , Signal Transduction
9.
Lab Anim Res ; 34(4): 147-159, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30671100

ABSTRACT

Genetically engineered mouse models are commonly preferred for studying the human disease due to genetic and pathophysiological similarities between mice and humans. In particular, Cre-loxP system is widely used as an integral experimental tool for generating the conditional. This system has enabled researchers to investigate genes of interest in a tissue/cell (spatial control) and/or time (temporal control) specific manner. A various tissue-specific Cre-driver mouse lines have been generated to date, and new Cre lines are still being developed. This review provides a brief overview of Cre-loxP system and a few commonly used promoters for expression of tissue-specific Cre recombinase. Also, we finally introduce some available links to the Web sites that provides detailed information about Cre mouse lines including their characterization.

10.
Exp Neurobiol ; 26(3): 158-167, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28680301

ABSTRACT

Brain is a rich environment where neurons and glia interact with neighboring cells as well as extracellular matrix in three-dimensional (3D) space. Astrocytes, which are the most abundant cells in the mammalian brain, reside in 3D space and extend highly branched processes that form microdomains and contact synapses. It has been suggested that astrocytes cultured in 3D might be maintained in a less reactive state as compared to those growing in a traditional, two-dimensional (2D) monolayer culture. However, the functional characterization of the astrocytes in 3D culture has been lacking. Here we cocultured neurons and astrocytes in 3D and examined the morphological, molecular biological, and electrophysiological properties of the 3D-cultured hippocampal astrocytes. In our 3D neuron-astrocyte coculture, astrocytes showed a typical morphology of a small soma with many branches and exhibited a unique membrane property of passive conductance, more closely resembling their native in vivo counterparts. Moreover, we also induced reactive astrocytosis in culture by infecting with high-titer adenovirus to mimic pathophysiological conditions in vivo. Adenoviral infection induced morphological changes in astrocytes, increased passive conductance, and increased GABA content as well as tonic GABA release, which are characteristics of reactive gliosis. Together, our study presents a powerful in vitro model resembling both physiological and pathophysiological conditions in vivo, and thereby provides a versatile experimental tool for studying various neurological diseases that accompany reactive astrocytes.

11.
Nat Commun ; 8: 14346, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28146148

ABSTRACT

In native tissues, cellular and acellular components are anisotropically organized and often aligned in specific directions, providing structural and mechanical properties for actuating biological functions. Thus, engineering alignment not only allows for emulation of native tissue structures but might also enable implementation of specific functionalities. However, achieving desired alignment is challenging, especially in three-dimensional constructs. By exploiting the elastomeric property of polydimethylsiloxane and fibrillogenesis kinetics of collagen, here we introduce a simple yet effective method to assemble and align fibrous structures in a multi-modular three-dimensional conglomerate. Applying this method, we have reconstructed the CA3-CA1 hippocampal neural circuit three-dimensionally in a monolithic gel, in which CA3 neurons extend parallel axons to and synapse with CA1 neurons. Furthermore, we show that alignment of the fibrous scaffold facilitates the establishment of functional connectivity. This method can be applied for reconstructing other neural circuits or tissue units where anisotropic organization in a multi-modular structure is desired.


Subject(s)
Cell Culture Techniques/methods , Hippocampus/physiology , Nerve Net/physiology , Neurons/physiology , Tissue Scaffolds/chemistry , Animals , Anisotropy , Cells, Cultured , Collagen/chemistry , Hippocampus/cytology , Mice, Inbred ICR , Microscopy, Confocal , Nerve Net/cytology , Neurons/cytology , Rats, Sprague-Dawley , Synapses/physiology , Time-Lapse Imaging/methods , Tissue Engineering/methods
12.
Neural Plast ; 2016: 5056418, 2016.
Article in English | MEDLINE | ID: mdl-27872763

ABSTRACT

Several studies have demonstrated the therapeutic potential of applying microtubule- (MT-) stabilizing agents (MSAs) that cross the blood-brain barrier to promote axon regeneration and prevent axonal dystrophy in rodent models of spinal cord injury and neurodegenerative diseases. Paradoxically, administration of MSAs, which have been widely prescribed to treat malignancies, is well known to cause debilitating peripheral neuropathy and axon degeneration. Despite the growing interest of applying MSAs to treat the injured or degenerating central nervous system (CNS), consequences of MSA exposure to neurons in the central and peripheral nervous system (PNS) have not been thoroughly investigated. Here, we have examined and compared the effects of a brain-penetrant MSA, epothilone B, on cortical and sensory neurons in culture and show that epothilone B exhibits both beneficial and detrimental effects, depending on not only the concentration of drug but also the type and age of a neuron, as seen in clinical settings. Therefore, to exploit MSAs to their full benefit and minimize unwanted side effects, it is important to understand the properties of neuronal MTs and strategies should be devised to deliver minimal effective concentration directly to the site where needed.


Subject(s)
Epothilones/pharmacology , Microtubules/physiology , Neurons/physiology , Tubulin Modulators/pharmacology , Age Factors , Animals , Animals, Newborn , Brain/cytology , Brain/drug effects , Brain/physiology , Cells, Cultured , Female , Mice , Mice, Inbred ICR , Microtubules/drug effects , Neurons/drug effects
13.
J Neuroimmunol ; 295-296: 21-9, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27235345

ABSTRACT

Tonicity-responsive enhancer (TonE) binding protein (TonEBP) is known as an osmosensitive transcription factor that regulates cellular homeostasis during states of hypo- and hypertonic stress. In addition to its role in osmoadaptation, growing lines of evidence suggest that TonEBP might have tonicity-independent functions. In particular, a number of studies suggest that inflammatory stimuli induce the expression and activation of TonEBP in peripheral immune cells. However, whether TonEBP is expressed in microglia, resident immune cells of the central nervous system, is unknown. Here we show that inflammatory signals induce the expression of TonEBP in microglia both in vitro and in vitro. In cultured primary microglia, treatment with lipopolysaccharide (LPS), interferon-γ, and interleukin 4 increased the expression of TonEBP. Moreover, we found that stereotaxic injection of LPS into the substantia nigra region of rat brain increased TonEBP expression in OX-42-positive cells. Furthermore, expression of TonEBP was induced in OX-42-positive cells in a rat model of transient middle cerebral artery occlusion. Together these results show that the expression of TonEBP is regulated by inflammatory signals in mammalian brain, suggesting that TonEBP might play a part during neuroinflammation.


Subject(s)
Encephalitis/pathology , Mesencephalon/pathology , Microglia/metabolism , Transcription Factors/metabolism , Animals , CD11b Antigen/metabolism , Disease Models, Animal , Encephalitis/chemically induced , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Microglia/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics
14.
Biomicrofluidics ; 9(2): 024115, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25945141

ABSTRACT

We present an engineered three-dimensional (3D) in vitro brain microvasculature system embedded within the bulk of a collagen matrix. To create a hydrogel template for the functional brain microvascular structure, we fabricated an array of microchannels made of collagen I using microneedles and a 3D printed frame. By culturing mouse brain endothelial cells (bEnd.3) on the luminal surface of cylindrical collagen microchannels, we reconstructed an array of brain microvasculature in vitro with circular cross-sections. We characterized the barrier function of our brain microvasculature by measuring transendothelial permeability of 40 kDa fluorescein isothiocyanate-dextran (Stoke's radius of ∼4.5 nm), based on an analytical model. The transendothelial permeability decreased significantly over 3 weeks of culture. We also present the disruption of the barrier function with a hyperosmotic mannitol as well as a subsequent recovery over 4 days. Our brain microvasculature model in vitro, consisting of system-in-hydrogel combined with the widely emerging 3D printing technique, can serve as a useful tool not only for fundamental studies associated with blood-brain barrier in physiological and pathological settings but also for pharmaceutical applications.

15.
PLoS One ; 9(3): e91402, 2014.
Article in English | MEDLINE | ID: mdl-24614691

ABSTRACT

Serotonin (5-HT) receptors of type 6 (5-HT6R) play important roles in mood, psychosis, and eating disorders. Recently, a growing number of studies support the use of 5-HT6R-targeting compounds as promising drug candidates for treating cognitive dysfunction associated with Alzheimer's disease. However, the mechanistic linkage between 5-HT6R and such functions remains poorly understood. By using yeast two-hybrid, GST pull-down, and co-immunoprecipitation assays, here we show that human 5-HT6R interacts with the light chain 1 (LC1) subunit of MAP1B protein (MAP1B-LC1), a classical microtubule-associated protein highly expressed in the brain. Functionally, we have found that expression of MAP1B-LC1 regulates serotonin signaling in a receptor subtype-specific manner, specifically controlling the activities of 5-HT6R, but not those of 5-HT4R and 5-HT7R. In addition, we have demonstrated that MAP1B-LC1 increases the surface expression of 5-HT6R and decreases its endocytosis, suggesting that MAP1B-LC1 is involved in the desensitization and trafficking of 5-HT6R via a direct interaction. Together, we suggest that signal transduction pathways downstream of 5-HT6R are regulated by MAP1B, which might play a role in 5-HT6R-mediated signaling in the brain.


Subject(s)
Microtubule-Associated Proteins/metabolism , Protein Subunits/metabolism , Receptors, Serotonin/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Endocytosis , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunoprecipitation , Ligands , Mice , Protein Binding , Receptors, Serotonin/chemistry
16.
PLoS One ; 9(2): e88811, 2014.
Article in English | MEDLINE | ID: mdl-24558432

ABSTRACT

During gastrulation, distinct lineage specification into three germ layers, the mesoderm, endoderm and ectoderm, occurs through an elaborate harmony between signaling molecules along the embryonic proximo-distal and anterior-posterior axes, and Nodal signaling plays a key role in the early embryonic development governing embryonic axis formation, mesoderm and endoderm specification, and left-right asymmetry determination. However, the mechanism by which Nodal expression is regulated is largely unknown. Here, we show that Meteorin regulates Nodal expression and is required for mesendoderm development. It is highly expressed in the inner cell mass of blastocysts and further in the epiblast and extra-embryonic ectoderm during gastrulation. Genetic ablation of the Meteorin gene resulted in early embryonic lethality, presumably due to impaired lineage allocation and subsequent cell accumulation. Embryoid body culture using Meteorin-null embryonic stem (ES) cells showed reduced Nodal expression and concomitant impairment of mesendoderm specification. Meteorin-null embryos displayed reduced levels of Nodal transcripts before the gastrulation stage, and impaired expression of Goosecoid, a definitive endoderm marker, during gastrulation, while the proximo-distal and anterior-posterior axes and primitive streak formation were preserved. Our results show that Meteorin is a novel regulator of Nodal transcription and is required to maintain sufficient Nodal levels for endoderm formation, thereby providing new insights in the regulation of mesendoderm allocation.


Subject(s)
Gene Expression Regulation, Developmental , Mesoderm/embryology , Nerve Tissue Proteins/metabolism , Nodal Protein/genetics , Nodal Protein/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Embryoid Bodies/cytology , Female , Gastrulation , Gene Knockout Techniques , Mesoderm/cytology , Mice , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Pregnancy , Signal Transduction , Transcription, Genetic
17.
PLoS One ; 8(3): e58511, 2013.
Article in English | MEDLINE | ID: mdl-23526992

ABSTRACT

Sorting nexin 5 (Snx5) has been posited to regulate the degradation of epidermal growth factor receptor and the retrograde trafficking of cation-independent mannose 6-phosphate receptor/insulin-like growth factor II receptor. Snx5 has also been suggested to interact with Mind bomb-1, an E3 ubiquitin ligase that regulates the activation of Notch signaling. However, the in vivo functions of Snx5 are largely unknown. Here, we report that disruption of the Snx5 gene in mice (Snx5(-/-) mice) resulted in partial perinatal lethality; 40% of Snx5(-/-) mice died shortly after birth due to cyanosis, reduced air space in the lungs, and respiratory failure. Histological analysis revealed that Snx5(-/-) mice exhibited thickened alveolar walls associated with undifferentiated alveolar epithelial type I cells. In contrast, alveolar epithelial type II cells were intact, exhibiting normal surfactant synthesis and secretion. Although the expression levels of surfactant proteins and saturated phosphatidylcholine in the lungs of Snx5(-/-) mice were comparable to those of Snx5(+/+) mice, the expression levels of T1α, Aqp5, and Rage, markers for distal alveolar epithelial type I cells, were significantly decreased in Snx5 (-/-) mice. These results demonstrate that Snx5 is necessary for the differentiation of alveolar epithelial type I cells, which may underlie the adaptation to air breathing at birth.


Subject(s)
Alveolar Epithelial Cells/physiology , Respiratory Insufficiency/etiology , Sorting Nexins/deficiency , Alveolar Epithelial Cells/classification , Alveolar Epithelial Cells/pathology , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Differentiation/physiology , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Pulmonary Alveoli/abnormalities , Pulmonary Surfactant-Associated Proteins/genetics , Pulmonary Surfactant-Associated Proteins/metabolism , Respiratory Insufficiency/pathology , Respiratory Insufficiency/physiopathology , Sorting Nexins/genetics , Sorting Nexins/physiology
18.
PLoS One ; 8(1): e53577, 2013.
Article in English | MEDLINE | ID: mdl-23308255

ABSTRACT

Mitochondria are key organelles dedicated to energy production. Crif1, which interacts with the large subunit of the mitochondrial ribosome, is indispensable for the mitochondrial translation and membrane insertion of respiratory subunits. To explore the physiological function of Crif1 in the heart, Crif1(f/f) mice were crossed with Myh6-cre/Esr1 transgenic mice, which harbor cardiomyocyte-specific Cre activity in a tamoxifen-dependent manner. The tamoxifen injections were given at six weeks postnatal, and the mutant mice survived only five months due to hypertrophic heart failure. In the mutant cardiac muscles, mitochondrial mass dramatically increased, while the inner structure was altered with lack of cristae. Mutant cardiac muscles showed decreased rates of oxygen consumption and ATP production, suggesting that Crif1 plays a critical role in the maintenance of both mitochondrial structure and respiration in cardiac muscles.


Subject(s)
Cardiomyopathies/pathology , Cell Cycle Proteins/genetics , Heart Failure/pathology , Mitochondria, Heart/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cell Cycle Proteins/deficiency , Cell Respiration , Crosses, Genetic , Gene Deletion , Heart Failure/genetics , Heart Failure/metabolism , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Mitochondria, Heart/genetics , Mitochondria, Heart/ultrastructure , Myocardium/metabolism , Myocytes, Cardiac/pathology , Oxygen Consumption , Tamoxifen
19.
PLoS One ; 5(12): e15254, 2010 Dec 13.
Article in English | MEDLINE | ID: mdl-21179456

ABSTRACT

Vaccinia-related kinase 1 (VRK1) is a crucial protein kinase for mitotic regulation. VRK1 is known to play a role in germ cell development, and its deficiency results in sterility. Here we describe that VRK1 is essential for the maintenance of spermatogonial stem cells. To determine whether VRK1 plays a role in these cells, we assessed the population size of undifferentiated spermatogonia. Flow cytometry analyses showed that the number of undifferentiated spermatogonia was markedly reduced in VRK1-deficient testes. VRK1 was highly expressed in spermatogonial populations, and approximately 66% of undifferentiated spermatogonia that were sorted as an Ep-CAM+/c-kit-/alpha-6-integrin+ population showed a positive signal for VRK1. Undifferentiated stem cells expressing Plzf and Oct4 but not c-kit also expressed VRK1, suggesting that VRK1 is an intrinsic factor for the maintenance of spermatogonial stem cells. Microarray analyses of the global testicular transcriptome and quantitative RT-PCR of VRK1-deficient testes revealed significantly reduced expression levels of undifferentiated spermatogonial marker genes in early postnatal mice. Together, these results suggest that VRK1 is required for the proliferation and differentiation of undifferentiated spermatogonia, which are essential for spermatogenic cell maintenance.


Subject(s)
Gene Expression Regulation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Spermatogonia/cytology , Animals , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Cell Differentiation , Epithelial Cell Adhesion Molecule , In Situ Hybridization , Integrin alpha6/metabolism , Male , Mice , Models, Biological , Octamer Transcription Factor-3/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spermatogonia/physiology , Stem Cells/cytology , Testis/cytology
20.
J Clin Invest ; 119(11): 3290-300, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19855135

ABSTRACT

The heterogeneous cellular composition of the mammalian renal collecting duct enables regulation of fluid, electrolytes, and acid-base homeostasis, but the molecular mechanism of its development has yet to be elucidated. The Notch signaling pathway is involved in cell fate determination and has been implicated in proximal-distal patterning in the mammalian kidney. To investigate the role of Notch signaling in renal collecting duct development, we generated mice in which Mind bomb-1 (Mib1), an E3 ubiquitin ligase required for the initiation of Notch signaling, was specifically inactivated in the ureteric bud of the developing kidney. Mice lacking Mib1 in the renal collecting duct displayed increased urinary production, decreased urinary osmolality, progressive hydronephrosis, sodium wasting, and a severe urinary concentrating defect manifested as nephrogenic diabetes insipidus. Histological analysis revealed a diminished number of principal cells and corresponding increase in the number of intercalated cells. Transgenic overexpression of Notch intracellular domain reversed the altered cellular composition of mutant renal collecting duct, with principal cells occupying the entire region. Our data demonstrate that Notch signaling is required for the development of the mammalian renal collecting duct and principal cell differentiation and indicate that pathway dysregulation may contribute to distal renal tubular disorders.


Subject(s)
Diabetes Insipidus, Nephrogenic/physiopathology , Kidney Tubules, Collecting/physiopathology , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Differentiation , Gene Expression , Hydronephrosis/physiopathology , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/embryology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...