Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Am J Physiol Cell Physiol ; 320(3): C448-C461, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33471620

ABSTRACT

Gram-negative bacterial lipopolysaccharide (LPS) increases the susceptibility of cells to pathogenic diseases, including inflammatory diseases and septic syndrome. In our experiments, we examined whether LPS induces epithelial barrier disruption in secretory epithelia and further investigated its underlying mechanism. The activities of Ca2+-activated Cl- channels (CACC) and epithelial Na+ channels (ENaC) were monitored with a short-circuit current using an Ussing chamber. Epithelial membrane integrity was estimated via transepithelial electrical resistance and paracellular permeability assays. We found that the apical application of LPS evoked short-circuit current (Isc) through the activation of CACC and ENaC. Although LPS disrupted epithelial barrier integrity, this was restored with the inhibition of CACC and ENaC, indicating the role of CACC and ENaC in the regulation of paracellular pathways. We confirmed that LPS, CACC, or ENaC activation evoked apical membrane depolarization. The exposure to a high-K+ buffer increased paracellular permeability. LPS induced the rapid redistribution of zonula occludens-1 (ZO-1) and reduced the expression levels of ZO-1 in tight junctions through apical membrane depolarization and tyrosine phosphorylation. However, the LPS-induced epithelial barrier disruption and degradation of ZO-1 were largely recovered by blocking CACC and ENaC. Furthermore, although LPS-impaired epithelial barrier became vulnerable to secondary bacterial infections, this vulnerability was prevented by inhibiting CACC and ENaC. We concluded that LPS induces the disruption of epithelial barrier integrity through the activation of CACC and ENaC, resulting in apical membrane depolarization and the subsequent tyrosine phosphorylation of ZO-1.


Subject(s)
Chloride Channels/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelium/drug effects , Epithelium/metabolism , Lipopolysaccharides/pharmacology , Sodium Channels/metabolism , Animals , Cells, Cultured , Male , Membrane Potentials/drug effects , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Tight Junctions/drug effects , Tight Junctions/metabolism , Zonula Occludens-1 Protein/metabolism
2.
Int J Mol Sci ; 20(24)2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31847128

ABSTRACT

Anoctamin1 (ANO1), a calcium activated chloride channel, is known to play a critical role in salivary secretion. In the salivary gland, ANO1 is expressed exclusively in the acinar cells, with no expression in the ductal cells. However, the mechanisms that determine this distinctive cell type-dependent expression pattern of ANO1 remain unknown. In this study, we discovered that the cell-dependent expression of ANO1 during salivary gland organogenesis is regulated by DNA methylation of ANO1 CpG islands. ANO1 CpG islands in e12 embryonic submandibular glands (eSMG) are highly methylated, but those in e14 eSMG or adult SMG are significantly unmethylated. The differential expression pattern of ANO1 in duct and acini is defined at e14. Artificial demethylation by treatment with the demethylating agent 5-aza-2'-deoxycytidine (5-Aza-CdR), induced the expression of ANO1 in both the ductal cell line Human Submandibular Gland (HSG) and in the duct cells of adult mouse SMG. During the trans-differentiation in Matrigel of duct-origin HSG cells into acinar-like phenotype, significant demethylation of ANO1 CpG islands is observed. This may be due to the reduced expression of DNA methyltransferase (DNMT) 3a and 3b. These results suggest that the differential expression of ANO1 in salivary glands during organogenesis and differentiation is mainly regulated by epigenetic demethylation of the ANO1 gene.


Subject(s)
Anoctamin-1/biosynthesis , CpG Islands , DNA Methylation , Epigenesis, Genetic , Neoplasm Proteins/biosynthesis , Salivary Glands/metabolism , Animals , Cell Line , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methyltransferase 3A , Decitabine/pharmacology , Humans , Mice , Salivary Glands/cytology , DNA Methyltransferase 3B
3.
Biochem Biophys Res Commun ; 481(1-2): 31-37, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27833020

ABSTRACT

Cystic fibrosis transmembrane conductance regulator (CFTR) plays a key role in exocrine secretion, including salivary glands. However, its functional expression in salivary glands has not been rigorously studied. In this study, we investigated the expression pattern and regulatory mechanism of CFTR in salivary glands using immunohistochemistry, western blot analysis, Ussing chamber study, methylation-specific PCR, and bisulfite sequencing. Using an organ culture technique, we found that CFTR expression was first detected on the 15th day at the embryonic stage (E15) and was observed in ducts but not in acini. CFTR expression was confirmed in HSG and SIMS cell lines, which both originated from ducts, but not in the SMG C-6 cell line, which originated from acinar cells. Treatment of SMG C-6 cells with 5-aza-2'-deoxycytidine (5-Aza-CdR) restored the expression level of CFTR mRNA in a time-dependent manner. Restoration of CFTR was further confirmed by a functional study. In the Ussing chamber study, 10 µM Cact-A1, a CFTR activator, did not evoke any currents in SMG C-6 cells. In contrast, in SMG C-6 cells pretreated with 5-Aza-CdR, Cact-A1 evoked a robust increase of currents, which were inhibited by the CFTR inhibitor CFTRinh-172. Furthermore, forskolin mimicked the currents activated by Cact-A1. In our epigenetic study, SMG C-6 cells showed highly methylated CG pairs in the CFTR CpG island and most of the methylated CG pairs were demethylated by 5-Aza-CdR. Our results suggest that epigenetic regulation is involved in the development of salivary glands by silencing the CFTR gene in a tissue-specific manner.


Subject(s)
Acinar Cells/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Salivary Glands/embryology , Salivary Glands/physiology , Cell Line , Gene Expression Regulation/genetics , Humans
4.
Molecules ; 21(7)2016 Jun 25.
Article in English | MEDLINE | ID: mdl-27347918

ABSTRACT

Capsaicin (trans-8-methyl-N-vanilyl-6-nonenamide) is a unique alkaloid isolated from hot chili peppers of the capsicum family. Capsaicin is an agonist of transient receptor potential vanilloid subtype 1 (TRPV1), which is expressed in nociceptive sensory neurons and a range of secretory epithelia, including salivary glands. Capsaicin has analgesic and anti-inflammatory properties in sensory neurons. Recently, increasing evidence has indicated that capsaicin also affects saliva secretion and inflammation in salivary glands. Applying capsaicin increases salivary secretion in human and animal models. Capsaicin appears to increase salivation mainly by modulating the paracellular pathway in salivary glands. Capsaicin activates TRPV1, which modulates the permeability of tight junctions (TJ) by regulating the expression and function of putative intercellular adhesion molecules in an ERK (extracelluar signal-regulated kinase) -dependent manner. Capsaicin also improved dysfunction in transplanted salivary glands. Aside from the secretory effects of capsaicin, it has anti-inflammatory effects in salivary glands. The anti-inflammatory effect of capsaicin is, however, not mediated by TRPV1, but by inhibition of the NF-κB pathway. In conclusion, capsaicin might be a potential drug for alleviating dry mouth symptoms and inflammation of salivary glands.


Subject(s)
Capsaicin/pharmacology , Salivary Glands/drug effects , Salivary Glands/physiopathology , Sensory System Agents/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Capsaicin/therapeutic use , Humans , Inflammation/drug therapy , Inflammation/metabolism , Receptors, Muscarinic/metabolism , Salivary Glands/metabolism , Salivation/drug effects , Sensory System Agents/therapeutic use , TRPV Cation Channels/metabolism
5.
Lab Invest ; 95(2): 237-45, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25485536

ABSTRACT

Muscarinic receptors, particularly the type 3 subtype (M3R), have an important role in exocrine secretion. M3R normally function in HSG cells originated from human submandibular gland ducts, but not in A253 and SGT cells, derived from human submandibular carcinoma and salivary gland adenocarcinoma. However, the underlying mechanism of this suppression has remained elusive. In this study, we examined whether M3R function is suppressed by epigenetic modulation of the receptor. To this end, we investigated the mRNA transcript and protein levels of the M3R using reverse transcriptase-PCR, western blot, and confocal microscopy analyses. Global DNA methylation assays, methylation-specific PCR, and bisulfite sequencing were also performed to understand the epigenetic status of the M3R CpG island. We found that A253 cells expressed all subtypes of muscarinic receptors, except M3R, on the mRNA level. However, treatment of cells with 5-aza-2'-deoxycytidine (5-Aza-CdR), a DNA-demethylating agent, increased the expression levels of both M3R mRNA transcript and protein in proportion to the incubation period. 5-Aza-CdR completely restored the carbachol-induced calcium response, which was not observed in untreated A253 cells. In untreated A253 cells, all CG pairs from the 1st to 14th were methylated and 5-Aza-CdR treatment demethylated one of the methylated CG pairs. We also examined the methylation pattern of M3R CpG island in human cancer tissue. Interestingly, the result was very similar to those of A253 cells. All CG pairs in M3R CpG island were also methylated. Another salivary gland tumor cell line, SGT, also showed the similar methylation pattern, heavy methylation in M3R CpG island. It is concluded that CpG island in M3R is hypermethylated in cancer cell lines and human cancer. Our results further suggest that 5-Aza-CdR could potentially be used to restore the function of M3R, suppressed in some cancer cell types.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic/physiology , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Receptor, Muscarinic M3/metabolism , Salivary Glands/cytology , Amino Acid Sequence , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Blotting, Western , Cell Line , CpG Islands/genetics , DNA Primers/genetics , Decitabine , Humans , Microscopy, Confocal , Molecular Sequence Data , Receptor, Muscarinic M3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Salivary Glands/metabolism , Sequence Analysis, DNA
6.
Biochem Biophys Res Commun ; 466(4): 704-10, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26399685

ABSTRACT

Purinergic receptors, particularly type 7 (P2RX7), are involved in apoptotic cell death. However, the expression and function of P2RX7 are suppressed in HSG cells. In the present study, we explored whether P2RX7 function is regulated by epigenetic alteration of the receptors in two different cell lines, HSG cells derived from human submandibular ducts, and A253 cells, originated from human submandibular carcinoma. We discovered that HSG cells expressed all subtypes of purinergic receptors, excluding P2RX7, at the mRNA level. However, treatment of the cells with 5-Aza-CdR, a DNA demethylating agent, increased the mRNA expression levels of P2RX7 in a time-dependent manner. Furthermore, 5-Aza-CdR completely rescued the calcium response induced by P2RX7 agonist BzATP, a response that was absent in untreated HSG cells. In contrast, A253 cells showed a moderate methylation pattern in the P2RX7 CpG island. Most CG pairs from the first to the 21st were methylated in untreated HSG cells, but 5-Aza-CdR-treatment partially demethylated the methylated CG pairs. We obtained similar results when investigated human tissues; the CG pairs in the P2RX7 CpG islands showed hypermethylation and hypomethylation patterns in human normal and cancer tissues, respectively. Our results suggest that the expression level and function of P2RX7 are regulated by DNA methylation in epithelial cells.


Subject(s)
Epigenesis, Genetic , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Salivary Glands/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Cell Line , CpG Islands , DNA Methylation , Decitabine , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Molecular Sequence Data , Purinergic P2X Receptor Agonists/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Salivary Glands/cytology , Salivary Glands/drug effects
7.
J Biol Chem ; 288(9): 6295-305, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23303189

ABSTRACT

Regulation of intracellular pH is critical for the maintenance of cell homeostasis in response to stress. We used yeast two-hybrid screening to identify novel interacting partners of the pH-regulating transporter NBCe1-B. We identified Hsp70-like stress 70 protein chaperone (STCH) as interacting with NBCe1-B at the N-terminal (amino acids 96-440) region. Co-injection of STCH and NBCe1-B cRNA into Xenopus oocytes significantly increased surface expression of NBCe1-B and enhanced bicarbonate conductance compared with NBCe1-B cRNA alone. STCH siRNA decreased the rate of Na(+)-dependent pHi recovery from NH4(+) pulse-induced acidification in an HSG (human submandibular gland ductal) cell line. We observed that in addition to NBCe1-B, Na(+)/H(+) exchanger (NHE)-dependent pHi recovery was also impaired by STCH siRNA and further confirmed the interaction of STCH with NHE1 but not plasma membrane Ca(2+) ATPase. Both NBCe1-B and NHE1 interactions were dependent on a specific 45-amino acid region of STCH. In conclusion, we identify a novel role of STCH in the regulation of pHi through site-specific interactions with NBCe1-B and NHE1 and subsequent modulation of membrane transporter expression. We propose STCH may play a role in pHi regulation at times of cellular stress by enhancing the recovery from intracellular acidification.


Subject(s)
Cation Transport Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Sodium-Bicarbonate Symporters/metabolism , Sodium-Hydrogen Exchangers/metabolism , Stress, Physiological/physiology , Amino Acid Sequence , Animals , Cation Transport Proteins/genetics , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , HSP70 Heat-Shock Proteins/genetics , Humans , Hydrogen-Ion Concentration , Ion Transport/physiology , Protein Binding , Sodium-Bicarbonate Symporters/genetics , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/genetics , Xenopus
8.
J Spinal Disord Tech ; 27(5): 271-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23897055

ABSTRACT

STUDY DESIGN: A retrospective review. OBJECTIVE: To compare the effectiveness of the posterior cervical foraminotomy (PCF) with and without discectomy for the treatment of cervical disk herniation. SUMMARY OF BACKGROUND DATA: Although PCF is effective and does not require a fusion procedure, it has certain disadvantages, including a narrow operating field, the need for cervical nerve root retraction, and the obstacle of epidural venous bleeding. Surgeons often find that these limitations complicate the removal of the disk material during PCF. METHODS: The study evaluated 135 consecutive patients who underwent PCF for cervical disk herniation and were followed up for a mean of 36.1 months. Of these patients, 117 were treated with posterior foraminotomy with discectomy (group A); the remaining 18 patients were treated with PCF alone because the disk could not be removed (group B). The clinical data, neurological status, radiologic findings, location of pathology, clinical outcomes, and postoperative satisfaction were compared between the 2 groups. RESULTS: The 2 groups had similar clinical outcomes. The mean visual analogue scale (VAS) score for radicular arm and neck pain improved from 7.8 and 5.7 to 4.6 and 3.6 in group A and from 6.6 and 6.2 to 3.4 and 3.6 in group B, and the mean Oswestry disability index (ODI) score from 39.6 and 38.7 to 23.2 and 17.6 in groups A and B, respectively. The success rates in groups A and B were 90.6% and 88.8%, respectively. Radiologic examination found significant differences in operative-level disk softness and disk type between the groups (P<0.05). Protruding, mixed-type, and C4-5 level were more difficult to remove and required more extensive foraminal unroofing. CONCLUSIONS: Despite the surgical and anatomic limitations, PCF with or without discectomy is an effective treatment for cervical disk herniation.


Subject(s)
Cervical Vertebrae/surgery , Decompression, Surgical/methods , Diskectomy/methods , Foraminotomy/methods , Intervertebral Disc Displacement/surgery , Adult , Female , Follow-Up Studies , Humans , Intervertebral Disc/surgery , Male , Middle Aged , Neck Pain/surgery , Postoperative Complications/pathology , Retrospective Studies , Spinal Nerve Roots/surgery , Treatment Outcome
9.
Korean J Physiol Pharmacol ; 18(6): 525-30, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25598668

ABSTRACT

Transient receptor potential vanilloid subtype 1 (TRPV1) was originally found in sensory neurons. Recently, it has been reported that TRPV1 is expressed in salivary gland epithelial cells (SGEC). However, the physiological role of TRPV1 in salivary secretion remains to be elucidated. We found that TRPV1 is expressed in mouse and human submandibular glands (SMG) and HSG cells, originated from human submandibular gland ducts at both mRNA and protein levels. However, capsaicin (CAP), TRPV1 agonist, had little effect on intracellular free calcium concentration ([Ca(2+)]i) in these cells, although carbachol consistently increased [Ca(2+)]i. Exposure of cells to high temperature (>43℃) or acidic bath solution (pH5.4) did not increase [Ca(2+)]i, either. We further examined the role of TRPV1 in salivary secretion using TRPV1 knock-out mice. There was no significant difference in the pilocarpine (PILO)-induced salivary flow rate between wild-type and TRPV1 knock-out mice. Saliva flow rate also showed insignificant change in the mice treated with PILO plus CAP compared with that in mice treated with PILO alone. Taken together, our results suggest that although TRPV1 is expressed in SGEC, it appears not to play any direct roles in saliva secretion via transcellular pathway.

10.
Sci Rep ; 14(1): 11595, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773164

ABSTRACT

Despite growing evidence implicating the calcium-activated chloride channel anoctamin1 (ANO1) in cancer metastasis, its direct impact on the metastatic potential of prostate cancer and the possible significance of epigenetic alteration in this process are not fully understood. Here, we show that ANO1 is minimally expressed in LNCap and DU145 prostate cancer cell lines with low metastatic potential but overexpressed in high metastatic PC3 prostate cancer cell line. The treatment of LNCap and DU145 cells with DNMT inhibitor 5-aza-2'-deoxycytidine (5-Aza-CdR) potentiates ANO1 expression, suggesting that DNA methylation is one of the mechanisms controlling ANO1 expression. Consistent with this notion, hypermethylation was detected at the CpG island of ANO1 promoter region in LNCap and DU145 cells, and 5-Aza-CdR treatment resulted in a drastic demethylation at promoter CpG methylation sites. Upon 5-Aza-CdR treatment, metastatic indexes, such as cell motility, invasion, and metastasis-related gene expression, were significantly altered in LNCap and DU145 cells. These 5-Aza-CdR-induced metastatic hallmarks were, however, almost completely ablated by stable knockdown of ANO1. These in vitro discoveries were further supported by our in vivo observation that ANO1 expression in xenograft mouse models enhances the metastatic dissemination of prostate cancer cells into tibial bone and the development of osteolytic lesions. Collectively, our results help elucidate the critical role of ANO1 expression in prostate cancer bone metastases, which is epigenetically modulated by promoter CpG methylation.


Subject(s)
Anoctamin-1 , Bone Neoplasms , DNA Methylation , Gene Expression Regulation, Neoplastic , Neoplasm Proteins , Promoter Regions, Genetic , Prostatic Neoplasms , Male , Anoctamin-1/metabolism , Anoctamin-1/genetics , Humans , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Bone Neoplasms/secondary , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Mice , CpG Islands , Decitabine/pharmacology , Cell Movement/genetics , Epigenesis, Genetic , Azacitidine/pharmacology
11.
Medicine (Baltimore) ; 103(29): e39051, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39029010

ABSTRACT

The interwoven relationship between gut microbiota and the epigenetic landscape constitutes a pivotal axis in understanding human health and disease. Governed by a myriad of dietary, genetic, and environmental influences, the gut microbiota orchestrates a sophisticated metabolic interplay, shaping nutrient utilization, immune responses, and defenses against pathogens. Recent strides in genomics and metabolomics have shed light on the intricate connections between these microbial influencers and the host's physiological dynamics, presenting a dynamic panorama across diverse disease spectra. DNA methylation and histone modifications, as key players in epigenetics, intricately align with the dynamic orchestration of the gut microbiota. This seamless collaboration, notably evident in conditions like inflammatory bowel disease and obesity, has captured the attention of researchers, prompting an exploration of its nuanced choreography. Nevertheless, challenges abound. Analyzing data is intricate due to the multifaceted nature of the gut microbiota and the limitations of current analytical methods. This underscores the need for a multidisciplinary approach, where diverse disciplines converge to pave innovative research pathways. The integration of insights from microbiome and epigenome studies assumes paramount importance in unraveling the complexities of this intricate partnership. Deciphering the synchronized interactions within this collaboration offers a deeper understanding of these delicate interplays, potentially heralding revolutionary strides in treatment modalities and strategies for enhancing public health.


Subject(s)
Epigenesis, Genetic , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/physiology , DNA Methylation , Obesity/microbiology , Obesity/genetics , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/genetics
12.
Mol Oncol ; 18(8): 2001-2019, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38600695

ABSTRACT

Matrix metalloproteinase 9 (MMP-9) is a member of the MMP family and has been recently identified as a nuclear protease capable of clipping histone H3 N-terminal tails (H3NT). This MMP-9-dependent H3NT proteolysis is critical for establishing an active state of gene transcription during osteoclast differentiation and melanoma development. However, whether H3NT cleavage by MMP-9 plays a similar role in other cellular events has not been explored. Here, we dissect the functional contribution of MMP-9-dependent H3NT clipping to colonic tumorigenesis by using a combination of genome-wide transcriptome data, ChIP/ChIPac-qPCR, CRISPR/dCas9 gene-targeting system, and in vivo xenograft models. We show that MMP-9 is overexpressed in colon cancer cells and catalyzes H3NT proteolysis to drive transcriptional activation of growth stimulatory genes. Our studies using knockdown and inhibition approaches clearly indicate that MMP-9 mediates transcriptional activation and promotes colonic tumorigenesis in a manner dependent on its protease activity toward H3NT. Remarkably, artificial H3NT proteolysis at target gene promoters with dCAS9-MMP-9 is sufficient for establishing their transcriptional competence in colon cancer cells, underscoring the importance of MMP-9-dependent H3NT proteolysis per se in the transactivation process. Our data establish new functions and mechanisms for MMP-9 in driving the oncogenic transcription program in colon cancer through H3NT proteolysis, and demonstrate how this epigenetic pathway can be exploited as a potential therapeutic target for cancer treatment.


Subject(s)
Carcinogenesis , Colonic Neoplasms , Epigenesis, Genetic , Histones , Matrix Metalloproteinase 9 , Proteolysis , Humans , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Histones/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Mice , Transcription, Genetic
13.
Biochim Biophys Acta ; 1822(2): 161-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22137887

ABSTRACT

OBJECTIVES: Primary Sjögren's syndrome (pSS) is a systemic autoimmune disease characterized by lymphocyte infiltration into the salivary and lachrymal glands, leading to dry mouth and eyes. The presence of functional autoantibodies against muscarinic type 3 receptor (M3R) has been reported in pSS patients. However, the pathological role of anti-M3R autoantibodies in pSS salivary dysfunction remains controversial. METHODS: Purified IgGs were obtained from normal (control) and primary SS patients' sera (pSS IgG). Internalization of M3R and clathrin was analyzed by biochemical assay and immunofluorescence confocal microscopy using human submandibular gland (hSMG) cells. Cytoplasmic free Ca(2+) concentration ([Ca(2+)](i)) was measured by microspectrofluorimetry. RESULTS: Incubation of hSMG cells with pSS IgG (1mg/ml) significantly decreased M3R expression levels at the membrane. Carbachol-induced [Ca(2+)](i) transients (CICTs) in these cells were also inhibited by pSS IgG. In contrast to pSS IgG, control IgG had no effect on both the M3R expression level and CICTs. We found that binding of pSS IgG to M3R induces phosphorylation of the receptor, and that the pSS IgG-induced M3R internalization is prevented by the lysosomal inhibitor, chloroquine. In addition, pSS IgG decreased membrane clathrin expression, which was inhibited by atropine. Our immunofluorescence study further confirmed that pSS IgG induces a co-localization of M3R with clathrin and subsequent internalization of M3R. CONCLUSION: pSS IgG induces internalization of M3R partly through a clathrin-mediated pathway. The results suggest M3R internalization as a potential mechanism to explain the exocrinopathy seen in pSS patients.


Subject(s)
Autoantibodies/metabolism , Immunoglobulin G/metabolism , Receptor, Muscarinic M3/metabolism , Sjogren's Syndrome/immunology , Sjogren's Syndrome/metabolism , Adult , Aged , Autoantibodies/blood , Autoantibodies/immunology , Calcium/metabolism , Carbachol/pharmacology , Chloroquine/pharmacology , Clathrin/genetics , Clathrin/metabolism , Down-Regulation , Female , Fluorescent Antibody Technique/methods , Humans , Immunoglobulin G/blood , Microscopy, Confocal/methods , Middle Aged , Phosphorylation/drug effects , Receptor, Muscarinic M3/genetics , Sjogren's Syndrome/blood , Sjogren's Syndrome/genetics , Submandibular Gland/drug effects , Submandibular Gland/immunology , Submandibular Gland/metabolism
14.
Biomedicines ; 11(9)2023 Sep 17.
Article in English | MEDLINE | ID: mdl-37760992

ABSTRACT

Vpr binding protein (VprBP), also known as DDB1- and CUL4-associated factor1 (DCAF1), is a recently identified atypical kinase and plays an important role in downregulating the transcription of tumor suppressor genes as well as increasing the risk for colon and prostate cancers. Melanoma is the most aggressive form of skin cancer arising from pigment-producing melanocytes and is often associated with the dysregulation of epigenetic factors targeting histones. Here, we demonstrate that VprBP is highly expressed and phosphorylates threonine 120 (T120) on histone H2A to drive the transcriptional inactivation of growth-regulatory genes in melanoma cells. As is the case for its epigenetic function in other types of cancers, VprBP acts to induce a gene silencing program dependent on H2AT120 phosphorylation (H2AT120p). The significance of VprBP-mediated H2AT120p is further underscored by the fact that VprBP knockdown- or VprBP inhibitor-induced lockage of H2AT120p mitigates melanoma tumor growth in xenograft models. Collectively, our results establish VprBP-mediated H2AT120p as a key epigenetic signal for melanomagenesis and suggest the therapeutic potential of targeting VprBP kinase activity for effective melanoma treatment.

15.
Res Sq ; 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37293029

ABSTRACT

Background: Melanoma is the most aggressive form of skin cancer arising from pigment-producing melanocytes and is often associated with dysregulation of epigenetic factors targeting histones. VprBP, also known as DCAF1, is a recently identified kinase and plays an important role in downregulating the transcription of tumor suppressor genes as well as increasing the risk for colon and prostate cancers. However, it remains unknown whether VprBP is also involved in triggering the pathogenesis of other types of cancer. Results: We demonstrate that VprBP is highly expressed and phosphorylates threonine 120 (T120) on histone H2A to drive transcriptional inactivation of growth regulatory genes in melanoma cells. As is the case for its epigenetic function in colon and prostate cancers, VprBP acts to induce gene silencing program dependently of H2AT120 phosphorylation (H2AT120p). The significance of VprBP-mediated H2AT120p is further underscored by the fact that VprBP knockdown- or VprBP inhibitor-induced lockage of H2AT120p mitigates melanoma tumor growth in xenograft models. Moreover, artificial tethering of VprBP wild type, but not VprBP kinase-dead mutant, to its responsive genes is sufficient for achieving an inactive transcriptional state in VprBP-depleted cells, indicating that VprBP drives gene silencing program in an H2AT120p-dependent manner. Conclusions: Our results establish VprBP-mediated H2AT120p as a key epigenetic signal for melanomagenesis and suggest the therapeutic potential of targeting VprBP kinase activity for effective melanoma treatment.

16.
Oncogene ; 42(17): 1405-1416, 2023 04.
Article in English | MEDLINE | ID: mdl-37041410

ABSTRACT

VprBP (also known as DCAF1) is a recently identified kinase that is overexpressed in cancer cells and serves as a major determinant for epigenetic gene silencing and tumorigenesis. The role of VprBP in driving target gene inactivation has been largely attributed to its ability to mediate histone H2A phosphorylation. However, whether VprBP also phosphorylates non-histone proteins and whether these phosphorylation events drive oncogenic signaling pathways have not been explored. Here we report that serine 367 phosphorylation (S367p) of p53 by VprBP is a key player in attenuating p53 transcriptional and growth suppressive activities. VprBP catalyzes p53S367p through a direct interaction with the C-terminal domain of p53. Mechanistically, VprBP-mediated S367p inhibits p53 function in the wake of promoting p53 proteasomal degradation, because blocking p53S367p increases p53 protein levels, thereby enhancing p53 transactivation. Furthermore, abrogation of VprBP-p53 interaction by p53 acetylation is critical for preventing p53S367p and potentiating p53 function in response to DNA damage. Together, our findings establish VprBP-mediated S367p as a negative regulator of p53 function and identify a previously uncharacterized mechanism by which S367p modulates p53 stability.


Subject(s)
Signal Transduction , Tumor Suppressor Protein p53 , Humans , Phosphorylation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Phosphotransferases
17.
Nat Commun ; 14(1): 2140, 2023 04 17.
Article in English | MEDLINE | ID: mdl-37069142

ABSTRACT

Our recent work has shown that DCAF1 (also known as VprBP) is overexpressed in colon cancer and phosphorylates histone H2AT120 to drive epigenetic gene inactivation and oncogenic transformation. We have extended these observations by investigating whether DCAF1 also phosphorylates non-histone proteins as an additional mechanism linking its kinase activity to colon cancer development. We now demonstrate that DCAF1 phosphorylates EZH2 at T367 to augment its nuclear stabilization and enzymatic activity in colon cancer cells. Consistent with this mechanistic role, DCAF1-mediated EZH2 phosphorylation leads to elevated levels of H3K27me3 and altered expression of growth regulatory genes in cancer cells. Furthermore, our preclinical studies using organoid and xenograft models revealed that EZH2 requires phosphorylation for its oncogenic function, which may have therapeutic implications for gene reactivation in colon cancer cells. Together, our data define a mechanism underlying DCAF1-driven colonic tumorigenesis by linking DCAF1-mediated EZH2 phosphorylation to EZH2 stability that is crucial for establishing H3K27me3 and gene silencing program.


Subject(s)
Colonic Neoplasms , Enhancer of Zeste Homolog 2 Protein , Histones , Protein Serine-Threonine Kinases , Ubiquitin-Protein Ligases , Humans , Colonic Neoplasms/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Silencing , Genes, Regulator , Histones/genetics , Histones/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism
18.
Oncogene ; 41(4): 560-570, 2022 01.
Article in English | MEDLINE | ID: mdl-34785776

ABSTRACT

Melanoma is a type of skin cancer that develops in pigment-producing melanocytes and often spreads to other parts of the body. Aberrant gene expression has been considered as a crucial step for increasing the risk of melanomagenesis, but how chromatin reorganization contributes to this pathogenic process is still not well understood. Here we report that matrix metalloproteinase 9 (MMP-9) localizes to the nucleus of melanoma cells and potentiates gene expression by proteolytically clipping the histone H3 N-terminal tail (H3NT). From genome-wide studies, we discovered that growth-regulatory genes are selectively targeted and activated by MMP-9-dependent H3NT proteolysis in melanoma cells. MMP-9 cooperates functionally with p300/CBP because MMP-9 cleaves H3NT in a manner that is dependent on p300/CBP-mediated acetylation of H3K18. The functional significance of MMP-9-dependent H3NT proteolysis is further underscored by the fact that RNAi knockdown and small-molecule inhibition of MMP-9 and p300/CBP impede melanomagenic gene expression and melanoma tumor growth. Together, our data establish new functions and mechanisms for nuclear MMP-9 in promoting melanomagenesis and demonstrate how MMP-9-dependent H3NT proteolysis can be exploited to prevent and treat melanoma skin cancer.


Subject(s)
Histones/metabolism , Matrix Metalloproteinase 9/metabolism , Melanoma/genetics , Animals , Humans , Melanoma/pathology , Mice , Proteolysis
19.
Front Psychol ; 12: 678803, 2021.
Article in English | MEDLINE | ID: mdl-35185663

ABSTRACT

Many messengers and social networking services (SNSs) use emojis and stickers as a means of communication. Stickers express individual emotions well, allowing long texts to be replaced with small pictures. As the use of stickers increased, stickers were commercialized on a few platforms and showed remarkable growth as people bought and used stickers with their favorite characters, products, or entertainers online. Depending on their personality, individuals have different motivations for using stickers that determine the usefulness and enjoyment of stickers, affecting their purchase decisions. In the present study, participants (n = 302) who were randomly recruited from a university completed an online questionnaire assessing the Big Five personality characteristics, motivations for using stickers, and the technology acceptance model (TAM). Results using partial least squares structural equation modeling (PLS-SEM) revealed that each personality trait affected different motivations for using stickers. Moreover, motivations for using stickers also influenced different technology acceptance variables. Finally, perceived usefulness, enjoyment, and ease of use had a positive effect on the intention to purchase stickers. This study has implications in that it is an exploratory approach to the intention to purchase stickers, which has been investigated by few prior studies, and it sheds light on the relationship between personality, motivation, and TAM in purchasing stickers. It also suggests that personality and motivation factors can be considered in personalized recommendation services.

20.
Mol Oncol ; 15(10): 2801-2817, 2021 10.
Article in English | MEDLINE | ID: mdl-34312968

ABSTRACT

Histone modification is aberrantly regulated in cancer and generates an unbalanced state of gene transcription. VprBP, a recently identified kinase, phosphorylates histone H2A on threonine 120 (T120) and is involved in oncogenic transcriptional dysregulation; however, its specific role in colon cancer is undefined. Here, we show that VprBP is overexpressed in colon cancer and directly contributes to epigenetic gene silencing and cancer pathogenesis. Mechanistically, the observed function of VprBP is mediated through H2AT120 phosphorylation (H2AT120p)-driven transcriptional repression of growth regulatory genes, resulting in a significantly higher proliferative capacity of colon cancer cells. Our preclinical studies using organoid and xenograft models demonstrate that treatment with the VprBP inhibitor B32B3 impairs colonic tumor growth by blocking H2AT120p and reactivating a transcriptional program resembling that of normal cells. Collectively, our work describes VprBP as a master kinase contributing to the development and progression of colon cancer, making it a new molecular target for novel therapeutic strategies.


Subject(s)
Colonic Neoplasms , Histones , Protein Serine-Threonine Kinases , Ubiquitin-Protein Ligases , Colonic Neoplasms/genetics , Epigenesis, Genetic , Gene Silencing , Histones/metabolism , Humans , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Ubiquitin-Protein Ligases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL