Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Int J Mol Sci ; 21(9)2020 May 09.
Article in English | MEDLINE | ID: mdl-32397561

ABSTRACT

Although various advancements in radical surgery and neoadjuvant chemotherapy have been developed in treating osteosarcoma (OS), their clinical prognosis remains poor. A synthetic chemical compound, 3-hydroxylflavone, that is reported to regulate ROS production is known to inhibit human bone osteosarcoma cells. However, its role and mechanism in human OS cells remains unclear. In this study, we have determined the potential of 3-Hydroxy-2-phenylchromone (3-HF) against OS using human osteosarcoma (HOS) cells. Our previous studies showed that Zipper sterile-alpha-motif kinase (ZAK), a kinase member of the MAP3K family, was involved in various cellular events such as cell proliferation and cell apoptosis, and encoded two transcriptional variants, ZAKα and ß. In this study, we show that 3-HF induces the expression of ZAK and thereby enhances cellular apoptosis. Using gain of function and loss of function studies, we have demonstrated that ZAK activation by 3-HF in OS cells is confined to a ZAKß form that presumably plays a leading role in triggering ZAKα expression, resulting in an aggravated cancer apoptosis. Our results also validate ZAKß as the predominant form of ZAK to drive the anticancer mechanism in HOS cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Neoplasms/pathology , Flavonoids/pharmacology , MAP Kinase Kinase 4/drug effects , MAP Kinase Kinase Kinases/drug effects , Osteosarcoma/pathology , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Gain of Function Mutation , Humans , Loss of Function Mutation , MAP Kinase Kinase Kinases/genetics , Membrane Potential, Mitochondrial/drug effects , Protein Isoforms/drug effects , Protein Isoforms/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
2.
Environ Toxicol ; 34(8): 902-911, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31044527

ABSTRACT

Osteosarcoma (OS) is a tumor entity that can cause a large number of cancer-related deaths. Although chemotherapy can decrease proliferation and increase apoptosis of human OS cells, the clinical prognosis remains poor. Fisetin is a flavonol found in fruits and vegetables and is reported to inhibit cell growth in numerous cancers. But the molecular mechanism underlying fisetin in human OS cells is not clear. It is known that sterile-alpha motif and leucine zipper containing kinase (ZAK), a kinase in the MAP3K family, is involved in various cell processes, including proliferation and apoptosis. In our lab, we have demonstrated that overexpression of ZAK can induce apoptosis in human OS cells. In the previous studies, MAP4K, the upstream of MAP3K, can act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Turning on the Hippo pathway can decrease proliferation and otherwise cause cell apoptosis in cancer cells. In this study, we found that fisetin can upregulate ZAK expression to induce the Hippo pathway and mediate the activation of JNK/ERK, the downstream of ZAK, to trigger cell apoptosis via AP-1 dependent manner in human OS cells. These findings reveal a novel molecular mechanism underlying fisetin effect on human OS cells.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/metabolism , Flavonoids/pharmacology , MAP Kinase Signaling System , Osteosarcoma/metabolism , Protein Kinases/metabolism , Apoptosis , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonols , Hippo Signaling Pathway , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases , Osteosarcoma/enzymology , Osteosarcoma/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factor AP-1/metabolism , Tumor Suppressor Proteins/metabolism
3.
J Cell Biochem ; 119(9): 7855-7864, 2018 09.
Article in English | MEDLINE | ID: mdl-29932238

ABSTRACT

Aberrant expression of leucine zipper- and sterile ɑ motif-containing kinase (ZAK) observed in pathological human myocardial tissue is associated with the progression and elevation of hypertrophy. Our previous reports have correlated high levels of estrogen (E2) and abundant estrogen receptor (ER) α with a low incidence of pathological cardiac-hypertrophy and heart failure in the premenopause female population. However, the effect of elevated ERß expression is not well known yet. Therefore, in this study, we have analyzed the cardioprotective effects and mechanisms of E2 and/or ERß against ZAK overexpression-induced cellular hypertrophy. We have used transient transfection to overexpress ERß into the ZAK tet-on H9c2 cells that harbor the doxycycline-inducible ZAK plasmid. The results show that ZAK overexpression in H9c2 cells resulted in hypertrophic effects, which was correlated with the upregulation of p-JNK and p-p38 MAPKs and their downstream transcription factors c-Jun and GATA-4. However, ERß and E2 with ERß overexpressions totally suppressed the effects of ZAK overexpression and inhibited the levels of p-JNK, p-p38, c-Jun, and GATA-4 effectively. Our results further reveal that ERß directly binds with ZAK under normal conditions; however, ZAK overexpression reduced the association of ZAK-ERß. Interestingly, increase in ERß and E2 along with ERß overexpression both enhanced the binding strengths of ERß and ZAK and reduced the ZAK protein level. ERß overexpression also suppressed the E3 ligase-casitas B-lineage lymphoma (CBL) and attenuated CBL-phosphoinositide 3-kinase (PI3K) protein association to prevent PI3K protein degradation. Moreover, ERß and/or E2 blocked ZAK nuclear translocation via the inhibition of small ubiquitin-like modifier (SUMO)-1 modification. Taken together, our results further suggest that ERß overexpression strongly suppresses ZAK-induced cellular hypertrophy and myocardial damage.


Subject(s)
Estrogen Receptor beta/genetics , Myoblasts, Cardiac/cytology , Protein Kinases/metabolism , SUMO-1 Protein/metabolism , Animals , Cell Enlargement , Cell Line , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Gene Expression Regulation , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/genetics , Proto-Oncogene Proteins c-cbl , Rats
4.
Cell Biochem Funct ; 36(4): 176-182, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29654619

ABSTRACT

ZAK is a novel mixed lineage kinase-like protein that contains a leucine-zipper and a sterile-alpha motif as a protein-protein interaction domain, and it is located in the cytoplasm. There are 2 alternatively spliced forms of ZAK: ZAKα and ZAKß. Previous studies showed that ZAKα is involved in various cell processes, including cell proliferation, cell differentiation, and cardiac hypertrophy, but the molecular mechanism of ZAKß is not yet known. In a recent study in our laboratory, we found that ZAKß can ameliorate the apoptotic effect induced by ZAKα in H9c2 cells. We further hypothesized that ZAKß could also improve the apoptotic effect induced by ZAKα in human osteosarcoma cells. The results of this study show that ZAKß can induce apoptosis and decrease cell viability similar to the effects of ZAKα. Interestingly, our ZAKα-specific inhibitor assay shows that the expression of ZAKß is highly dependent on ZAKα expression. However, ZAKß expression effectively induces ZAKα expression and results in synergistic enhancement of apoptosis in human osteosarcoma cells. Furthermore, co-immunoprecipitation results revealed that ZAKα can directly interact with ZAKß, and this interaction may contribute to the enhanced apoptotic effects. SIGNIFICANCE OF THE STUDY: ZAK is a mixed lineage kinase involved in cell differentiation, proliferation, and hypertrophic growth. ZAKα isoform of ZAK is associated with tumorigenesis, but the function of ZAKß is not yet known. In H9c2 cells, ZAKß was found to ameliorate the apoptotic effect induced by ZAKα. However, in osteosarcoma cells, ZAKß elevates the apoptotic effect induced by ZAKα. In this study, we show that similar to ZAKα, the ZAKß induces apoptosis and decreases cell viability. Interestingly, the expression of ZAKß is dependent on ZAKα expression, and ZAKß further enhances ZAKα expression and results in synergistic enhancement of apoptosis in osteosarcoma cells.


Subject(s)
Apoptosis/drug effects , Osteosarcoma/metabolism , Protein Kinases/biosynthesis , Antibiotics, Antineoplastic/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Drug Synergism , Humans , MAP Kinase Kinase Kinases , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Environ Toxicol ; 33(2): 191-197, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29105997

ABSTRACT

Human osteosarcoma (OS) is a malignant cancer of the bone. It exhibits a characteristic malignant osteoblastic transformation and produces a diseased osteoid. A previous study demonstrated that doxorubicin (DOX) chemotherapy decreases human OS cell proliferation and might enhance the relative RNA expression of ZAK. However, the impact of ZAKα overexpression on the OS cell proliferation that is inhibited by DOX and the molecular mechanism underlying this effect are not yet known. ZAK is a protein kinase of the MAPKKK family and functions to promote apoptosis. In our study, we found that ZAKα overexpression induced an apoptotic effect in human OS cells. Treatment of human OS cells with DOX enhanced ZAKα expression and decreased cancer cell viability while increasing apoptosis of human OS cells. In the meantime, suppression of ZAKα expression using shRNA and inhibitor D1771 both suppressed the DOX therapeutic effect. These findings reveal a novel molecular mechanism underlying the DOX effect on human OS cells. Taken together, our findings demonstrate that ZAKα enhances the apoptotic effect and decreases cell viability in DOX-treated human OS cells.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Doxorubicin/toxicity , Protein Kinases/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , MAP Kinase Kinase Kinases , NF-kappa B/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Protein Kinases/chemistry , Protein Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism , bcl-X Protein/metabolism
6.
Chin J Physiol ; 61(1): 25-34, 2018 Feb 28.
Article in English | MEDLINE | ID: mdl-29374956

ABSTRACT

Sterile alpha motif (SAM)- and leucine-zipper-containing kinase (ZAK) plays a role in the regulation of cell cycle progression and oncogenic transformation. The ZAK gene generates two transcript variants, ZAKα and ZAKß, through alternative splicing. In this study, we identified that ZAKα proteins were upregulated in tumor tissues, whereas ZAKß proteins were mostly expressed in corresponding normal tissues. The ectopically expressed ZAKß proteins in cancer cells inhibited cancer cell proliferation as well as anchorage-independent growth. The ZAKß:ZAKα protein ratio played a role in the regulation of the cyclic adenosine monophosphate (cAMP) signaling pathway, whereas high ZAKß protein levels led to the activation of cAMP response element binding protein 1 (CREB1) and exerted antitumor properties. Overexpression of ZAKß or CREB1 cDNAs in cancer cells inhibited anchorage-independent growth and also reduced the levels of cyclooxygenase 2 (Cox2) and ß-catenin proteins. Cancer cells treated with doxorubicin (Doxo) resulted in the switching from the expression of ZAKα to ZAKß and also inhibited cancer cell growth in soft agar, demonstrating that pharmacological drugs could be used to manipulate endogenous reprogramming splicing events and resulting in the activation of endogenous antitumorigenic properties. We showed that the two ZAK transcript variants, ZAKα and ZAKß, had opposite biological functions in the regulation of tumor cell proliferation in that ZAKß had powerful antitumor properties and that ZAKα could promote tumor growth.


Subject(s)
Neoplasms/prevention & control , Protein Kinases/physiology , Alternative Splicing , Cell Line, Tumor , Cell Proliferation , Cyclic AMP/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Doxorubicin/pharmacology , Humans , MAP Kinase Kinase Kinases , Neoplasms/pathology , Protein Isoforms , Protein Kinases/genetics , Signal Transduction
7.
Int J Med Sci ; 14(12): 1284-1291, 2017.
Article in English | MEDLINE | ID: mdl-29104486

ABSTRACT

Cardiomyopathy involves changes in the myocardial ultra-structure, hypertrophy, apoptosis, fibrosis and inflammation. Angiotensin II (AngII) stimulates the expression of insulin like-growth factors (IGF-2) and IGF-2 receptor (IGF-2R) in H9c2 cardiomyoblasts and subsequently leads to apoptosis. Estrogen receptors protect cardiomyocytes from apoptosis and fibrosis. Tanshinone IIA (TSN), a main active ingredient from Danshen, has been shown to protect cardiomyocytes from death caused by different stress signals. Estrogen receptor α (ER) is required for the rapid activation of the IGF-1R signaling cascade. This study aimed to investigate whether TSN protected H9c2 cardiomyocytes from AngII-induced activation of IGF-2R pathway and hypertrophy via ERs. We found that AngII caused the reduction in IGF-1R phosphorylation and the elevation of ß-catenin and IGF-2R levels. This was reversed by increasing doses of TSN and of caspase-3 and ERK1/2 phosphorylation mediated by ERs. The phytoestrogen significantly attenuated AngII-induced apoptosis and suppressed the subsequent cardiac remodeling effect. Therefore, TSN reduced the AngII-induced activation of ß-catenin and IGF-2R pathways, apoptosis and cardiac remodeling via ERs in H9c2 cardiomyoblasts.


Subject(s)
Abietanes/pharmacology , Angiotensin II/metabolism , Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Myocytes, Cardiac/physiology , Abietanes/therapeutic use , Animals , Cardiomyopathy, Hypertrophic/drug therapy , Cardiomyopathy, Hypertrophic/pathology , Cell Line , Cell Nucleus/metabolism , Drugs, Chinese Herbal/therapeutic use , Humans , Phosphorylation , Protein Transport/drug effects , Rats , Receptor, IGF Type 2/metabolism , Receptors, Estrogen/metabolism , Salvia miltiorrhiza , Signal Transduction/drug effects , beta Catenin/metabolism
8.
Chin J Physiol ; 60(5): 267-274, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-28950690

ABSTRACT

Curcumin, a popular yellow pigment of the dietary spice turmeric, has been reported to inhibit cell growth and to induce apoptosis in a wide variety of cancer cells. Although numerous studies have investigated anticancer effects of curcumin, the precise molecular mechanism of action remains unidentified. Whereas curcumin mediates cell survival and apoptosis through mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) signaling cascades, its impact on the upstream regulation of MAPK is unclear. The leucine-zipper and sterile-α motif kinase alpha (ZAKα), a mitogen-activated protein kinase kinase kinase (MAP3K), activates the c-Jun N-terminal kinase (JNK) and NF-κB pathway. This paper investigated the prospective involvement of ZAKα in curcumin-induced effects on cancer cells. Our results suggest that the antitumor activity of curcumin is mediated via a mechanism involving inhibition of ZAKα activity.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Curcumin/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/physiology , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/analysis , Cyclin-Dependent Kinase Inhibitor p27/analysis , Humans , MAP Kinase Kinase Kinases
9.
Cell Biochem Funct ; 34(8): 606-612, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27859413

ABSTRACT

ZAK (sterile alpha motif and leucine zipper containing kinase AZK), a serine/threonine kinase with multiple biochemical functions, has been associated with various cell processes, including cell proliferation, cell differentiation, and cardiac hypertrophy. In our previous reports, we found that the activation of ZAKα signaling was critical for cardiac hypertrophy. In this study, we show that the expression of ZAKα activated apoptosis through both a FAS-dependent pathway and a mitochondria-dependent pathway by subsequently inducing caspase-3. ZAKß, an isoform of ZAKα, is dramatically expressed during cardiac hypertrophy and apoptosis. The interaction between ZAKα and ZAKß was demonstrated here using immunoprecipitation. The results show that ZAKß has the ability to diminish the expression level of ZAKα. These findings reveal an inherent regulatory role of ZAKß to antagonize ZAKα and to subsequently downregulate the cardiac hypertrophy and apoptosis induced by ZAKα.


Subject(s)
Apoptosis , Cardiomegaly/metabolism , Cardiomegaly/pathology , Protein Kinases/metabolism , Animals , Cell Survival , Intracellular Space/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Protein Binding , Rats , Signal Transduction
10.
J Biomed Sci ; 21: 67, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25091805

ABSTRACT

BACKGROUND: The DEP domain is a globular domain containing approximately 90 amino acids, which was first discovered in 3 proteins: Drosophila disheveled, Caenorhabditis elegans EGL-10, and mammalian Pleckstrin; hence the term, DEP. DEPDC1B is categorized as a potential Rho GTPase-activating protein. The function of the DEP domain in signal transduction pathways is not fully understood. The DEPDC1B protein exhibits the characteristic features of a signaling protein, and contains 2 conserved domains (DEP and RhoGAP) that are involved in Rho GTPase signaling. Small GTPases, such as Rac, CDC42, and Rho, regulate a multitude of cell events, including cell motility, growth, differentiation, cytoskeletal reorganization and cell cycle progression. RESULTS: In this study, we found that it was a guanine nucleotide exchange factor and induced both cell migration in a cultured embryonic fibroblast cell line and cell invasion in cancer cell lines; moreover, it was observed to promote anchorage-independent growth in oral cancer cells. We also demonstrated that DEPDC1B plays a role in regulating Rac1 translocated onto cell membranes, suggesting that DEPDC1B exerts a biological function by regulating Rac1. We examined oral cancer tissue; 6 out of 7 oral cancer tissue test samples overexpressed DEPDC1B proteins, compared with normal adjacent tissue. CONCLUSIONS: DEPDC1B was a guanine nucleotide exchange factor and induced both cell migration in a cultured embryonic fibroblast cell line and cell invasion in cancer cell lines; moreover, it was observed to promote anchorage-independent growth in oral cancer cells. We also demonstrated that DEPDC1B exerts a biological function by regulating Rac1. We found that oral cancer samples overexpressed DEPDC1B proteins, compared with normal adjacent tissue. Suggest that DEPDC1B plays a role in the development of oral cancer. We revealed that proliferation was linked to a novel DEPDC1B-Rac1-ERK1/2 signaling axis in oral cancer cell lines.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cell Proliferation , GTPase-Activating Proteins/metabolism , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mouth Neoplasms/metabolism , Neoplasm Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Female , GTPase-Activating Proteins/genetics , Humans , Male , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Protein Transport/genetics , rac1 GTP-Binding Protein/genetics
11.
Phytother Res ; 28(7): 1096-101, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24399768

ABSTRACT

Puerh tea has been proposed to promote weight loss and favorably modify glucose, insulin and blood lipids. This study tested the effect of daily Puerh tea consumption for 3 months on weight and body mass index (BMI), and select metabolic parameters. The effect of daily Puerh tea intake on weight, BMI and changes in glucose, HbA1c and lipids was evaluated in patients with metabolic syndrome. The patients (N = 70) were randomized into two groups: those taking Puerh tea extract capsule (333 mg Puerh tea extract) three times a day and those taking a placebo tea for 3 months. There was a decrease in body weight of 1.3 kg in the Puerh tea group (p = 0.077) versus 0.23 kg in the placebo arm (p = 0.186). There was also a slight decrease in BMI 0.47 kg/m(2) in the Puerh tea group (p = 0.076) versus 0.09 kg/m(2) in the placebo arm (p = 0.185), suggesting a trend of weight change, but without statistical significance. Subgroup analysis of the male patients demonstrated statistically significant improvements in body weight reduction (p = 0.004) and BMI (p = 0.004). However, the change in other metabolic parameters (cholesterol or triglyceride) or HbA1c was not statistically significant. Intake of Puerh tea for 3 months was associated with a slight reduction in body weight and BMI, especially in the male patients. Therefore, daily Puerh tea consumption may be an alternative choice to modify body weight.


Subject(s)
Body Weight/drug effects , Metabolic Syndrome/drug therapy , Plant Extracts/therapeutic use , Tea/chemistry , Weight Loss , Adult , Aged , Body Composition , Body Mass Index , Cholesterol/blood , Double-Blind Method , Glycated Hemoglobin/metabolism , Humans , Insulin/blood , Male , Metabolic Syndrome/metabolism , Middle Aged , Triglycerides/blood
12.
Exp Cell Res ; 318(14): 1759-66, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22659163

ABSTRACT

Tumor-associated NADH oxidase (tNOX; ENOX2) is a growth-related protein expressed in transformed cells. Consistent with this function, tNOX knockdown by RNA interference leads to a significant reduction in cell proliferation and migration in HeLa cells, whereas tNOX overexpression confers an aggressive phenotype. Here, for the first time, we report that tNOX is phosphorylated by protein kinase Cδ (PKCδ) both in vitro and in vivo. Replacement of serine-504 with alanine significantly reduces phosphorylation by PKCδ. Co-immunoprecipitation experiments reveal an interaction between tNOX and PKCδ. Moreover, whereas overexpression of wild-type tNOX in NIH3T3 cells increases cell proliferation and migration, overexpression of the S504A tNOX mutant leads to diminished cell proliferation and migration, reflecting reduced stability of the unphosphorylatable tNOX mutant protein. Collectively, these results suggest that phosphorylation of serine-504 by PKCδ modulates the biological function of tNOX.


Subject(s)
NADH, NADPH Oxidoreductases/metabolism , Serine/metabolism , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , HEK293 Cells , Humans , Mice , NADH, NADPH Oxidoreductases/genetics , NIH 3T3 Cells , Phosphorylation , Protein Kinase C-delta/metabolism
13.
Curr Oncol ; 30(10): 9004-9018, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37887550

ABSTRACT

The research domain investigating bacterial factors in the development of oral cancer from January 2013 to December 2022 was examined with a bibliometric analysis. A bibliometric analysis is a mathematical and statistical method used to examine extensive datasets. It assesses the connections between prolific authors, journals, institutions, and countries while also identifying commonly used keywords. A comprehensive search strategy identified 167 relevant articles, revealing a progressive increase in publications and citations over time. China and the United States were the leading countries in research productivity, while Harvard University and the University of Helsinki were prominent affiliations. Prolific authors such as Nezar Al-Hebshi, Tsute Chen, and Yaping Pan were identified. The analysis also highlights the contributions of different journals and identifies the top 10 most cited articles in the field, all of which focus primarily on molecular research. The article of the highest citation explored the role of a Fusobacterium nucleatum surface protein in tumor immune evasion. Other top-cited articles investigated the correlation between the oral bacteriome and cancer using 16S rRNA amplicon sequencing, showing microbial shifts associated with oral cancer development. The functional prediction analysis used by recent studies has further revealed an inflammatory bacteriome associated with carcinogenesis. Furthermore, a keyword analysis reveals four distinct research themes: cancer mechanisms, periodontitis and microbiome, inflammation and Fusobacterium, and risk factors. This analysis provides an objective assessment of the research landscape, offers valuable information, and serves as a resource for researchers to advance knowledge and collaboration in the search for the influence of bacteria on the prevention, diagnosis, and treatment of oral cancer.


Subject(s)
Bibliometrics , Mouth Neoplasms , Humans , United States , RNA, Ribosomal, 16S , Bacteria , China
14.
Cell Biochem Biophys ; 80(3): 547-554, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35776316

ABSTRACT

Oxidized low-density lipoprotein (ox-LDL) is a type of modified cholesterol that promotes apoptosis and inflammation and advances the progression of heart failure. Leucine-zipper and sterile-α motif kinase (ZAK) is a kinase of the MAP3K family which is highly expressed in the heart and encodes two variants, ZAKα and ZAKß. Our previous study serendipitously found opposite effects of ZAKα and ZAKß in which ZAKß antagonizes ZAKα-induced apoptosis and hypertrophy of the heart. This study aims to test the hypothesis of whether ZAKα and ZAKß are involved in the damaging effects of ox-LDL in the cardiomyoblast. Cardiomyoblast cells H9c2 were treated with different concentrations of ox-LDL. Cell viability and apoptosis were measured by MTT and TUNEL assay, respectively. Western blot was used to detect apoptosis, hypertrophy, and pro-survival signaling proteins. Plasmid transfection, pharmacological inhibition with D2825, and siRNA transfection were utilized to upregulate or downregulate ZAKß, respectively. Ox-LDL concentration-dependently reduces the viability and expression of several pro-survival proteins, such as phospho-PI3K, phospho-Akt, and Bcl-xL. Furthermore, ox-LDL increases cleaved caspase-3, cleaved caspase-9 as indicators of apoptosis and increases B-type natriuretic peptide (BNP) as an indicator of hypertrophy. Overexpression of ZAKß by plasmid transfection attenuates apoptosis and prevents upregulation of BNP. Importantly, these effects were abolished by inhibiting ZAKß either by D2825 or siZAKß application. Our results suggest that ZAKß upregulation in response to ox-LDL treatment confers protective effects on cardiomyoblast.


Subject(s)
Lipoproteins, LDL , Natriuretic Peptide, Brain , Animals , Apoptosis , Hypertrophy , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Natriuretic Peptide, Brain/genetics , Protein Kinases , Rats , Up-Regulation
15.
Biochem Biophys Res Commun ; 414(2): 337-43, 2011 Oct 22.
Article in English | MEDLINE | ID: mdl-21951852

ABSTRACT

Our previous study shows that caveolin-1 colocalizes and interacts with ATP-binding cassette transporter A1 (ABCA1), which is intimately involved in cellular cholesterol efflux. In this study, we further clarified the region of caveolin-1 that interacts with ABCA1. We also examined the interaction between mutant caveolin-1 and ABCA1 in HDL-mediated cholesterol efflux. We constructed a panel of mutant caveolin-1 proteins and co-transfected them into rat aortic endothelial and human embryonic kidney 293 (HEK293) cells. The co-immunoprecipitation shows that mutant oligomerization domain of caveolin-1, caveolin-1(Δ62-100), is required for the interaction of caveolin-1 with ABCA1. Caveolin-1(Δ62-100) did not colocalize with ABCA1 in the cholesterol-loaded cells after HDL incubation as observed by immunofluorescence confocal microscopy. Concomitantly, caveolin-1(Δ62-100) suppressed HDL-mediated cholesterol efflux. The results suggest that the region of caveolin-1 between amino acids 62 and 100 is an oligomerization domain as well as an attachment site for ABCA1 interaction that regulates HDL-mediated cholesterol efflux.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Caveolin 1/metabolism , Cholesterol/metabolism , Lipoproteins, HDL/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Caveolin 1/genetics , Cells, Cultured , HEK293 Cells , Humans , Lipoproteins, HDL/pharmacology , Rats , Rats, Sprague-Dawley , Sequence Deletion
16.
Cancer Sci ; 101(6): 1374-81, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20331627

ABSTRACT

Novel mixed-lineage kinase protein zipper sterile-alpha-motif kinase (ZAK) was first cloned by our laboratory. Lung cancer is the leading cause of cancer-related death in the world, including in Taiwan. Here, we wanted to investigate whether ZAK plays a potential role in lung cancer development. First, Western blot analysis results demonstrated that four cell lines expressed high levels of ZAK from among a panel of 10 lung cancer cell lines, and two of three normal lung cells expressed ZAK. ZAK gene expressions were down-regulated in lung cancers by real-time PCR analysis. Overexpression of ZAK suppressed cell proliferation in parallel with increased phosphorylated levels of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK). In contrast, ZAK silencing cells inhibited the expressions of phosphorylated ERK and JNK without affecting the expression of phosphorylated p38. The effect of the decreased cell growth rate was significantly but incompletely reversed when ZAK-overexpressing cells were treated with a specific ERK or JNK inhibitor. Moreover, c-Fos and c-Jun, the major downstream components of MAPKs, were up-regulated by ERK and JNK, respectively. When ZAK-overexpressing cells introduced with c-Jun RNA interference (RNAi), the activator protein-1 (AP-1) transcription activity detected by a secreted alkaline phosphatase (SEAP) assay was suppressed and the decreased cell number was reversed compared with the control RNAi-treated group. More importantly, ZAK significantly depressed tumor growth in in vivo study. Taken together, results from both in vitro and in vivo studies indicated that the decrease of lung cancer cell proliferation by ZAK may involve the ERK and JNK pathways via an AP-1 transcription factor.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , JNK Mitogen-Activated Protein Kinases/physiology , Lung Neoplasms/prevention & control , Protein Kinases/physiology , Transcription Factor AP-1/physiology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Lung Neoplasms/pathology , MAP Kinase Kinase Kinases , MAP Kinase Signaling System , Male , Mice , Middle Aged , Protein Kinases/genetics
17.
Biochem Biophys Res Commun ; 391(1): 698-702, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19945436

ABSTRACT

In this report, we identified the novel protein heart protein phosphatase 1-binding protein (Hepp1), encoded by FLJ23654. Hepp1 associated with protein phosphatase 1 (PP1) by yeast two-hybrid, GST pull-down, co-immunoprecipitation, and far Western blotting assays. Northern blot analysis revealed that Hepp1 mRNA was only expressed in human heart and testis. Recombinant Hepp1 slightly enhanced the enzymatic activity of PP1 and antagonized the ability of phospho-inhibitor-1 or inhibitor-2 to inhibit PP1. Hepp1 protein in human heart tissues was detected by Western blot analysis. Together, our data suggest that Hepp1 can play a role in cardiac functions by working in concert with PP1.


Subject(s)
Myocardium/metabolism , Protein Phosphatase 1/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Humans , Male , Molecular Sequence Data , Oncogene Proteins , Protein Phosphatase 1/antagonists & inhibitors , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Testis/metabolism , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases
18.
J Biomed Sci ; 16: 11, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-19272173

ABSTRACT

We found that overexpression of RhoGDIbeta, a Rho GDP dissociation inhibitor, induced hypertrophic growth and suppressed cell cycle progression in a cultured cardiomyoblast cell line. Knockdown of RhoGDIbeta expression by RNA interference blocked hypertrophic growth. We further demonstrated that RhoGDIbeta physically interacts with ZAK and is phosphorylated by ZAK in vitro, and this phosphorylation negatively regulates RhoGDIbeta functions. Moreover, the ZAK-RhoGDIbeta interaction may maintain ZAK in an inactive hypophosphorylated form. These two proteins could negatively regulate one another such that ZAK suppresses RhoGDIbeta functions through phosphorylation and RhoGDIbeta counteracts the effects of ZAK by physical interaction. Knockdown of ZAK expression in ZAK- and RhoGDIbeta-expressing cells by ZAK-specific RNA interference restored the full functions of RhoGDIbeta.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , Hypertrophy/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Cycle/physiology , Cell Line , Cell Movement/physiology , Guanine Nucleotide Dissociation Inhibitors/genetics , Humans , Hypertrophy/pathology , Leucine Zippers , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , rho-Specific Guanine Nucleotide Dissociation Inhibitors
19.
J Biomed Sci ; 16: 56, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19538723

ABSTRACT

RhoGDIbeta, a Rho GDP dissociation inhibitor, induced hypertrophic growth and cell migration in a cultured cardiomyoblast cell line, H9c2. We demonstrated that RhoGDIbeta plays a previously undefined role in regulating Rac1 expression through transcription to induce hypertrophic growth and cell migration and that these functions are blocked by the expression of a dominant-negative form of Rac1. We also demonstrated that knockdown of RhoGDIbeta expression by RNA interference blocked RhoGDIbeta-induced Rac1 expression and cell migration. We demonstrated that the co-expression of ZAK and RhoGDIbeta in cells resulted in an inhibition in the activity of ZAK to induce ANF expression. Knockdown of ZAK expression in ZAK-RhoGDIbeta-expressing cells by ZAK-specific RNA interference restored the activities of RhoGDIbeta.


Subject(s)
Cell Movement , Guanine Nucleotide Dissociation Inhibitors/metabolism , Protein Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Enlargement , Cells, Cultured , Guanine Nucleotide Dissociation Inhibitors/genetics , Rats , Transfection , rho-Specific Guanine Nucleotide Dissociation Inhibitors
20.
Mol Cell Biochem ; 325(1-2): 69-77, 2009 May.
Article in English | MEDLINE | ID: mdl-19184368

ABSTRACT

Leucine-zipper and sterile-alpha motif kinase (ZAK) is the key intra-cellular mediator protein in cardiomyocyte hypertrophy induction by transforming growth factor beta 1 (TGF-beta1) which has also been identified as a profibrotic cytokine involved in cardiac fibrosis progression. We hypothesized whether ZAK over-expression causes cardiac scar formation due to the extra-cellular matrix (ECM) degraded enzyme regulation in this paper. Using immuno-histochemical analysis of the human cardiovascular tissue array, we found a positively significant association between ZAK over-expression and myocardial scars. ZAK over-expression in H9c2 cardiomyoblast cells increases the metalloproteinase tissue inhibitor 1/2 (TIMP-1/2) protein level, which reduces matria metalloproteinase-9 (MMP-9) activity and also activates c-JNK N-terminal kinase 1/2 (JNK1/2) and p38 signaling, which induces MMP-2, possibly resulting in cardiac fibrosis. Taken together, ZAK activity inhibition may be a good strategy to prevent the cardiac fibrosis progression.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Myocardium/metabolism , Protein Kinases/physiology , Signal Transduction , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Immunohistochemistry , MAP Kinase Signaling System , Myocardium/cytology , Myocardium/enzymology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL