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

Country/Region as subject
Publication year range
1.
Mol Cell ; 69(2): 279-291.e5, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29351847

ABSTRACT

Sustained energy starvation leads to activation of AMP-activated protein kinase (AMPK), which coordinates energy status with numerous cellular processes including metabolism, protein synthesis, and autophagy. Here, we report that AMPK phosphorylates the histone methyltransferase EZH2 at T311 to disrupt the interaction between EZH2 and SUZ12, another core component of the polycomb repressive complex 2 (PRC2), leading to attenuated PRC2-dependent methylation of histone H3 at Lys27. As such, PRC2 target genes, many of which are known tumor suppressors, were upregulated upon T311-EZH2 phosphorylation, which suppressed tumor cell growth both in cell culture and mouse xenografts. Pathologically, immunohistochemical analyses uncovered a positive correlation between AMPK activity and pT311-EZH2, and higher pT311-EZH2 correlates with better survival in both ovarian and breast cancer patients. Our finding suggests that AMPK agonists might be promising sensitizers for EZH2-targeting cancer therapies.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Animals , Carcinogenesis/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , DNA Methylation , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/physiology , Epigenesis, Genetic , Female , Histones/metabolism , Humans , Mice , Neoplasm Proteins , Nuclear Proteins/metabolism , Oncogenes , Ovarian Neoplasms/metabolism , Phosphorylation , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/physiology , Transcription Factors , Up-Regulation
2.
Mol Med ; 30(1): 73, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822233

ABSTRACT

Human malignant pleural mesothelioma (hMPM) is an aggressive, rare disease with a poor prognosis. Histologically, MPM is categorized into epithelioid, biphasic, and sarcomatoid subtypes, with the epithelioid subtype generally displaying a better response to treatment. Conversely, effective therapies for the non-epithelioid subtypes are limited. This study aimed to investigate the potential role of FK228, a histone deacetylase inhibitor, in the suppression of hMPM tumor growth. We conducted a comprehensive analysis of the histological and molecular characteristics of two MPM cell lines, CRL-5820 (epithelioid) and CRL-5946 (non-epithelioid). CRL-5946 cells and non-epithelioid patient-derived xenografted mice exhibited heightened growth rates compared to those with epithelioid MPM. Both CRL-5946 cells and non-epithelioid mice displayed a poor response to cisplatin. However, FK228 markedly inhibited the growth of both epithelioid and non-epithelioid tumor cells in vitro and in vivo. Cell cycle analysis revealed FK228-induced G1/S and mitotic arrest in MPM cells. Caspase inhibitor experiments demonstrated that FK228-triggered apoptosis occurred via a caspase-dependent pathway in CRL-5946 but not in CRL-5820 cells. Additionally, a cytokine array analysis showed that FK228 reduced the release of growth factors, including platelet-derived and vascular endothelial growth factors, specifically in CRL-5946 cells. These results indicate that FK228 exhibits therapeutic potential in MPM by inducing cytotoxicity and modulating the tumor microenvironment, potentially benefiting both epithelioid and non-epithelioid subtypes.


Subject(s)
Apoptosis , Cell Proliferation , Depsipeptides , Mesothelioma, Malignant , Mesothelioma , Xenograft Model Antitumor Assays , Humans , Animals , Mesothelioma, Malignant/drug therapy , Mesothelioma, Malignant/pathology , Cell Line, Tumor , Mice , Mesothelioma/drug therapy , Mesothelioma/pathology , Apoptosis/drug effects , Cell Proliferation/drug effects , Depsipeptides/pharmacology , Depsipeptides/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Pleural Neoplasms/drug therapy , Pleural Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Female , Epithelioid Cells/pathology , Cell Cycle/drug effects
3.
Curr Issues Mol Biol ; 45(6): 4518-4528, 2023 May 24.
Article in English | MEDLINE | ID: mdl-37367035

ABSTRACT

Among genetically engineered mouse models of breast cancer, MMTV-PyVT is a mouse strain in which the oncogenic polyoma virus middle T antigen is driven by the mouse mammary tumor virus promoter. The aim of the present study was to perform morphologic and genetic analyses of mammary tumors arising from MMTV-PyVT mice. To this end, mammary tumors were obtained at 6, 9, 12, and 16 weeks of age for histology and whole-mount analyses. We conducted whole-exome sequencing to identify constitutional and tumor-specific mutations, and genetic variants were identified using the GRCm38/mm10 mouse reference genome. Using hematoxylin and eosin analysis and whole-mount carmine alum staining, we demonstrated the progressive proliferation and invasion of mammary tumors. Frameshift insertions/deletions (indels) were noted in the Muc4. Mammary tumors showed small indels and nonsynonymous single-nucleotide variants but no somatic structural alterations or copy number variations. In summary, we validated MMTV-PyVT transgenic mice as a multistage model for mammary carcinoma development and progression. Our characterization may be used as a reference for guidance in future research.

4.
Mol Carcinog ; 62(12): 1935-1946, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37642311

ABSTRACT

Cellular retinoic acid binding protein 1 (CRABP1) participates in the regulation of retinoid signaling. Previous studies showed conflicting results regarding the role of CRABP1 in tumor biology, including protumorigenic and tumor-suppressive effects in different types of cancer. Our bioinformatics analyses suggested that CRABP1 expression was downregulated in thyroid cancer. Ectopic expression of CRABP1 in thyroid cancer cells suppressed migratory and invasive activity without affecting cell growth or cell cycle distribution. In transformed normal thyroid follicular epithelial cells, silencing of CRABP1 expression increased invasiveness. Additionally, CRABP1 overexpression was associated with downregulation of the mesenchymal phenotype. Kinase phosphorylation profiling indicated that CRABP1 overexpression was accompanied by a decrease in phosphorylation of epidermal growth factor (EGF) receptor and downstream phosphorylation of Akt, STAT3, and FAK, which were reversed by exogenous EGF treatment. Immunohistochemical analysis of our tissue microarrays revealed an inverse association between CRABP1 expression and disease stage of differentiated thyroid cancer. Taken together, our results suggest that CRABP1 expression is aberrantly lost in thyroid cancer, and this downregulation promotes the epithelial-mesenchymal transition at least partly through modulating EGF receptor signaling.


Subject(s)
Epidermal Growth Factor , Thyroid Neoplasms , Humans , Epidermal Growth Factor/metabolism , Down-Regulation , Epithelial-Mesenchymal Transition , Cell Line, Tumor , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism
5.
J Fish Biol ; 103(3): 593-602, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37269234

ABSTRACT

Uropterygius concolor Rüppell, the type species of the genus Uropterygius, is a small, uniformly brown moray considered to be widely distributed in the Indo-Pacific region. However, a recent study indicated that the real U. concolor is currently known only from the type locality in the Red Sea, and species recorded outside the Red Sea may represent a species complex that comprises several species. In this study, we assess the genetic and morphological variations of this species complex based on available data. Analyses of cytochrome c oxidase subunit I sequences revealed at least six distinct genetic lineages recognized under 'U. concolor'. After carefully comparing the morphologies, one of the lineages is described herein as a new species, Uropterygius mactanensis sp. nov., based on 21 specimens collected from Mactan Island, Cebu, Philippines. Another distinct lineage is considered to be a possibly undescribed species based on diagnostic morphological characters. Although the taxonomic status of junior synonyms of U. concolor and some lineages still remain unresolved, this study provides informative morphological characters (i.e., tail length, trunk length, vertebrae number, and arrangement of teeth) that can be used in future studies on this species complex.


Subject(s)
Eels , Animals , Eels/anatomy & histology , Philippines , Indian Ocean , Phylogeny
6.
Molecules ; 28(2)2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36677579

ABSTRACT

It is both important and required to quickly and accurately detect chemical warfare agents, such as the highly toxic nerve agent sarin. Surface-enhanced Raman scattering (SERS) has received considerable attention due to its rapid results, high sensitivity, non-destructive data acquisition, and unique spectroscopic fingerprint. In this work, we successfully prepared SERS cotton swabs (CSs) for the detection of the sarin simulant agent dimethyl methyl phosphonate (DMMP) by anchoring N1-(3-trimethoxysilylpropyl) diethylenetriamine (ATS)/silver nanoparticle (AgNP) nanocomposites on CSs using ATS as the stabilizer and coupling agent. Simultaneously, the binding mode and reaction mechanics between the AgNP, ATS, and CS were confirmed by XPS. The modified CSs exhibited good uniformity, stability, and adsorption capability for SERS measurements, enabling the adsorption and detection of DMMP residue from an irregular surface via a simple swabbing process, with a detection limit of 1 g/L. The relative standard deviations (RSDs) of RSD710 = 5.6% had high reproducibility. In this research, the fabrication method could easily be extended to other cellulose compounds, such as natural fibers and paper. Furthermore, the versatile SERS CSs can be used for the on-site detection of DMMP, particularly in civil and defense applications, to guarantee food security and the health of the population.

7.
Int J Mol Sci ; 23(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35163585

ABSTRACT

BACKGROUND: The treatment of non-small-cell lung cancer (NSCLC) involves platinum-based chemotherapy. It is typically accompanied by chemoresistance resulting from antioxidant properties conferred by cancer stem cells (CSCs). Human epidermal growth factor receptor 2 (HER2) enhances CSCs and antioxidant properties in cancers, including NSCLC. METHODS: Here, we elucidated the role of histamine N-methyltransferase (HNMT), a histamine metabolism enzyme significantly upregulated in NSCLC and coexpressed with HER2. HNMT expression in lung cancer tissues was determined using quantitative reverse transcription PCR (RT-qPCR). A publicly available dataset was used to determine HNMT's potential as an NSCLC target molecule. Immunohistochemistry and coimmunoprecipitation were used to determine HNMT-HER2 correlations and interactions, respectively. HNMT shRNA and overexpression plasmids were used to explore HNMT functions in vitro and in vivo. We also examined miRNAs that may target HNMT and investigated HNMT/HER2's role on NSCLC cells' antioxidant properties. Finally, how HNMT loss affects NSCLC cells' sensitivity to cisplatin was investigated. RESULTS: HNMT was significantly upregulated in human NSCLC tissues, conferred a worse prognosis, and was coexpressed with HER2. HNMT depletion and overexpression respectively decreased and increased cell proliferation, colony formation, tumorsphere formation, and CSCs marker expression. Coimmunoprecipitation analysis indicated that HNMT directly interacts with HER2. TARGETSCAN analysis revealed that HNMT is a miR-223 and miR-3065-5p target. TBHp treatment increased HER2 expression, whereas shHNMT disrupted the Nuclear factor erythroid 2-related factor 2 (Nrf2)/ hemeoxygenase-1 (HO-1)/HER2 axis and increased reactive oxygen species accumulation in NSCLC cells. Finally, shHNMT sensitized H441 cells to cisplatin treatment in vitro and in vivo. CONCLUSIONS: Therefore, HNMT upregulation in NSCLC cells may upregulate HER2 expression, increasing tumorigenicity and chemoresistance through CSCs maintenance and antioxidant properties. This newly discovered regulatory axis may aid in retarding NSCLC progression and chemoresistance.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histamine N-Methyltransferase/biosynthesis , Lung Neoplasms/enzymology , Neoplastic Stem Cells/enzymology , Oxidative Stress , Receptor, ErbB-2/metabolism , Up-Regulation , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Female , Histamine N-Methyltransferase/genetics , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Receptor, ErbB-2/genetics
8.
Acta Neurol Taiwan ; 2022 Sep 18.
Article in English | MEDLINE | ID: mdl-35040109

ABSTRACT

Here we present a case of Japanese encephalitis with an interesting MRI image. The patient is a previously healthy 27 years old male living around a hog farm. Initially, he went to a local hospital and was treated with Levofloxacin as a pneumonia infection. He presented with fever and headache for two days before he sought medical assistance. For two days, his symptoms didn't improve, and progressive consciousness declining was noted. Hence the family decided to transfer to our hospital for further evaluation. On examination, his consciousness was stupor, cannot obey orders, and febrile. The pupils were equal with preserved light reflex. His muscle powers were symmetric bilaterally near his baseline. CSF examination showed normal opening pressure, elevated WBC count with 196 nucleated cells/mm3, normal glucose, and elevated protein level. Brain MRI showed left medial thalamic hyperintensity on T2WI and DWI (Figure 1). Finally, the patient was diagnosed with Japanese encephalitis based on the positive result of the Nucleic acid amplification test. The patient received supportive care with a gradual recovery of his consciousness and became able to obey commands. However, subtle learning problems persisted after one week. Based on the literature review, the MRI or CT finding on thalamic lesions on imaging has high specificity, which could be an assistance tool diagnosis of Japanese encephalitis.[1] The typical Japanese MRI feature consists of hyperintense lesions on T2WI or DWI, and the thalamus was the most commonly involved region. [2][3][4] Although the majority of Japanese encephalitis had bilateral thalamic lesions, the unilateral lesion is uncommon. [4][5] Thus the case presented here provides a rare image of reference for Japanese encephalitis with a unilateral thalamic lesion Reference 1. Dung NM, et al. An evaluation of the usefulness of neuroimaging for the diagnosis of Japanese encephalitis. J Neurol. 2009;256(12): 2052-60. 2. Maschke M, et al. Update on neuroimaging in infectious central nervous system disease. Curr Opin Neurol. 2004;17(4):475-80. 3. Sunwoo, J.-S., et al., Clinical Characteristics of Severe Japanese Encephalitis: A Case Series from South Korea. The American journal of tropical medicine and hygiene, 2017. 97(2): p. 369-375. 4. Phukan, P., et al., MRI Spectrum of Japanese Encephalitis in Northeast India: A Cross-Sectional Study. Journal of neurosciences in rural practice, 2021. 12(2): p. 281-289. 5. Yakushiji, Y., et al., [A case of Japanese encephalitis presenting with unilateral lesions in diffusion-weighted MRI]. Rinsho Shinkeigaku, 2001. 41(9): p. 602-5.

9.
Invest New Drugs ; 39(6): 1493-1506, 2021 12.
Article in English | MEDLINE | ID: mdl-34031786

ABSTRACT

Background Human 3ß-hydroxysteroid dehydrogenase type 1 (HSD3B1) is an enzyme associated with steroidogenesis, however its' role in hepatocellular carcinoma (HCC) biology is unknown. Trilostane is an inhibitor of HSD3B1 and has been tested as a treatment for patients with breast cancer but has not been studied in patients with HCC. Methods and Results The expression of HSD3B1 in HCC tumors in 57 patients were examined. A total of 44 out of 57 tumors (77.2%) showed increased HSD3B1 expression. The increased HSD3B1 in tumors was significantly associated with advanced HCC. In vitro, the knockdown of HSD3B1 expression in Mahlavu HCC cells by a short hairpin RNA (shRNA) led to significant decreases in colony formation and cell migration. The suppression of clonogenicity in the HSD3B1-knockdown HCC cells was reversed by testosterone and 17ß-estradiol. Trilostane-mediated inhibition of HSD3B1 in different HCC cells also caused significant inhibition of clonogenicity and cell migration. In subcutaneous HCC Mahlavu xenografts, trilostane (30 or 60 mg/kg, intraperitoneal injection) significantly inhibited tumor growth in a dose-dependent manner. Furthermore, the combination of trilostane and sorafenib significantly enhanced the inhibition of clonogenicity and xenograft growth, surpassing the effects of each drug used alone, with no documented additional toxicity to animals. HSD3B1 blockade was found to suppress the phosphorylation of extracellular signal-regulated kinase (ERK). The decreased ERK phosphorylation was reversed by testosterone or 17b-estradiol. Conclusions Trilostane significantly inhibited the growth of HCC by inhibiting HSD3B1 function and augmenting the efficacy of sorafenib.


Subject(s)
Carcinoma, Hepatocellular/pathology , Dihydrotestosterone/analogs & derivatives , Liver Neoplasms/pathology , Multienzyme Complexes/antagonists & inhibitors , Progesterone Reductase/antagonists & inhibitors , Sorafenib/pharmacology , Steroid Isomerases/antagonists & inhibitors , Aged , Animals , Cell Line, Tumor , Cell Movement/drug effects , Dihydrotestosterone/administration & dosage , Dihydrotestosterone/pharmacology , Drug Therapy, Combination , Estradiol/pharmacology , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , RNA, Small Interfering/drug effects , Sorafenib/administration & dosage , Testosterone/pharmacology , Xenograft Model Antitumor Assays
10.
Toxicol Appl Pharmacol ; 422: 115493, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33727089

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) has a more aggressive phenotype and poorer prognosis than hormone receptor (HR+) and human epidermal growth factor receptor (HER2 -) subtypes. Inhibition of cyclin-dependent kinase (CDK)4 and CDK6 was successful in patients with advanced metastatic HR+/HER2- breast cancer, but those with TNBC exhibited low or no response to this therapeutic approach. This study investigated the dual therapeutic targeting of CDK2 and CDK4 by using 4-acetyl-antroquinonol B (4-AAQB) against TNBC cells. METHODS: We examined the effects of CDK2, CDK4, and CDK6 inhibition through 4-AAQB treatment on TNBC cell lines and established an orthotropic xenograft mouse model to confirm the in vitro results of inhibiting CDK2, CDK4, and CDK6 by 4-AAQB treatment. RESULTS: High expression and alteration of CDK2 and CDK4 but not CDK6 significantly correlated with poor overall survival of patients with breast cancer. CDK2 and CDK4 were positively correlated with damage in DNA replication and repair pathways. Docking results indicated that 4-AAQB was bound to CDK2 and CDK4 with high affinity. Treatment of TNBC cells with 4-AAQB suppressed the expression of CDK2 and CDK4 in vitro. Additionally, 4-AAQB induced cell cycle arrest, DNA damage, and apoptosis in TNBC cells. In vivo study results confirmed that the anticancer activity of 4-AAQB suppressed tumor growth through the inhibition of CDK2 and CDK4. CONCLUSION: The expression level of CDK2 and CDK4 and DNA damage response (DDR) signaling are prominent in TNBC cell cycle regulation. Thus, 4-AAQB is a potential agent for targeting CDK2/4 and DDR in TNBC cells.


Subject(s)
4-Butyrolactone/analogs & derivatives , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclohexanones/pharmacology , DNA Damage , DNA Repair/drug effects , Triple Negative Breast Neoplasms/drug therapy , 4-Butyrolactone/pharmacology , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 4/genetics , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred NOD , Mice, SCID , Signal Transduction , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Nature ; 527(7576): 100-104, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26479035

ABSTRACT

The development of life-threatening cancer metastases at distant organs requires disseminated tumour cells' adaptation to, and co-evolution with, the drastically different microenvironments of metastatic sites. Cancer cells of common origin manifest distinct gene expression patterns after metastasizing to different organs. Clearly, the dynamic interaction between metastatic tumour cells and extrinsic signals at individual metastatic organ sites critically effects the subsequent metastatic outgrowth. Yet, it is unclear when and how disseminated tumour cells acquire the essential traits from the microenvironment of metastatic organs that prime their subsequent outgrowth. Here we show that both human and mouse tumour cells with normal expression of PTEN, an important tumour suppressor, lose PTEN expression after dissemination to the brain, but not to other organs. The PTEN level in PTEN-loss brain metastatic tumour cells is restored after leaving the brain microenvironment. This brain microenvironment-dependent, reversible PTEN messenger RNA and protein downregulation is epigenetically regulated by microRNAs from brain astrocytes. Mechanistically, astrocyte-derived exosomes mediate an intercellular transfer of PTEN-targeting microRNAs to metastatic tumour cells, while astrocyte-specific depletion of PTEN-targeting microRNAs or blockade of astrocyte exosome secretion rescues the PTEN loss and suppresses brain metastasis in vivo. Furthermore, this adaptive PTEN loss in brain metastatic tumour cells leads to an increased secretion of the chemokine CCL2, which recruits IBA1-expressing myeloid cells that reciprocally enhance the outgrowth of brain metastatic tumour cells via enhanced proliferation and reduced apoptosis. Our findings demonstrate a remarkable plasticity of PTEN expression in metastatic tumour cells in response to different organ microenvironments, underpinning an essential role of co-evolution between the metastatic cells and their microenvironment during the adaptive metastatic outgrowth. Our findings signify the dynamic and reciprocal cross-talk between tumour cells and the metastatic niche; importantly, they provide new opportunities for effective anti-metastasis therapies, especially of consequence for brain metastasis patients.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/secondary , Exosomes/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , MicroRNAs/genetics , PTEN Phosphohydrolase/deficiency , Tumor Microenvironment , Adaptation, Physiological/genetics , Animals , Astrocytes/cytology , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Calcium-Binding Proteins , Cell Proliferation/genetics , Chemokine CCL2/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Evolution, Molecular , Exosomes/metabolism , Female , Genes, Tumor Suppressor , Humans , Male , Mice , Microfilament Proteins , PTEN Phosphohydrolase/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Tumor Microenvironment/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
12.
J Fish Biol ; 97(5): 1462-1480, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32844437

ABSTRACT

Strophidon McClelland is a muraenid genus with characteristic appearance of a very elongated body, a large mouth cleft and anteriorly placed eyes. The nomenclature and taxonomic history of species within Strophidon are contentious and its members are easily misidentified. In the present study, species of the genus Strophidon are revised based on morphological and molecular data, and five species are considered valid, including S. dawydoffi Prokofiev, S. dorsalis (Seale), S. sathete (Hamilton), S. ui Tanaka and a new species, S. tetraporus. Strophidon tetraporus sp. nov. is described based on 15 specimens from Indonesia, the Philippines, Taiwan and Vietnam with the unique characteristic of the constant presence of the fourth infraorbital pore among species of Strophidon. The intraspecific variation of vertebral formula within S. dorsalis is discussed based on molecular data. Muraena macrurus Bleeker and Thyrsoidea longissima Kaup are synonyms of S. sathete that can be distinguished from the most similar congener S. ui by a longer tail, smaller eyes and more inner maxillary and inner dentary teeth. A key to identify species of Strophidon is provided. The distribution and maximum size of each species are also re-evaluated.


Subject(s)
Eels/classification , Animals , Asia , Eels/anatomy & histology , Eels/genetics , Genetic Variation , Species Specificity
13.
Cancer Immunol Immunother ; 68(7): 1087-1094, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31089757

ABSTRACT

We evaluated the analytical and clinical performance of a novel circulating tumor cell (CTC)-based blood test for determination of programmed death ligand 1 (PD-L1) protein expression status in real time in treatment-naïve non-small cell lung cancer (NSCLC) patients. CTCs were detected in 86% of patients with NSCLC (I-IV) at the time of diagnosis, with a 67% PD-L1 positivity rate (≥ 1 PDL + CTC). Among 33 NSCLC patients with PD-L1 results available via both tissue immunohistochemistry (IHC) and CTC assays, 78.9% were positive according to both methods. The CTC test identified an additional ten cases that were positive for PD-L1 expression but that tested negative via IHC analysis. Detection of higher PD-L1 expression on CTCs compared to that in the corresponding tissue was concordant with data obtained using other platforms in previously treated patients. The concordance in PD-L1 expression between tissue and CTCs was approximately 57%, which is higher than that reported by others. In summary, evaluation of PD-L1 protein expression status on CTCs isolated from NSCLC patients is feasible. PD-L1 expression status on CTCs can be determined serially during the disease course, thus overcoming the myriad challenges associated with tissue analysis.


Subject(s)
B7-H1 Antigen/analysis , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/blood , Lung Neoplasms/blood , Neoplastic Cells, Circulating/metabolism , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , False Negative Reactions , Feasibility Studies , Female , Humans , Immunohistochemistry , Lung/pathology , Lung Neoplasms/pathology , Male , Middle Aged
14.
BMC Cancer ; 19(1): 634, 2019 Jun 27.
Article in English | MEDLINE | ID: mdl-31248373

ABSTRACT

BACKGROUND: Metastasis is a leading cause of breast cancer mortality. The induction of epithelial-to-mesenchymal transition (EMT) and complex oncogenic signaling is a vital step in the evolution of highly metastatic and therapeutically-intractable breast cancer; necessitating novel target discovery or development of therapeutics that target metastatic breast cells (MBCs). METHODS: To achieve this, this study employs a combination of in silico bioinformatics analyses, protein and transcript analyses, drug sensitivity assays, functional assays and animal studies. RESULTS: The present study identified CDH11 as an inductor and/or facilitator of metastatic signaling, and biomarker of poor prognosis in MBCs. Furthermore, we showed that in the presence of CDH11-rich cancer-associated fibroblasts (CAFs), MCF7 and MDA-MB-231 MBC cell lines acquired enhanced metastatic phenotype with increased CDH11, ß-catenin, vimentin, and fibronectin (FN) expression. We also demonstrated, for the first time to the best of our knowledge that exposure to anti-CDH11 antibody suppresses metastasis, reduces CDH11, FN and ß-catenin expression, and abrogate the cancer stem cell (CSC)-like traits of MBC cells. Interestingly, ectopic expression of miR-335 suppressed CDH11, ß-catenin and vimentin expression, in concert with attenuated metastatic and CSC potentials of the MBC cells; conversely, inhibition of miR-335 resulted in increased metastatic potential. Finally, corroborating the in silica and in vitro findings, in vivo assays showed that the administration of anti-CDH11 antibody or miR-335 mimic suppressed tumorigenesis and inhibited cancer metastasis. CONCLUSIONS: These findings validate our hypotheses that miR-335 mediates anti-CDH11 antibody therapy response and that an enhanced miR-335/CDH11 ratio elicits marked suppression of the MBC CSC-like and metastatic phenotypes, thus revealing a therapeutically-exploitable inverse correlation between CDH11-enhanced CSC-like and metastatic phenotype and miR-335 expression in MBCs. Thus, we highlight the therapeutic promise of humanized anti-CDH11 antibodies or miR-335-mimic, making a case for their clinical application as efficacious therapeutic option in patients with MBC.


Subject(s)
Breast Neoplasms/pathology , Cadherins/antagonists & inhibitors , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Cadherins/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Mice, Inbred NOD , Mice, SCID , MicroRNAs/pharmacology , Neoplasm Metastasis , Prognosis , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
15.
J Nanosci Nanotechnol ; 18(10): 7190-7196, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29954557

ABSTRACT

Magnetite nanoparticles (MNPs) Fe3O4 and r-Fe2O3 were surface-modified with poly(ethylene glycol) (PEG) in order to improve their specificity and bioactivity. PEG and the anti-MEL monoclonal antibody (mAb) were successfully immobilized on the surface of MNPs and characterized using FTIR, UV-Vis and TEM analyses. Surface modification of MNPs-PEG-mAb conjugates of a variety of sizes and magnetite types was employed to design and prepare labels for use in a lateral flow immunoassay (LFIA) to test whether the size of the conjugate can affect the performance of the assay. The results showed that the detection limit was mainly determined by the size of the MNPs-PEG-mAb conjugate. Under optimized conditions, a detection limit of 0.4 ppm for melamine was achieved using Fe2O3-PEG-mAb, which was almost 5-fold lower than that of the Fe3O4-PEG-mAb conjugate (2.2 ppm).


Subject(s)
Antibodies, Immobilized/chemistry , Chromatography, Affinity/methods , Magnetite Nanoparticles/chemistry , Triazines/analysis , Limit of Detection , Polyethylene Glycols/chemistry , Reagent Strips/analysis
16.
Ann Surg Oncol ; 24(13): 4033-4041, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28744792

ABSTRACT

BACKGROUND: Human 3ß-hydroxysteroid dehydrogenase type 1 (HSD3B1) plays a vital role in steroidogenesis in breast tumors and may therefore be a suitable target for treatment of breast cancer. This study investigated the role of HSD3B1 in the pathogenesis of breast cancer in clinical and experimental settings. METHODS: Expression of HSD3B1 in primary tumors of 258 breast cancer patients was evaluated by immunohistochemistry. Screening of breast cancer cell lines indicated that triple-negative MDA-MB-231 cells expressed HSD3B1. The effects from genetic and pharmacologic inhibition of HSD3B1 were assessed in vitro and in vivo. RESULTS: The findings showed that 44% of the 258 breast cancers were HSD3B1-positive. The HSD3B1-positivity was associated with advanced-stage disease (p = 0.009) and reduced recurrence-free survival (p = 0.048) but not with tumor subtype or estrogen receptor status. Silencing of HSD3B1 or treatment with an HSD3B1 inhibitor (trilostane) reduced colony formation in breast cancer cells. Knockdown of HSD3B1 inhibited cell proliferation and migration. Analysis of a murine xenograft tumor model indicated that trilostane significantly slowed tumor growth. CONCLUSIONS: Expression of HSD3B1 in breast cancer is negatively associated with prognosis. The study found HSD3B1 to be a potential therapeutic target for breast cancer independent of estrogen receptor status.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Multienzyme Complexes/metabolism , Progesterone Reductase/metabolism , Receptors, Estrogen/metabolism , Steroid Isomerases/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Dihydrotestosterone/analogs & derivatives , Dihydrotestosterone/pharmacology , Female , Follow-Up Studies , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/genetics , Progesterone Reductase/antagonists & inhibitors , Progesterone Reductase/genetics , Prognosis , RNA, Small Interfering/genetics , Steroid Isomerases/antagonists & inhibitors , Steroid Isomerases/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Toxicol Appl Pharmacol ; 325: 48-60, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28408137

ABSTRACT

Targeting residual self-renewing, chemoresistant cancerous cells may represent the key to overcoming therapy resistance. The entry of these quiescent cells into an activated state is associated with high metabolic demand and autophagic flux. Therefore, modulating the autophagy pathway in aggressive carcinomas may be beneficial as a therapeutic modality. In this study, we evaluated the anti-tumor activities of 4-acetylantroquinonol B (4-AAQB) in chemoresistant ovarian cancer cells, particularly its ability to modulate autophagy through autophagy-related genes (Atg). Atg-5 was overexpressed in invasive ovarian cancer cell lines and tissue (OR: 5.133; P=0.027) and depleting Atg-5 in ES-2 cell lines significantly induced apoptosis. 4-AAQB effectively suppressed viability of various subtypes of ovarian cancer. Cells with higher cisplatin-resistance were more responsive to 4-AAQB. For the first time, we demonstrate that 4-AAQB significantly suppress Atg-5 and Atg-7 expression with decreased autophagic flux in ovarian cancer cells via inhibition of the PI3K/Akt/mTOR/p70S6K signaling pathway. Similar to Atg-5 silencing, 4-AAQB-induced autophagy inhibition significantly enhanced cell death in vitro. These results are comparable to those of hydroxychloroquine (HCQ). In addition, 4-AAQB/cisplatin synergistically induced apoptosis in ovarian cancer cells. In vivo, 4-AAQB/cisplatin also significantly induced apoptosis and autophagy in an ES-2 mouse xenografts model. This is the first report demonstrating the efficacy of 4-AAQB alone or in combination with cisplatin on the suppression of ovarian cancer via Atg-5-dependent autophagy. We believe these findings will be beneficial in the development of a novel anti-ovarian cancer therapeutic strategy.


Subject(s)
4-Butyrolactone/analogs & derivatives , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Cisplatin/pharmacology , Cyclohexanones/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplasms, Glandular and Epithelial/drug therapy , Ovarian Neoplasms/drug therapy , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , 4-Butyrolactone/pharmacology , Animals , Apoptosis/drug effects , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasms, Glandular and Epithelial/enzymology , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
18.
Tumour Biol ; 39(5): 1010428317691689, 2017 May.
Article in English | MEDLINE | ID: mdl-28466786

ABSTRACT

Current standard chemotherapy for late stage ovarian cancer is found unsuccessful due to relapse after completing the regimens. After completing platinum-based chemotherapy, 70% of patients develop relapse and resistance. Recent evidence proves ovarian cancer stem cells as the source of resistance. Therefore, treatment strategy to target both cancer stem cells and normal stem cells is essential. In this study, we developed a novel chalcone derivative as novel drug candidate for ovarian cancer treatment. We found that methoxyphenyl chalcone was effective to eliminate ovarian cancer cells when given either as monotherapy or in combination with cisplatin. We found that cell viability of ovarian cancer cells was decreased through apoptosis induction. Dephosphorylation of Bcl2-associated agonist of cell death protein was increased after methoxyphenyl chalcone treatment that led to activation of caspases. Interestingly, this drug also worked as a G2/M checkpoint modulator with alternative ways of DNA damage signal-evoking potential that might work to increase response after cisplatin treatment. In addition, methoxyphenyl chalcone was able to suppress autophagic flux and stemness regulator in ovarian spheroids that decreased their survival. Therefore, combination of methoxyphenyl chalcone and cisplatin showed synergistic effects. Taken together, we believe that our novel compound is a promising novel therapeutic agent for effective clinical treatment of ovarian cancer.


Subject(s)
Chalcone/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Neoplasms, Glandular and Epithelial/diet therapy , Ovarian Neoplasms/diet therapy , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/administration & dosage , DNA Damage/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Humans , Neoplasm Recurrence, Local/pathology , Neoplasms, Glandular and Epithelial/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Phosphorylation/drug effects
19.
BMC Cancer ; 16: 160, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26917489

ABSTRACT

BACKGROUND: Triple negative breast cancers (TNBC) possess cell dedifferentiation characteristics, carry out activities connate to those of cancer stem cells (CSCs) and are associated with increased metastasis, as well as, poor clinical prognosis. The regulatory mechanism of this highly malignant phenotype is still poorly characterized. Accruing evidence support the role of non-coding RNAs (ncRNAs) as potent regulators of CSC and metastatic gene expression, with their dysregulation implicated in tumorigenesis and disease progression. METHODS: In this study, we investigated TNBC metastasis, metastasis-associated genes and potential inhibitory mechanisms using bioinformatics, tissue microarray analyses, immunoblotting, polymerase chain reaction, loss and gain of gene function assays and comparative analyses of data obtained. RESULTS: Compared with other breast cancer types, the highly metastatic MDA-MB-231 cells concurrently exhibited increased expression levels of Lysine-specific demethylase 5B protein (KDM5B) and long non-coding RNA (lncRNA), MALAT1, suggesting their functional association. KDM5B-silencing in the TNBC cells correlated with the upregulation of hsa-miR-448 and led to suppression of MALAT1 expression with decreased migration, invasion and clonogenic capacity in vitro, as well as, poor survival in vivo. This projects MALAT1 as a mediator of KDM5B oncogenic potential and highlights the critical role of this microRNA, lncRNA and histone demethylase in cancer cell motility and metastatic colonization. Increased expression of KDM5B correlating with disease progression and poor clinical outcome in breast cancer was reversed by hsa-miR-448. CONCLUSIONS: Our findings demonstrate the critical role of KDM5B and its negative regulator hsa-miR-448 in TNBC metastasis and progression. Hsa-miR-448 disrupting KDM5B-MALAT1 signalling axis and associated activities in TNBC cells, projects it as a putative therapeutic factor for selective eradication of TNBC cells. Graphical abstract KDM5B, MALAT1 and hsa-miR-448 are active looped components of the epigenetic poculo mortis in aggressive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Jumonji Domain-Containing Histone Demethylases/genetics , MicroRNAs/genetics , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Disease Progression , Female , Gene Expression Profiling , Gene Knockdown Techniques , Gene Silencing , Humans , Immunohistochemistry , Jumonji Domain-Containing Histone Demethylases/chemistry , Jumonji Domain-Containing Histone Demethylases/metabolism , MicroRNAs/chemistry , Models, Biological , Models, Molecular , Molecular Conformation , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Prognosis , Protein Binding , RNA Interference , RNA, Long Noncoding/chemistry , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Risk Factors , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
20.
Ecotoxicol Environ Saf ; 124: 37-41, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26454073

ABSTRACT

Diarrheagenic Escherichia coli (DEC) are the most common agents of diarrhea. Waterborne DEC could pose a potential health risk to human through agricultural, household, recreational, and industrial use. There are few published reports on the detection of DEC and its seasonal distribution in aquatic environments. The presence of DEC in different types of aquatic environments was investigated in this study. Water samples were collected from major rivers, water reservoirs, and recreational hot springs throughout Taiwan. Moreover, an intensive water sampling plan was carried out along Puzih River. The detection of DEC target genes was used to determine the presence of enterotoxigenic E. coli (ETEC), enteropathogenic E. coli (EPEC), and Shiga toxin-producing E. coli (STEC). Among the 383 water samples analyzed, DEC was found in 122 (31.8%) samples. The detection rate varied by genotype, raging from 3.6% for STEC to 17.2% for EPEC. The DEC detection rate was higher from river waters than reservoirs and hot springs. In addition, DEC was detected at a higher rate in spring and summer. The presence of EPEC was significantly associated with total coliform levels among hot spring samples. Moreover, the presence of ETEC in river water samples was associated with heterotrophic plate counts. Water with EPEC differed significantly in pH from Puzih River samples. These results suggest that seasonal characteristics may affect the presence of DEC in different aquatic environments, and water quality indicators may be indicative of the presence of DEC.


Subject(s)
Enteropathogenic Escherichia coli/isolation & purification , Enterotoxigenic Escherichia coli/isolation & purification , Hot Springs/microbiology , Rivers/microbiology , Shiga-Toxigenic Escherichia coli/isolation & purification , Water Supply , Diarrhea , Escherichia coli Infections , Humans , Seasons , Taiwan
SELECTION OF CITATIONS
SEARCH DETAIL