Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Parasitol ; 124: 57-89, 2024.
Article in English | MEDLINE | ID: mdl-38754927

ABSTRACT

For over a century, vector ecology has been a mainstay of vector-borne disease control. Much of this research has focused on the sensory ecology of blood-feeding arthropods (black flies, mosquitoes, ticks, etc.) with terrestrial vertebrate hosts. Of particular interest are the cues and sensory systems that drive host seeking and host feeding behaviours as they are critical for a vector to locate and feed from a host. An important yet overlooked component of arthropod vector ecology are the phenotypic changes observed in infected vectors that increase disease transmission. While our fundamental understanding of sensory mechanisms in disease vectors has drastically increased due to recent advances in genome engineering, for example, the advent of CRISPR-Cas9, and high-throughput "big data" approaches (genomics, proteomics, transcriptomics, etc.), we still do not know if and how parasites manipulate vector behaviour. Here, we review the latest research on arthropod vector sensory systems and propose key mechanisms that disease agents may alter to increase transmission.


Subject(s)
Arthropod Vectors , Animals , Arthropod Vectors/physiology , Humans , Arthropods/physiology , Vector Borne Diseases/transmission , Vector Borne Diseases/prevention & control , Host-Parasite Interactions
2.
Mol Cells ; 45(10): 718-728, 2022 Oct 31.
Article in English | MEDLINE | ID: mdl-35996826

ABSTRACT

Splicing factor B subunit 4 (SF3B4), a component of the U2-pre-mRNA spliceosomal complex, contributes to tumorigenesis in several types of tumors. However, the oncogenic potential of SF3B4 in lung cancer has not yet been determined. The in vivo expression profiles of SF3B4 in non-small cell lung cancer (NSCLC) from publicly available data revealed a significant increase in SF3B4 expression in tumor tissues compared to that in normal tissues. The impact of SF3B4 deletion on the growth of NSCLC cells was determined using a siRNA strategy in A549 lung adenocarcinoma cells. SF3B4 silencing resulted in marked retardation of the A549 cell proliferation, accompanied by the accumulation of cells at the G0/G1 phase and increased expression of p27, p21, and p53. Double knockdown of SF3B4 and p53 resulted in the restoration of p21 expression and partial recovery of cell proliferation, indicating that the p53/p21 axis is involved, at least in part, in the SF3B4-mediated regulation of A549 cell proliferation. We also provided ubiquitination factor E4B (UBE4B) is essential for p53 accumulation after SF3B4 depletion based on followings. First, co-immunoprecipitation showed that SF3B4 interacts with UBE4B. Furthermore, UBE4B levels were decreased by SF3B4 depletion. UBE4B depletion, in turn, reproduced the outcome of SF3B4 depletion, including reduction of polyubiquitinated p53 levels, subsequent induction of p53/p21 and p27, and proliferation retardation. Collectively, our findings indicate the important role of SF3B4 in the regulation of A549 cell proliferation through the UBE4B/p53/p21 axis and p27, implicating the therapeutic strategies for NSCLC targeting SF3B4 and UBE4B.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , RNA Precursors/metabolism , RNA Splicing Factors/genetics , RNA, Small Interfering/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
Biochem Biophys Res Commun ; 619: 42-48, 2022 09 03.
Article in English | MEDLINE | ID: mdl-35732079

ABSTRACT

Bcl-2-interacting cell death suppressor (BIS), also called as BAG3, regulates numerous physiological processes, such as apoptosis, protein quality control, and senescence. Whole-body Bis-knockout (KO) mice exhibit early lethality following cardiac and skeletal muscle dysfunction. The first attempt to generate organ-specific knockout mice resulted in constitutive or inducible heart-specific Bis-knockout mice, which exhibited cardiac dilation and underwent premature death. Here, we generated hepatocyte-specific Bis-knockout (Bis-HKO) mice and found no abnormalities in metabolic function and survival. However, depletion of HSPB8 and accumulation of p62 indicated impaired autophagy in Bis-HKO livers. Interestingly, the number of peroxisomes wrapped by phagophore membranes increased as evidenced by transmission electron microscopy analysis, indicating defects in the progression of pexophagy. In addition, increased dihydroethidine intensities and histone H3 K9me3-positive nuclei indicated increased oxidative stress and senescence induction in Bis-HKO livers. Mechanistically, p27 was upregulated in Bis-HKO livers. In SNU368 hepatocellular carcinoma cells, BIS depletion led to p27 upregulation, and increase in histone H3 K9me3 levels and senescence-associated ß-galactosidase staining; therefore, reproducing the in vivo senescence phenotype. Despite the observation of no metabolic abnormalities, BIS depletion led to defective autophagy, increased oxidative stress, and senescence in Bis-HKO livers. Collectively, our results suggest a role for BIS in maintaining liver regeneration potential under pathological conditions.


Subject(s)
Histones , Liver Neoplasms , Animals , Cellular Senescence/genetics , Hepatocytes/metabolism , Histones/metabolism , Liver/metabolism , Liver Neoplasms/pathology , Liver Regeneration/physiology , Mice , Mice, Knockout
4.
Cells ; 9(10)2020 10 19.
Article in English | MEDLINE | ID: mdl-33086735

ABSTRACT

BCL-2 interacting cell death suppressor (BIS), also known as BAG3, is a multifunctional protein. Aberrant expression and mutation of BIS have been implicated in cancers and myopathy. However, there have only been a few studies on the splicing of BIS pre-mRNA. In the present study, through RT-PCR and sequencing in various cell lines and mouse tissues, we identified for the first time the presence of BIS mRNA isomers in which exon 3 or exons 2-3 are skipped. We also demonstrated that the depletion of SRSF3 promoted the skipping of exon 3 of BIS pre-mRNA in endogenous BIS and the GFP-BIS minigene. SRSF3 specifically interacts with the putative binding sites in exon 3, in which deletion promoted the skipping of exon 3 in the GFP-BIS minigene, which was comparable to the effect of SRSF knockdown. Even though acceleration of exon 3 skipping was not observed in response to various stimuli, SRSF3 depletion, accompanied by the production of a truncated BIS protein, inhibited the nuclear translocation of HSF1, which was restored by the wild-type BIS, not by exon 3-depleted BIS. Therefore, our results suggested that the maintenance of SRSF3 levels and subsequent preservation of the intact BIS protein is an important factor in modulating HSF1 localization upon cellular stress.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Exons/genetics , RNA Precursors/genetics , Serine-Arginine Splicing Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Cell Line , Heat Shock Transcription Factors/metabolism , Heat-Shock Response , Humans , Mice , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Anticancer Res ; 40(4): 2033-2042, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32234894

ABSTRACT

BACKGROUND: SF3B4, a critical component of U2 pre-mRNA spliceosomal complex, has been recently indicated as a potential oncogene in hepatocellular carcinoma (HCC). However, limited information exists on how SF3B4 expression is regulated in HCC. MATERIALS AND METHODS: To determine the regulatory factor for SF3B4 expression, small interfering RNA (siRNA), real-time polymerase chain reaction (qRT-PCR) and western blotting assay were performed. The in vivo expression profiles of SRSF3 and SF3B4 were analyzed using public datasets and clinical samples. RESULTS: Among 10 liver-specific splicing factors, only SRSF3 knockdown resulted in a significant increase in SF3B4 mRNA and consequently protein levels in SNU-368 HCC cells, probably via the retardation of SF3B4 mRNA decay rates. Using green fluorescent protein-SF3B4 fusion construct, the coding region of SF3B4 was found to be involved in SRSF3-mediated regulation of SF3B4 expression. Publicly available data from paired normal and tumor tissues in HCC and results from patients with HCC suggest that SRSF3 and SF3B4 possess an inverse relationship. CONCLUSION: SRSF3 is a key molecule for determining SF3B4 levels in HCC cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA Splicing Factors/genetics , Serine-Arginine Splicing Factors/genetics , Alternative Splicing/genetics , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/pathology , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics
6.
Anticancer Res ; 38(9): 5057-5065, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30194150

ABSTRACT

BACKGROUND/AIM: High expression of the Bcl-2-interacting cell death suppressor (BIS), an anti-apoptotic protein, in various human cancers is linked to a poor outcome. The purpose of this study was to clarify whether BIS is associated with the migration and invasive characteristics of A549 cells. MATERIALS AND METHODS: BIS-knockout (KO) cells were prepared by the CRISPR/Cas9 method. The aggressive behaviors of A549 cells were analyzed by wound healing and a transwell invasion assay as well as 3D spheroid culture. RESULTS: BIS depletion resulted in significant inhibition of the migration and invasive potential of A549 cells which was accompanied by an increased ratio of E-cadherin/N-cadherin and a decrease in the mRNA levels of Zeb1, Snail, Slug and MMP-2. NF-ĸB activity was suppressed in BIS-KO A549 cells due to the decrease in p65 protein levels, but not in mRNA levels. CONCLUSION: BIS regulates cell invasion and the induction of the epithelial-mesenchymal transition (EMT) phenotype in A549 cells probably via the NF-ĸB signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , A549 Cells , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Lung Neoplasms/metabolism , NF-kappa B/metabolism , Neoplasm Invasiveness , Signal Transduction , Spheroids, Cellular
7.
Anticancer Res ; 38(8): 4525-4533, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30061218

ABSTRACT

BACKGROUND/AIM: High expression of the Bcl-2-interacting cell death suppressor (BIS), an anti-apoptotic protein, in various human cancers is linked to a poor outcome. The purpose of this study was to clarify whether BIS is associated with the migration and invasive characteristics of A549 cells. MATERIALS AND METHODS: BIS-knockout (KO) cells were prepared by the CRISPR/Cas9 method. The aggressive behaviors of A549 cells were analyzed by wound healing and a transwell invasion assay as well as 3D spheroid culture. RESULTS: BIS depletion resulted in significant inhibition of the migration and invasive potential of A549 cells which was accompanied by an increased ratio of E-cadherin/N-cadherin and a decrease in the mRNA levels of Zeb1, Snail, Slug and MMP-2. NF-κB activity was suppressed in BIS-KO A549 cells due to the decrease in p65 protein levels, but not in mRNA levels. CONCLUSION: BIS regulates cell invasion and the induction of the epithelial-mesenchymal transition (EMT) phenotype in A549 cells probably via the NF-κB signaling pathway.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/physiology , Lung Neoplasms/pathology , Neoplasm Invasiveness/pathology , A549 Cells , CRISPR-Cas Systems/physiology , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Communication/physiology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Humans , Lung Neoplasms/metabolism , Matrix Metalloproteinase 2/metabolism , NF-kappa B/metabolism , RNA, Messenger/metabolism , Signal Transduction/physiology , Snail Family Transcription Factors/metabolism , Wound Healing/physiology , Zinc Finger E-box-Binding Homeobox 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...