Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Toxicol Appl Pharmacol ; 485: 116888, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38452945

ABSTRACT

Upregulation of the multidrug efflux pump ABCB1/MDR1 (P-gp) and the anti-apoptotic protein BIRC5/Survivin promotes multidrug resistance in various human cancers. GDC-0152 is a DIABLO/SMAC mimetic currently being tested in patients with solid tumors. However, it is still unclear whether GDC-0152 is therapeutically applicable for patients with ABCB1-overexpressing multidrug-resistant tumors, and the molecular mechanism of action of GDC-0152 in cancer cells is still incompletely understood. In this study, we found that the potency of GDC-0152 is unaffected by the expression of ABCB1 in cancer cells. Interestingly, through in silico and in vitro analysis, we discovered that GDC-0152 directly modulates the ABCB1-ATPase activity and inhibits ABCB1 multidrug efflux activity at sub-cytotoxic concentrations (i.e., 0.25×IC50 or less). Further investigation revealed that GDC-0152 also decreases BIRC5 expression, induces mitophagy, and lowers intracellular ATP levels in cancer cells at low cytotoxic concentrations (i.e., 0.5×IC50). Co-treatment with GDC-0152 restored the sensitivity to the known ABCB1 substrates, including paclitaxel, vincristine, and YM155 in ABCB1-expressing multidrug-resistant cancer cells, and it also restored the sensitivity to tamoxifen in BIRC5-overexpressing tamoxifen-resistant breast cancer cells in vitro. Moreover, co-treatment with GDC-0152 restored and potentiated the anticancer effects of paclitaxel in ABCB1 and BIRC5 co-expressing xenograft tumors in vivo. In conclusion, GDC-0152 has the potential for use in the management of cancer patients with ABCB1 and BIRC5-related drug resistance. The findings of our study provide essential information to physicians for designing a more patient-specific GDC-0152 clinical trial program in the future.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Benzodioxoles , Drug Resistance, Neoplasm , Indolizines , Survivin , Humans , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Survivin/genetics , Survivin/metabolism , Animals , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Female , Mice, Nude , Mice , Antineoplastic Agents/pharmacology , Xenograft Model Antitumor Assays , Apoptosis Regulatory Proteins/metabolism , Drug Resistance, Multiple/drug effects , Paclitaxel/pharmacology , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mice, Inbred BALB C , Inhibitor of Apoptosis Proteins/metabolism , Inhibitor of Apoptosis Proteins/genetics
2.
R I Med J (2013) ; 106(9): 23-27, 2023 10 02.
Article in English | MEDLINE | ID: mdl-37768158

ABSTRACT

Cross-cultural medical education has been suggested to train students to care for diverse patient populations and reform medical education systems. In this article, the authors conduct a cross-cultural comparison between two medical schools with a long-standing relationship - the Warren Alpert Medical School of Brown University in the United States and the School of Medicine of National Cheng Kung University in Taiwan - focusing on history, admissions, and curriculum.


Subject(s)
Education, Medical, Undergraduate , Education, Medical , United States , Humans , Schools, Medical , Cross-Cultural Comparison , Curriculum , Universities
3.
Am J Cancer Res ; 12(5): 2084-2101, 2022.
Article in English | MEDLINE | ID: mdl-35693094

ABSTRACT

The incidence of breast cancer is increasing, and is one of the leading causes of cancer death worldwide. Dysregulation of NOTCH1 signaling is reported in breast cancer. In present study, bioinformatics was utilized to study the expression of NOTCH1 gene in breast cancer from public databases, including the Kaplan-Meier Plotter, PrognoScan, Human Protein Atlas, and cBioPortal. The relationship between NOTCH1 mRNA expression and survival of patients was inconsistent in public databases. In addition, we performed immunohistochemistry (IHC) staining of 135 specimens from our hospital. Lower cytoplasmic staining of NOTCH1 protein was correlated with cancer recurrence, bone metastasis, and a worse disease-free survival of patients, especially those with estrogen receptor-positive and human epidermal growth factor receptor 2-positive (HER2+) cancers. In TCGA breast cancer dataset, lower expression of NOTCH1 in breast cancer specimens was correlated with higher level of CCND1 (protein: cyclin D1). Decreased expression of NOTCH1 was correlated with lower level of CCNA1 (protein: cyclin A1), CCND2 (protein: cyclin D2), CCNE1 (protein: cyclin E1), CDK6 (protein: CDK6), and CDKN2C (protein: p18). In conclusion, NOTCH1 mRNA expression is not consistently correlated with clinical outcomes of breast cancer patients. Low cytoplasmic expression of NOTCH1 in IHC study is correlated with poor prognosis of breast cancer patients. Cytoplasmic localization of NOTCH1 protein failed to initial oncogenic signaling in present study. Expression of NOTCH1 mRNA was discordant with cell cycle-related genes. Regulation of NOTCH1 in breast cancer involves gene expression, protein localization and downstream signaling.

4.
Aging Dis ; 13(3): 868-883, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35656119

ABSTRACT

Amyloid cascade hypothesis proposes that amyloid ß (Aß) accumulation is the initiator and major contributor to the development of Alzheimer's disease (AD). However, this hypothesis has recently been challenged by clinical studies showing that reduction of Aß accumulation in the brain does not accompany with cognitive improvement, suggesting that therapeutically targeting Aß in the brain may not be sufficient for restoring cognitive function. Since the molecular mechanism underlying the progressive development of cognitive impairment after Aß clearance is largely unknown, the reason of why there is no behavioral improvement after Aß clearance remains elusive. In the current study, we demonstrated that transient Aß expression caused learning deficit in later life, despite the accumulated Aß was soon being removed after the expression. Early Aß exposure decreased the cellular expression of XBP1 and both the antioxidants, catalase, and dPrx5, which made cells more vulnerable to oxidative stress in later life. Early induction of XBP1, catalase, and dPrx5 prevented the overproduction of ROS, improved the learning performance, and preserved the viability of cells in the later life with the early Aß induction. Treating the early Aß exposed flies with antioxidants such as vitamin E, melatonin and lipoic acid, after the removal of Aß also preserved the learning ability in later life. Taken together, we demonstrated that early and transient Aß exposure can have a profound impact on animal behavior in later life and also revealed the cellular and molecular mechanism underlying the development of learning impairment by the early and transient Aß exposure.

5.
Int J Mol Med ; 50(2)2022 08.
Article in English | MEDLINE | ID: mdl-35703361

ABSTRACT

Cancer cells can acquire resistance to targeted therapeutic agents when the designated targets or their downstream signaling molecules develop protein conformational or activity changes. There is an increasing interest in developing poly­pharmacologic anticancer agents to target multiple oncoproteins or signaling pathways in cancer cells. The microRNA 125a­5p (miR­125a­5p) is a tumor suppressor, and its expression has frequently been downregulated in tumors. By contrast, the anti­apoptotic molecule BIRC5/SURVIVIN is highly expressed in tumors but not in the differentiated normal tissues. In the present study, the development of a BIRC5 gene promoter­driven, miR­125a­5p expressing, poly­L­lysine­conjugated magnetite iron poly­pharmacologic nanodrug (pL­MNP­pSur­125a) was reported. The cancer cells self­activating property and the anticancer effects of this nanodrug were examined in both the multidrug efflux protein ABCB1/MDR1­expressing/­non­expressing cancer cells in vitro and in vivo. It was demonstrated that pL­MNP­pSur­125a decreased the expression of ERBB2/HER2, HDAC5, BIRC5, and SP1, which are hot therapeutic targets for cancer in vitro. Notably, pL­MNP­pSur­125a also downregulated the expression of TDO2 in the human KB cervical carcinoma cells. PL­MNP­pSur­125a decreased the viability of various BIRC5­expressing cancer cells, regardless of the tissue origin or the expression of ABCB1, but not of the human BIRC5­non­expressing HMEC­1 endothelial cells. In vivo, pL­MNP­pSur­125a exhibited potent antitumor growth effects, but without inducing liver toxicity, in various zebrafish human­ABCB1­expressing and ABCB1­non­expressing tumor xenograft models. In conclusion, pL­MNP­pSur­125a is an easy­to­prepare and a promising poly­pharmacological anticancer nanodrug that has the potential to manage numerous malignancies, particularly for patients with BIRC5/ABCB1­related drug resistance after prolonged chemotherapeutic treatments.


Subject(s)
Antineoplastic Agents , MicroRNAs , Nanoparticles , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Endothelial Cells , Humans , MicroRNAs/genetics , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Zebrafish/genetics
6.
Sci Rep ; 12(1): 241, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997132

ABSTRACT

The prognosis of patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer has considerably improved. However, no reliable treatment besides anti-HER2 strategies has been available. FTY720, a small-molecule compound used for treating refractory multiple sclerosis, has been reported to have beneficial effects against cancers. We therefore evaluated the efficacy of FTY720 in trastuzumab-resistant breast cancer cells and investigated the possible mechanism involved. This study evaluated morphological changes after FTY720 treatment. Antiproliferative WST-1 assays and LDH Cytotoxicity Assay Kits were used to determine the treatment effects of drugs, whereas Western blot analysis was used to evaluate protein expression. Apoptotic events were investigated through annexin V staining and TUNEL assays using flow cytometry. FTY720 was effective in trastuzumab-resistant breast cancer cell lines despite the presence of PIK3CA mutation. Studied on a xenograft mouse model, FTY720-treated groups had statistically significantly poorer HCC1954 xenograft growth in vivo compared with the control group. Our findings suggest that FTY720 can overcome resistance to trastuzumab therapy in patients with HER2-positive breast cancer, with FTY720 plus trastuzumab might offer even better efficacy in vitro and in vivo.


Subject(s)
Breast Neoplasms/drug therapy , Fingolimod Hydrochloride/administration & dosage , Receptor, ErbB-2/genetics , Animals , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Drug Resistance, Neoplasm , Female , Humans , Mice , Mice, Inbred BALB C , Receptor, ErbB-2/metabolism , Trastuzumab/administration & dosage , Xenograft Model Antitumor Assays
7.
J Cell Physiol ; 237(1): 178-188, 2022 01.
Article in English | MEDLINE | ID: mdl-34406646

ABSTRACT

Dysregulation of DNA damage response/repair and genomic instability promote tumorigenesis and the development of various neurological diseases. Autophagy is a dynamic catabolic process used for removing unnecessary or dysfunctional proteins and organelles in cells. Despite the consensus in the field that upregulation of autophagy promotes the initiation of the DNA damage response and assists the process of homologous recombination upon genotoxic stress, a few studies showed that upregulation of autophagy (or excessive autophagy), under certain circumstances, triggers caspase/apoptosis-independent DNA damage and promotes genomic instability in cells. As the cytoprotective and the DNA repairing roles of autophagy have been discussed extensively in different reviews, here, we mainly focus on describing the latest studies which reported the "opposite" roles of autophagy (or excessive autophagy). We will discuss whether the "dark side" (i.e., the opposite/unconventional effect) of autophagy on the maintenance of DNA integrity and genomic stability really does exist in cells and if it does, will it be one of the yet-to-be-identified causes of cancer, in this review.


Subject(s)
Autophagy , Genomic Instability , Autophagy/genetics , Carcinogenesis/metabolism , DNA Damage/genetics , DNA Repair , Genomic Instability/genetics , Humans
8.
Mol Neurobiol ; 58(10): 5224-5238, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34273104

ABSTRACT

Accumulated beta-amyloid (Aß) in the brain is the hallmark of Alzheimer's disease (AD). Despite Aß accumulation is known to trigger cellular dysfunctions and learning and memory damage, the detailed molecular mechanism remains elusive. Recent studies have shown that the onset of memory impairment and learning damage in the AD animal is different, suggesting that the underlying mechanism of the development of memory impairment and learning damage may not be the same. In the current study, with the use of Aß42 transgenic flies as models, we found that Aß induces memory damage and learning impairment via differential molecular signaling pathways. In early stage, Aß activates both Ras and PI3K to regulate Rac1 activity, which affects mostly on memory performance. In later stage, PI3K-Akt is strongly activated by Aß, which leads to learning damage. Moreover, reduced Akt, but not Rac1, activity promotes cell viability in the Aß42 transgenic flies, indicating that Akt and Rac1 exhibit differential roles in Aß regulating toxicity. Taken together, different molecular and cellular mechanisms are involved in Aß-induced learning damage and memory decline; thus, caution should be taken during the development of therapeutic intervention in the future.


Subject(s)
Amyloid beta-Peptides/toxicity , Drosophila Proteins/metabolism , Maze Learning/physiology , Memory Disorders/metabolism , Peptide Fragments/toxicity , Proto-Oncogene Proteins c-akt/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Drosophila , Drosophila Proteins/genetics , Female , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Memory Disorders/genetics , Mice, Transgenic , Proto-Oncogene Proteins c-akt/genetics , rac GTP-Binding Proteins/genetics
9.
Pharmacol Res ; 166: 105474, 2021 04.
Article in English | MEDLINE | ID: mdl-33549731

ABSTRACT

Activation of autophagy plays a critical role in DNA repair, especially for the process of homologous recombination. Despite upregulation of autophagy promotes both the survival and the death of cells, the pathways that govern the pro-cell death effects of autophagy are still incompletely understood. YM155 is originally developed as an expression suppressant of BIRC5 (an anti-apoptotic molecule) and it has reached Phase I/II clinical trials for the treatment of variety types of cancer. However, the target-specificity of YM155 has recently been challenged as several studies reported that YM155 exhibits direct DNA damaging effects. Recently, we discovered that BIRC5 is an autophagy negative-modulator. Using function-comparative analysis, we found in the current study that YM155 and BIRC5 siRNA both induced early "autophagy-dependent ROS production-mediated" DNA damage/strand breaks and concurrently downregulated the expression of RAD54L, RAD51, and MRE11, which are molecules known for their important roles in homologous recombination, in human cancer (MCF7, MDA-MB-231, and SK-BR-3) and mouse embryonic fibroblast (MEF) cells. Similar to the effects of YM155 and BIRC5 siRNA, downregulation of RAD54L and RAD51 by siRNA induced autophagy and DNA damage/strand breaks in cells, suggesting YM155/BIRC5 siRNA might also induce autophagy partly through RAD54L and RAD51 downregulations. We further observed that prolonged YM155 and BIRC5 siRNA treatment induced autophagic vesicle formation proximal to the nucleus and triggered DNA leakage. In conclusion, our findings reveal a novel mechanism of action of YM155 (i.e. induces autophagy-dependent ROS production-mediated DNA damage) in cancer cells and show the functional complexity of BIRC5 and autophagy involving the modulation of genome stability, highlighting that upregulation of autophagy is not always beneficial to the DNA repair process. Our findings can aid the development of a variety of BIRC5-directly/indirectly targeted anticancer therapies that are currently under pre-clinical and clinical investigations.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Repair/drug effects , Genomic Instability/drug effects , Imidazoles/pharmacology , Naphthoquinones/pharmacology , Neoplasms/drug therapy , Survivin/genetics , Autophagy/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Humans , Neoplasms/genetics , Neoplasms/metabolism , Reactive Oxygen Species/metabolism
10.
Front Cell Dev Biol ; 8: 485, 2020.
Article in English | MEDLINE | ID: mdl-32695778

ABSTRACT

BACKGROUND: Drug resistance is frequently found in estrogen receptor-positive (ER+) breast cancer patients during and after prolonged tamoxifen treatment. Although tamoxifen rechallenge has been proposed for treating recurrent breast tumors, the clinical benefit of this treatment is still controversial. The aims of this study are to identify the possible tamoxifen cytotoxicity-resistant subpopulation of MCF7 cells and to determine the effects of tamoxifen rechallenge on these cells. METHODS: Western blot analysis was used to determine the expression levels of various epithelial-mesenchymal transition- and cell survival/proliferation-related proteins in MCF7 and MCF7-derived, tamoxifen-mediated cytotoxicity-resistant MCF7-TAM12.5 breast cancer cells. Wound healing, Transwell migration, and invasion assays were used to examine the metastatic potential of cells. Clonogenic assays, trypan blue exclusion assays, and bromodeoxyuridine assays were used to examine clonogenicity and to determine the proliferation rate of cells. RESULTS: We found that MCF7-TAM12.5 cells exhibited higher tolerance to tamoxifen-mediated cytotoxicity, higher metastatic potential, higher expression levels of XIAP, and lower expression levels of ERα/ERß/HER2/Smac than MCF7 cells. In addition, MCF7 cells endogenously expressed Bcl-2α, whereas MCF7-TAM12.5 cells only expressed Bcl-2ß. Interestingly, tamoxifen rechallenge decreased the metastatic potential but increased the proliferation and clonogenicity of MCF7-TAM12.5 cells. At the molecular level, tamoxifen rechallenge upregulated the expression of phosphorylated Aurora A and Aurora B kinase in MCF7-TAM12.5 cells. CONCLUSION: Our findings further support the existence of highly heterogenetic cancer cell populations in ER+ breast tumors. It will be of clinical importance to determine the protein expression and the genetic profiles of tamoxifen-resistant/recurrent ER+ breast tumors to predict the potential effects of tamoxifen readministration in the future.

11.
Toxicol Appl Pharmacol ; 401: 115080, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32497533

ABSTRACT

Upregulation of ABCB1/MDR1 (P-gp) and BIRC5/Survivin promotes multidrug resistance in a variety of human cancers. LCL161 is an anti-cancer DIABLO/SMAC mimetic currently being tested in patients with solid tumors, but the molecular mechanism of action of LCL161 in cancer cells is still incompletely understood. It is still unclear whether LCL161 is therapeutically applicable for patients with ABCB1-overexpressing multidrug resistant tumors. In this study, we found that the potency of LCL161 is not affected by the expression of ABCB1 in KB-TAX50, KB-VIN10, and NTU0.017 cancer cells. Besides, LCL161 is equally potent towards the parental MCF7 breast cancer cells and its BIRC5 overexpressing, hormone therapy resistance subline MCF7-TamC3 in vitro. Mechanistically, we found that LCL161 directly modulates the ABCB1-ATPase activity and inhibits ABCB1 multi-drug efflux activity at low cytotoxic concentrations (i.e. 0.5xIC50 or less). Further analysis revealed that LCL161 also decreases intracellular ATP levels in part through BIRC5 downregulation. Therapeutically, co-treatment with LCL161 at low cytotoxic concentrations restored the sensitivity to the known ABCB1 substrate, paclitaxel, in ABCB1-expressing cancer cells and increased the sensitivity to tamoxifen in MCF7-TamC3 cells. In conclusion, LCL161 has the potential for use in the management of cancer patients with ABCB1 and BIRC5-related drug resistance. The findings of our study provide important information to physicians for designing a more "patient-specific" LCL161 clinical trial program in the future.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Mitochondrial Proteins/pharmacology , Survivin/antagonists & inhibitors , Thiazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adenosine Triphosphatases/metabolism , Antineoplastic Agents/chemistry , Apoptosis Regulatory Proteins/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mitochondrial Proteins/chemistry , Protein Structure, Secondary , Survivin/biosynthesis , Survivin/genetics , Thiazoles/chemistry
12.
J Biomed Sci ; 27(1): 31, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-32019552

ABSTRACT

X-linked inhibitor of apoptosis protein (XIAP), survivin, and BRUCE are members of the inhibitor-of-apoptosis protein (IAP) family known for their inhibitory effects on caspase activity and dysregulation of these molecules has widely been shown to cause embryonic defects and to promote tumorigenesis in human. Besides the anti-apoptotic functions, recent discoveries have revealed that XIAP, survivin, and BRUCE also exhibit regulatory functions for autophagy in cells. As the role of autophagy in human diseases has already been discussed extensively in different reviews; in this review, we will discuss the emerging autophagic role of XIAP, survivin, and BRUCE in cancer cells. We also provide an update on the anti-apoptotic functions and the roles in maintaining DNA integrity of these molecules. Second mitochondria-derived activator of caspases (Smac) is a pro-apoptotic protein and IAPs are the molecular targets of various Smac mimetics currently under clinical trials. Better understanding on the functions of XIAP, survivin, and BRUCE can enable us to predict possible side effects of these drugs and to design a more "patient-specific" clinical trial for Smac mimetics in the future.


Subject(s)
Apoptosis/genetics , Inhibitor of Apoptosis Proteins/physiology , Survivin/physiology , X-Linked Inhibitor of Apoptosis Protein/physiology , Autophagy , Humans , Inhibitor of Apoptosis Proteins/genetics , Survivin/genetics , Tumor Cells, Cultured , X-Linked Inhibitor of Apoptosis Protein/genetics
13.
Autophagy ; 16(7): 1296-1313, 2020 07.
Article in English | MEDLINE | ID: mdl-31612776

ABSTRACT

BIRC5/Survivin is known as a dual cellular functions protein that directly regulates both apoptosis and mitosis in embryonic cells during embryogenesis and in cancer cells during tumorigenesis and tumor metastasis. However, BIRC5 has seldom been demonstrated as a direct macroautophagy/autophagy regulator in cells. ATG7 expression and ATG12-ATG5-ATG16L1 complex formation are crucial for the phagophore elongation during autophagy in mammalian cells. In this study, we observed that the protein expression levels of BIRC5 and ATG7 were inversely correlated, whereas the expression levels of BIRC5 and SQSTM1/p62 were positively correlated in normal breast tissues and tumor tissues. Mechanistically, we found that BIRC5 negatively modulates the protein stability of ATG7 and physically binds to the ATG12-ATG5 conjugate, preventing the formation of the ATG12-ATG5-ATG16L1 protein complex in human cancer (MDA-MB-231, MCF7, and A549) and mouse embryonic fibroblast (MEF) cells. We also observed a concurrent physical dissociation between BIRC5 and ATG12-ATG5 (but not CASP3/caspase-3) and upregulation of autophagy in MDA-MB-231 and A549 cells under serum-deprived conditions. Importantly, despite the fact that upregulation of autophagy is widely thought to promote DNA repair in cells under genotoxic stress, we found that BIRC5 maintains DNA integrity through autophagy negative-modulations in both human cancer and MEF cells under non-stressed conditions. In conclusion, our study reveals a novel role of BIRC5 in cancer cells as a direct regulator of autophagy. BIRC5 may act as a "bridging molecule", which regulates the interplay between mitosis, apoptosis, and autophagy in embryonic and cancer cells. ABBREVIATIONS: ACTA1: actin; ATG: autophagy related; BIRC: baculoviral inhibitor of apoptosis repeat-containing; BAF: bafilomycin A1; CQ: chloroquine; CASP3: caspase 3; HSPB1/Hsp27: heat shock protein family B (small) member 1/heat shock protein 27; IAPs: inhibitors of apoptosis proteins; IP: immunoprecipitation; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; PLA: proximity ligation assay; SQSTM1/p62: sequestosome 1; siRNA: small interfering RNA.


Subject(s)
Autophagy-Related Protein 12/metabolism , Autophagy-Related Protein 5/metabolism , Autophagy , DNA Damage , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Neoplasms/metabolism , Survivin/metabolism , Animals , Cell Line, Tumor , Culture Media, Serum-Free , Down-Regulation , Humans , Mice , Neoplasms/pathology , Protein Binding , Protein Processing, Post-Translational , Protein Stability
14.
Sci Rep ; 9(1): 16359, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31704974

ABSTRACT

Breast cancer is the most common cancer in women, and some patients develop recurrence after standard therapy. Effective predictors are urgently needed to detect recurrence earlier. The activation of Hedgehog signaling in breast cancer is correlated with poor prognosis. PTCH1 is an essential membrane receptor of Hedgehog. However, there are few reports about mutations in Hedgehog genes in breast cancer. We conducted a comprehensive study via an experimental and bioinformatics approach to detect mutated genes in breast cancer. Twenty-two breast cancer patients who developed recurrence within 24 months postoperatively were enrolled with 22 control cancer patients. Targeted deep sequencing was performed to assess the mutations among individuals with breast cancer using a panel of 143 cancer-associated genes. Bioinformatics and public databases were used to predict the protein functions of the mutated genes. Mutations were identified in 44 breast cancer specimens, and the most frequently mutated genes were BRCA2, APC, ATM, BRCA1, NF1, TET2, TSC1, TSC2, NOTCH1, MSH2, PTCH1, TP53, PIK3CA, FBXW7, and RB1. Mutation of these genes was correlated with protein phosphorylation and autophosphorylation, such as peptidyl-tyrosine and protein kinase C phosphorylation. Among these highly mutated genes, mutations of PTCH1 were associated with poor prognosis and increased recurrence of breast cancer, especially mutations in exons 22 and 23. The public sequencing data from the COSMIC database were exploited to predict the functions of the mutations. Our findings suggest that mutation of PTCH1 is correlated with early recurrence of breast cancer patients and will become a powerful predictor for recurrence of breast cancer.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/surgery , Mutation , Neoplasm Recurrence, Local/diagnosis , Patched-1 Receptor/genetics , Adult , Aged , Breast Neoplasms/pathology , Case-Control Studies , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Incidence , Middle Aged , Neoplasm Metastasis , Neoplasm Recurrence, Local/epidemiology , Neoplasm Recurrence, Local/genetics , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Survival Rate , Taiwan/epidemiology
15.
Aging Cell ; 18(4): e12989, 2019 08.
Article in English | MEDLINE | ID: mdl-31183966

ABSTRACT

Multicellular signals are altered in the processes of both aging and neurodegenerative diseases, including Alzheimer's disease (AD). Similarities in behavioral and cellular functional changes suggest a common regulator between aging and AD that remains undetermined. Our genetics and behavioral approaches revealed the regulatory role of Akt in both aging and AD pathogenesis. In this study, we found that the activity of Akt is upregulated during aging through epidermal growth factor receptor activation by using the fruit fly as an in vivo model. Downregulation of Akt in neurons improved cell survival, locomotor activity, and starvation challenge in both aged and Aß42-expressing flies. Interestingly, increased cAMP levels attenuated both Akt activation-induced early death and Aß42-induced learning deficit in flies. At the molecular level, overexpression of Akt promoted Notch cleavage, suggesting that Akt is an endogenous activity regulator of γ-secretase. Taken together, this study revealed that Akt is involved in the aging process and Aß toxicity, and manipulating Akt can restore both neuronal functions and improve behavioral activity during the processes of aging and AD pathogenesis.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Drosophila melanogaster/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Aging/pathology , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/genetics , Animals , Animals, Genetically Modified , Cell Survival/genetics , Cyclic AMP/metabolism , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Locomotion/genetics , Longevity/genetics , Neurons/metabolism , Peptide Fragments/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Receptors, Notch/metabolism
16.
Protein Expr Purif ; 160: 73-83, 2019 08.
Article in English | MEDLINE | ID: mdl-31004782

ABSTRACT

Survivin is a well-known inhibitor-of-apoptosis proteins family member and a promising molecular target for anti-cancer treatment. However, it is widely accepted that survivin is only a "semi-druggable" target and development of survivin-specific small molecule inhibitors has shown to be difficult. In this study, we demonstrated that a histidine-tagged survivin T34A-C84A mutated protein (T34A-C84A-dNSur-His) can be produced using a bacterial recombinant protein expression system [E. coli ArcticExpress (DE3) cells] and solubilized using 1% (w/v) Sarkosyl. In addition, we showed that the purified T34A-C84A-dNSur-His protein formed dimers as predicted by in silico protein structure and molecular dynamics analysis. Importantly, results of the MTT assay revealed that the purified recombinant protein was biologically active in decreasing the viability of the human MDA-MB-231 breast adenocarcinoma and MIA-PaCa pancreatic carcinoma cells in vitro. Furthermore, the purified T34A-C84A-dNSur-His protein, but not of the histidine-peptide, induced apoptosis (i.e. caspase-9 activation and DNA fragmentation) in MDA-MB-231 cells at concentrations from 50 to 400 nM. In conclusion, our study provides a protocol of producing a biologically active survivin-targeting macromolecule, T34A-C84A-dNSur-His, which can be used as a tool for studying the molecular and cellular roles of survivin in cells. T34A-C84A-dNSur-His is also a potential therapeutic agent for augmenting cancer therapy.


Subject(s)
Cloning, Molecular , Escherichia coli/genetics , Survivin/genetics , Survivin/isolation & purification , Apoptosis/drug effects , Cell Line, Tumor , Escherichia coli/metabolism , Gene Expression , Humans , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Survivin/metabolism , Survivin/pharmacology
18.
FASEB J ; 32(3): 1375-1387, 2018 03.
Article in English | MEDLINE | ID: mdl-29127191

ABSTRACT

The endosomal-lysosomal system (ELS), autophagy, and ubiquitin-proteasome system (UPS) are cellular degradation pathways that each play a critical role in the removal of misfolded proteins and the prevention of the accumulation of abnormal proteins. Recent studies on Alzheimer's disease (AD) pathogenesis have suggested that accumulation of aggregated ß-amyloid (Aß) peptides in the AD brain results from a dysfunction in these cellular clearance systems. However, the specific roles of these pathways in the removal of Aß peptides and the pathogenesis underlying AD are unclear. Our in vitro and in vivo genetic approaches revealed that ELS mainly removed monomeric ß-amyloid42 (Aß42), while autophagy and UPS clear oligomeric Aß42. Although overproduction of phosphatidylinositol 4-phosphate-5 increased Aß42 clearance, it reduced the life span of Aß42 transgenic flies. Our behavioral studies further demonstrated impaired autophagy and UPS-enhanced Aß42-induced learning and memory deficits, but there was no effect on Aß42-induced reduction in life span. Results from genetic fluorescence imaging showed that these pathways were damaged in the following order: UPS, autophagy, and finally ELS. The results of our study demonstrate that different degradation pathways play distinct roles in the removal of Aß42 aggregates and in disease progression. These findings also suggest that pharmacologic treatments that are designed to stimulate cellular degradation pathways in patients with AD should be used with caution.-Ji, X.-R., Cheng, K.-C., Chen, Y.-R., Lin, T.-Y., Cheung, C. H. A., Wu, C.-L., Chiang, H.-C. Dysfunction of different cellular degradation pathways contributes to specific ß-amyloid42-induced pathologies.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Autophagy , Peptide Fragments/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila melanogaster , Humans , Peptide Fragments/genetics , Proteasome Endopeptidase Complex/genetics , Ubiquitin/genetics
19.
Front Pharmacol ; 8: 223, 2017.
Article in English | MEDLINE | ID: mdl-28529481

ABSTRACT

Alkali burn injury is a true ocular emergency of the conjunctiva and cornea that requires immediate precision. Lack of an immediate therapy can lead to a substantial damage in the ocular surface and anterior segment further causing visual impairment and disfigurement. We explored the regenerative capability of dominant negative survivin protein (SurR9-C84A) and histone deacetylase inhibitor trichostatin-A (TSA) in vivo, in a rat alkali burn model. A topical insult in rat eyes with NaOH led to degradation of the conjunctival and corneal epithelium. The integrity of the conjunctival and corneal tissue was increased by TSA and SurR9-C84A by improving the clathrin and claudin expressions. Wound healing was initiated by an increase in TGF-beta-1 and, increased endogenous survivin which inhibited apoptosis post-TSA and SurR9-C84A treatments. Protein expressions of fibronectin and alpha-integrin 5 were found to increase promoting corneal integrity. The cytokine analysis confirmed increased expressions of IL-1beta, IL-6, IL-12, IL-13, IFN-gamma, TNF-alpha, GMCSF, Rantes, and MMP-2 in injured cornea, which were found to be significantly downregulated by the combined treatment of SurR9-C84A and TSA. The ocular and systemic pharmacokinetic (PK) parameters were measured post-topical ocular administration of TSA and SurR9-C84A. The SurR9-C84A and TSA sustained relatively longer in the cornea, conjunctiva, and aqueous humor than in the tear fluid and plasma. Our results confirmed that a combination of TSA with SurR9-C8A worked in synergy and showed a promising healing and anti-inflammatory effect in a very short time against alkali burn. Therefore, a combination of TSA and SurR9-C84A can fulfill the need for an immediate response to wound healing in alkali burnt cornea. We also synthesized ultra-small chitosan nanoparticles (USC-NPs) targeted with alpha-SMA antibodies that can be used for delivery of TSA and SurR9-C84A specifically to the ocular burn site.

20.
Front Pharmacol ; 8: 902, 2017.
Article in English | MEDLINE | ID: mdl-29326587

ABSTRACT

Intrinsic or acquired resistance to hormone therapy is frequently reported in estrogen receptor positive (ER+) breast cancer patients. Even though dysregulations of histone deacetylases (HDACs) are known to promote cancer cells survival, the role of different HDACs in the induction of hormone therapy resistance in ER+ breast cancer remains unclear. Survivin is a well-known pro-tumor survival molecule and miR-125a-5p is a recently discovered tumor suppressor. In this study, we found that ER+, hormone-independent, tamoxifen-resistant MCF7-TamC3 cells exhibit increased expression of HDAC2, HDAC5, and survivin, but show decreased expression of miR-125a-5p, as compared to the parental tamoxifen-sensitive MCF7 breast cancer cells. Molecular down-regulations of HDAC2, HDAC5, and survivin, and ectopic over-expression of miR-125a-5p, increased the sensitivity of MCF7-TamC3 cells to estrogen deprivation and restored the sensitivity to tamoxifen. The same treatments also further increased the sensitivity to estrogen-deprivation in the ER+ hormone-dependent ZR-75-1 breast cancer cells in vitro. Kaplan-Meier analysis and receiver operating characteristic curve analysis of expression cohorts of breast tumor showed that high HDAC2 and survivin, and low miR-125a-5p, expression levels correlate with poor relapse-free survival in endocrine therapy and tamoxifen-treated ER+ breast cancer patients. Further molecular analysis revealed that HDAC2 and HDAC5 positively modulates the expression of survivin, and negatively regulates the expression miR-125a-5p, in ER+ MCF7, MCF7-TamC3, and ZR-75-1 breast cancer cells. These findings indicate that dysregulations of HDAC2 and HDAC5 promote the development of hormone independency and tamoxifen resistance in ERC breast cancer cells in part through expression regulation of survivin and miR-125a-5p.

SELECTION OF CITATIONS
SEARCH DETAIL
...