Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Biomedicines ; 11(4)2023 Mar 26.
Article in English | MEDLINE | ID: mdl-37189629

ABSTRACT

Mitochondria-associated membranes (MAMs) regulate several cellular processes, including calcium homeostasis and mitochondrial function, and dynamics. While MAMs are upregulated in Alzheimer's disease (AD), the mechanisms underlying this increase remain unknown. A possible mechanism may include dysregulation of protein phosphatase 2A (PP2A), which is reduced in the AD brain. Furthermore, PP2A has been previously reported to modulate MAM formation in hepatocytes. However, it is unknown whether PP2A and MAMs are linked in neuronal cells. Here, to test the correlation between PP2A and MAMs, we inhibited the activity of PP2A to mimic its low levels in AD brains and observed MAM formation, function, and dynamics. MAMs were significantly increased after PP2A inhibition, which correlated with elevated mitochondrial Ca2+ influx and disrupted mitochondrial membrane potential and mitochondrial fission. This study highlights the essential role PP2A plays in regulating MAM formation and mitochondrial function and dynamics for the first time in neuronal-like cells.

2.
Biomedicines ; 10(11)2022 Oct 23.
Article in English | MEDLINE | ID: mdl-36359200

ABSTRACT

Chronically activated microglia and brain vascular damage are major causes of neuroinflammation. The aim of this study was to determine the anti-inflammatory effects of nitro capsaicin, a newly modified capsaicin with less irritating characteristics, against microglial activation and brain microvascular endothelial cell damage. Using the SIMA9 microglia cell line, we found that nitro capsaicin reduced nitric oxide (NO) production in LPS-activated microglia better than its parent compound, capsaicin. Nitro capsaicin also decreased the expression of proinflammatory cytokines (TNF-α, IL-1ß, and IL-6) and enhanced the levels of anti-inflammatory factors, IL-4 and IL-10, both at the mRNA and protein levels. In the TNF-α-induced vascular damage model, nitro capsaicin decreased expression and secretion of the proinflammatory cytokines IL-1ß and IL-6. Phosphorylated NF-κB p65, a key transcription factor that stimulates the signaling of inflammatory pathways, was also reduced in the presence of nitro capsaicin, suggesting that the anti-inflammatory effects of nitro capsaicin were created through reducing NF-κB activation. Together, we concluded that nitro capsaicin has the potential to be further developed as an anti-neuroinflammatory agent.

3.
Biomedicines ; 10(10)2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36289849

ABSTRACT

Uncontrolled and excessive microglial activation is known to contribute to inflammation-mediated neurodegeneration. Therefore, reducing neurotoxic microglial activation may serve as a new approach to preventing neurodegeneration. Here, we investigated the anti-inflammatory effects of panduratin A against microglial activation induced by lipopolysaccharides (LPS) in the SIMA9 microglial cell line. We initially examined the anti-inflammatory properties of panduratin A by measuring LPS-induced nitric oxide (NO) production and the levels of pro-inflammatory cytokines (TNF-α, IL-1ß, and IL-6). Panduratin A significantly reduced NO levels and pro-inflammatory cytokines' production and secretion. In addition, panduratin A enhanced the production of anti-inflammatory cytokines IL-4 and IL-10. The anti-inflammatory effects of panduratin A are related to the suppression of the NF-κB signaling pathway. Together, these results demonstrate the anti-inflammatory properties of panduratin A against LPS-induced microglial activation, suggesting panduratin A has the potential to be further developed as a new agent for the prevention of neuroinflammation-associated neurodegenerative diseases.

4.
Asian Pac J Cancer Prev ; 23(7): 2379-2386, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35901345

ABSTRACT

OBJECTIVE: Glioblastoma is the most aggressive and lethal brain tumor in adults with highly invasive properties. In this present study, we explored the effects of Phyllanthus taxodiifolius Beille extract on molecules known to be hallmarks of aggressive glioblastoma including N-cadherin and vimentin, mesenchymal markers, as well as paxillin, a major adaptor protein that regulates the linking of focal adhesions to the actin cytoskeleton. METHODS: P. taxodiifolius were air-dried, powdered and percolated with methanol, filtered, concentrated and lyophilized to yield a crude methanol extract. C6 glioblastoma cell line was used in this study. The expression of N-cadherin and vimentin, as well as the activation of paxillin was determined using Western blot analysis. The effect of the extract on focal adhesions and actin cytoskeleton were investigated using immunofluorescence staining and confocal imaging. RESULTS: In the presence of 40 µg/ml Phyllanthus taxodiifolius Beille extract, the expression of N-cadherin and vimentin were significantly decreased (p<0.001 and p<0.05, respectively). Activation of paxillin was also diminished as indicated by a reduction of phosphorylated-paxillin (p<0.01). Consequently, actin stress fibers in glioblastoma cells were abolished as evidenced by the decrease in focal adhesion (p<0.001) and stress fibers numbers (p<0.001). CONCLUSION: Our study demonstrates for the first time that P. taxodiifolius interferes with multiple key molecules related to pathological hallmarks of glioblastoma. These molecules are involved with cell contacts, focal adhesions, and the formation and stabilization of actin stress fibers, which are required for glioblastoma metastatic behavior. These results provide further evidence supporting the potential of P. taxodiifolius and its bioactive compounds as anti-cancer agents.


Subject(s)
Glioblastoma , Phyllanthus , Actins/metabolism , Cadherins/metabolism , Cell Adhesion , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glioblastoma/pathology , Humans , Methanol , Paxillin/metabolism , Paxillin/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Phyllanthus/metabolism , Plant Extracts/pharmacology , Stress Fibers/metabolism , Stress Fibers/pathology , Vimentin
5.
Neuropsychopharmacol Rep ; 42(3): 281-287, 2022 09.
Article in English | MEDLINE | ID: mdl-35733332

ABSTRACT

Cognitive impairment is a prominent cause of disability in schizophrenia. Although antipsychotic drugs can rescue the psychotic symptoms, the cognitive impairments persist, with no treatment available. Alterations of BDNF, VEGF, TNF-α, and S100B have been linked to cognitive impairment in several neurological disorders. However, it remains unclear whether their levels are correlated with the cognitive functions of schizophrenia patients. Forty-one chronic, medicated schizophrenia patients were included in this study. Enzyme-linked, immunosorbent assays were used to measure the serum concentrations of BDNF, VEGF, TNF-α, and S100B. Associations between serum protein levels and various domains of the cognitive functions of the schizophrenia patients were observed. We found significant, positive correlations between serum BDNF and the processing speed and attention levels of the patients. Serum VEGF was also positively correlated with their memory and learning functions. In contrast, serum S100B and TNF-α were negatively correlated with the processing speed and attention of the schizophrenia patients. The findings warrant further investigation of these molecules as potential prognostic markers or treatment targets for cognitive impairment in schizophrenia patients.


Subject(s)
Antipsychotic Agents , Cognitive Dysfunction , Schizophrenia , Antipsychotic Agents/therapeutic use , Brain-Derived Neurotrophic Factor , Cognitive Dysfunction/etiology , Humans , Immunosorbents/therapeutic use , S100 Calcium Binding Protein beta Subunit , Schizophrenia/complications , Schizophrenia/drug therapy , Tumor Necrosis Factor-alpha , Vascular Endothelial Growth Factor A/therapeutic use
6.
PLoS One ; 17(4): e0266044, 2022.
Article in English | MEDLINE | ID: mdl-35377896

ABSTRACT

Colorectal cancer is the leading cause of cancer-related deaths worldwide, warranting the urgent need for a new treatment option. Plant-derived nanovesicles containing bioactive compounds represent new therapeutic avenues due to their unique characteristics as natural nanocarriers for bioactive molecules with therapeutic effects. Recent evidence has revealed potential anticancer activity of bioactive compounds from Boesenbergia rotunda (L.) Mansf. (fingerroot). However, the effect and the underlying mechanisms of fingerroot-derived nanovesicles (FDNVs) against colorectal cancer are still unknown. We isolated the nanovesicles from fingerroot and demonstrated their anticancer activity against two colorectal cancer cell lines, HT-29 and HCT116. The IC50 values were 63.9 ± 2.4, 57.8 ± 4.1, 47.8 ± 7.6 µg/ml for HT-29 cells and 57.7 ± 6.6, 47.2 ± 5.2, 34 ± 2.9 µg/ml for HCT116 cells at 24, 48, and 72 h, respectively. Interestingly, FDNVs were not toxic to a normal colon epithelial cell line, CCD 841 CoN. FDNVs exhibited selective uptake by the colorectal cancer cell lines but not the normal colon epithelial cell line. Moreover, dose- and time-dependent FDNV-induced apoptosis was only observed in the colorectal cancer cell lines. In addition, reactive oxygen species levels were substantially increased in colorectal cancer cells, but total glutathione decreased after treatment with FDNVs. Our results show that FDNVs exhibited selective anticancer activity in colorectal cancer cell lines via the disruption of intracellular redox homeostasis and induction of apoptosis, suggesting the utility of FDNVs as a novel intervention for colorectal cancer patients.


Subject(s)
Colorectal Neoplasms , Zingiberaceae , Apoptosis , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , HCT116 Cells , HT29 Cells , Humans
7.
Cell Mol Neurobiol ; 42(7): 2337-2353, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34059943

ABSTRACT

The Cath.a-differentiated (CAD) cell line is a central nervous system-derived catecholaminergic cell line originating from tyrosine hydroxylase (TH)-producing neurons located around the locus coeruleus area of the mouse brain. CAD cells have been used as an in vitro model for cellular and molecular studies due to their ability to differentiate under serum-free media conditions. However, the lack of serum-derived survival factors, limits the longevity for differentiated CAD cells to be maintained in healthy conditions; thereby, limiting their use in long-term culture studies. Here, we present a novel differentiation method that utilizes dexamethasone (Dex), a synthetic glucocorticoid receptor agonist. Specifically, we discovered that the addition of 100 µM of Dex into the 1% fetal bovine serum (FBS)-supplemented media effectively induced neuronal differentiation of CAD cells, as characterized by neurite formation and elongation. Dex-differentiated CAD cells exited the cell cycle, stopped proliferating, extended the neurites, and expressed neuronal markers. These effects were dependent on the glucocorticoid receptors (GR) as they were abolished by GR knockdown. Importantly, Dex-differentiated CAD cells showed longer survival duration than serum-free differentiated CAD cells. In addition, RNA-sequencing and qPCR data demonstrate that several genes involved in proliferation, neuronal differentiation, and survival pathways were differentially expressed in the Dex-differentiated cells. This is the first study to reveal Dex as a novel differentiation methodology used to generate postmitotic neuronal CAD cells, which may be utilized as an in vitro neuronal model for cellular and molecular neurobiology research.


Subject(s)
Central Nervous System , Neurites , Animals , Cell Differentiation , Dexamethasone , Mice , Neurons , Receptors, Glucocorticoid
9.
Exp Hematol ; 99: 12-20.e3, 2021 07.
Article in English | MEDLINE | ID: mdl-34077792

ABSTRACT

Red blood cell production, or erythropoiesis, is a proliferative process that requires tight regulation. Erythropoietin (Epo) is a glycoprotein cytokine that plays a major role in erythropoiesis by triggering erythroid progenitors/precursors of varying sensitivity. The concentration of Epo in bone marrow is hypothesized to be suboptimal, and the survival of erythroid cells has been suggested to depend on Epo sensitivity. However, the key factors that control Epo sensitivity remain unknown. Two types of transferrin receptors (TfRs), TfR1 and TfR2, are known to play a role in iron uptake in erythroid cells. Here, we hypothesized that TfRs may additionally modulate Epo sensitivity during erythropoiesis by modulating Epo receptor (EpoR) signaling. Using an Epo-sensitive UT-7 (UT7/Epo) erythroid cell and human erythroid progenitor cell models, we report that iron-loaded transferrin, that is, holo-transferrin (holo-Tf), synergizes with suboptimal Epo levels to improve erythroid cell survival, proliferation, and differentiation. This is accomplished via the major signaling pathways of erythropoiesis, which include signal transducer and activator of transcription 5 (STAT5), mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK), and phosphoinositide-3-kinase (PI3K)/AKT. Furthermore, we found that this cooperation is improved by, but does not require, the internalization of TfR1. Interestingly, we observed that loss of TfR2 stabilizes EpoR levels and abolishes the beneficial effects of holo-Tf. Overall, these data reveal novel signaling properties of TfRs, which involve the regulation of erythropoiesis through EpoR signaling.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation/drug effects , Erythroblasts/metabolism , Erythropoietin/pharmacology , Iron/pharmacology , MAP Kinase Signaling System/drug effects , Receptors, Transferrin/metabolism , Transferrin/pharmacology , Cell Line , Cell Survival/drug effects , Erythropoietin/metabolism , Humans , Iron/metabolism , Transferrin/metabolism
10.
Mol Psychiatry ; 26(6): 2402-2414, 2021 06.
Article in English | MEDLINE | ID: mdl-32327735

ABSTRACT

While several therapeutic strategies exist for depression, most antidepressant drugs require several weeks before reaching full biochemical efficacy and remission is not achieved in many patients. Therefore, biomarkers for depression and drug-response would help tailor treatment strategies. This study made use of banked human lymphoblast cell lines (LCLs) from normal and depressed subjects; the latter divided into remitters and non-remitters. Due to the fact that previous studies have shown effects on growth factors, cytokines, and elements of the cAMP-generating system as potential biomarkers for depression and antidepressant action, these were examined in LCLs. Initial gene and protein expression profiles for signaling cascades related to neuroendocrine and inflammatory functions differ among the three groups. Growth factor genes, including VEGFA and BDNF were significantly down-regulated in cells from depressed subjects. In addition, omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been reported to act as both antidepressants and anti-inflammatories, but the mechanisms for these effects are not established. Here we showed that n-3 PUFAs and escitalopram (selective serotonin reuptake inhibitors, SSRIs) treatment increased adenylyl cyclase (AC) and BDNF gene expression in LCLs. These data are consistent with clinical observations showing that n-3 PUFA and SSRI have antidepressant affects, which may be additive. Contrary to observations made in neuronal and glial cells, n-3 PUFA treatment attenuated cAMP accumulation in LCLs. However, while lymphoblasts show paradoxical responses to neurons and glia, patient-derived lymphoblasts appear to carry potential depression biomarkers making them an important tool for studying precision medicine in depressive patients. Furthermore, these data validate usefulness of n-3 PUFAs in treatment for depression.


Subject(s)
Fatty Acids, Omega-3 , Selective Serotonin Reuptake Inhibitors , Antidepressive Agents/pharmacology , Biomarkers , Cell Line , Depression , Humans , Selective Serotonin Reuptake Inhibitors/pharmacology
11.
ACS Chem Neurosci ; 11(11): 1634-1642, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32374999

ABSTRACT

Neurodegenerative diseases are irreversible conditions that result in progressive degeneration and death of nerve cells. Although the underlying mechanisms may vary, oxidative stress is considered to be one of the major causes of neuronal loss. Importantly, there are still no comprehensive treatments to completely cure these diseases. Therefore, protecting neurons from oxidative damage may be the most effective therapeutic strategy. Here we report a neuroprotective effects of a novel hybrid compound (dlx-23), obtained by conjugating α-lipoic acid (ALA), a natural antioxidant agent, and 3-n-butylphthalide (NBP), a clinical anti-ischemic drug. Dlx-23 protected against neuronal death induced by both H2O2 induced oxidative stress in Cath.-a-differentiated (CAD) cells and 6-OHDA, a toxin model of Parkinson's disease (PD) in SH-SY5Y cells. These activities proved to be more potent than the parent compound (ALA) alone. Dlx-23 scavenged free radicals, increased glutathione levels, and prevented mitochondria damage. In addition, live imaging of primary cortical neurons demonstrated that dlx-23 protected against neuronal growth cone damage induced by H2O2. Taken together these results suggest that dlx-23 has substantial potential to be further developed into a novel neuroprotective agent against oxidative damage and toxin induced neurodegeneration.


Subject(s)
Neuroprotective Agents , Thioctic Acid , Benzofurans , Cell Line, Tumor , Hydrogen Peroxide/toxicity , Neurons , Neuroprotective Agents/pharmacology , Oxidative Stress , Oxidopamine/toxicity , Reactive Oxygen Species , Thioctic Acid/pharmacology
12.
Biomed Pharmacother ; 112: 108645, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30798125

ABSTRACT

Glioblastoma is the most common and the most malignant form of brain tumor. This devastating tumor results in death within a year after diagnosis. Although the tumor mass can be surgically removed, glioma cells invade other areas in the brain leading to tumor recurrence and poor prognosis. Therefore, new agents that can overcome cancer cell invasion are urgently required. Phyllanthus taxodiifolius Beille (P. taxodiifolius), has been reported to have potent anti-cancer activities. However, its effects on glioblastoma cells and its underlying mechanisms have never been revealed. Here we investigated the effect and underlying mechanisms of P. taxodiifolius extract on aggressive properties of the glioblastoma, including adhesion, migration, and invasion. P. taxodiifolius extract disrupted adhesion, delayed migration and interfered with the invasion of glioblastoma cells. In addition, the extract suppressed microtubule dynamics as shown by live imaging of a microtubule plus tip protein and decreased focal adhesion by decreasing focal adhesion kinase activity. Our study is the first evidence showing that P. taxodiifolius extract suppresses invasive properties of glioblastoma cells by disrupting microtubule structure and interfering with microtubule dynamics, suggesting the possibility to further develop P. taxodiifolius and its bioactive compounds as an anti-cancer drug targeting microtubules in glioblastoma.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/pathology , Glioblastoma/pathology , Microtubules/drug effects , Phyllanthus/chemistry , Plant Components, Aerial/chemistry , Plant Extracts/pharmacology , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Microscopy, Confocal , Microtubules/ultrastructure , Neoplasm Invasiveness , Plant Extracts/isolation & purification , Rats
13.
Sci Rep ; 8(1): 7924, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29784906

ABSTRACT

The Wnt/ß-catenin signaling pathway plays a key role in the progression of human colorectal cancers (CRCs) and is one of the leading targets of chemotherapy agents developed for CRC. The present study aimed to investigate the anti-cancer effects and molecular mechanisms of 19-O-triphenylmethyl andrographolide (RS-PP-050), an andrographolide analogue and determine its activity in the Wnt/ß-catenin pathway. RS-PP-050 was found to potently inhibit the proliferation and survival of HT-29 CRC cells. It induces cell cycle arrest and promotes apoptotic cell death which was associated with the activation of PARP-1 and p53. Furthermore, RS-PP-050 exerts inhibitory effects on ß-catenin transcription by suppressing T-cell factor/lymphocyte enhancer factor (TCF/LEF) activity in cells overexpressing ß-catenin and by down-regulating the endogenous expression of Wnt target genes. RS-PP-050 also decreased the protein expression of the active form of ß-catenin but functions independently of GSK-3ß, a negative regulator of Wnt. Interestingly, RS-PP-050 extensively blocks phosphorylation at Ser675 of ß-catenin which links to interference of the nuclear translocation of ß-catenin and might contribute to Wnt inactivation. Collectively, our findings reveal the underlying anti-cancer mechanism of an andrographolide analogue and provide useful insight for exploiting a newly chemotherapeutic agent in Wnt/ß-catenin-overexpressing CRC cells.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/pathology , Diterpenes/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Antineoplastic Agents/chemistry , Apoptosis , Cell Cycle Checkpoints , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Humans , Phosphorylation , Wnt Proteins/genetics , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
14.
Biomed Pharmacother ; 101: 414-421, 2018 May.
Article in English | MEDLINE | ID: mdl-29501763

ABSTRACT

Hyperactivation of Wnt/ß-catenin signaling implicated in oncogenesis of colorectal cancer (CRC) is a potential molecular target for chemotherapy. An andrographolide analogue, 3A.1 (19-tert-butyldiphenylsilyl-8, 17-epoxy andrographolide) has previously been reported to be potently cytotoxic toward cancer cells by unknown molecular mechanisms. The present study explored the anti-cancer activity of analogue 3A.1 on Wnt/ß-catenin signaling in colon cancer cells (HT29 cells) which were more sensitive to the others (HCT116 and SW480 cells). Analogue 3A.1 inhibited viability of HT29 cells with IC50 value of 11.1 ±â€¯1.4 µM at 24 h, which was more potent than that of the parent andrographolide. Analogue 3A.1 also suppressed the proliferation of HT29 cells and induced cell apoptosis in a dose-dependent manner. Its apoptotic activity was accompanied with increased expressions of proteins related to DNA damages; PARP-1 and γ-H2AX. In addition, analogue 3A.1 significantly inhibited T-cell factor and lymphoid enhancer factor (TCF/LEF) promoter activity of Wnt/ß-catenin signaling. Accordingly, the expressions of Wnt target genes and ß-catenin protein were suppressed. Moreover, analogue 3A.1 increased the activity of GSK-3ß kinase, which is a negative regulator responsible for degradation of intracellular ß-catenin. This mode of action was further supported by the absence of the effects after treatment with a GSK-3ß inhibitor, and over-expression of a mutant ß-catenin (S33Y). Our findings reveal, for the first time, an insight into the molecular mechanism of the anti-cancer activity of analogue 3A.1 through the inhibition of Wnt/ß-catenin/GSK-3ß pathway and provide a therapeutic potential of the andrographolide analogue 3A.1 in CRC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Diterpenes/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Colonic Neoplasms/metabolism , DNA Damage/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , HCT116 Cells , HEK293 Cells , HT29 Cells , Histones/metabolism , Humans , Poly (ADP-Ribose) Polymerase-1/metabolism
15.
RSC Adv ; 8(52): 29548-29554, 2018 Aug 20.
Article in English | MEDLINE | ID: mdl-35547324

ABSTRACT

A simple and efficient protocol was developed for the syntheses of oridonin analogues, i.e. 6,20-epoxy ent-kaurane diterpenoid analogues from oridonin via diethylaminosulfur trifluoride (DAST) promoted rearrangement, most of which exhibited superior anticancer activities compared with their precursor.

16.
Asian Pac J Cancer Prev ; 18(10): 2845-2851, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29072435

ABSTRACT

Gastric cancer is the most common cancer in Eastern Asia. Increasing chemoresistance and general systemic toxicities have complicated the current chemotherapy leading to an urgent need of more effective agents. The present study reported a potent DNA topoisomerase IIα inhibitory activity of an andrographolide analogue (19-triisopropyl andrographolide, analogue-6) in gastric cancer cells; MKN-45, and AGS cells. The analogue was potently cytotoxic to both gastric cancer cell lines with the half maximal inhibitory concentration (IC50 values) of 6.3±0.7 µM, and 1.7±0.05 µM at 48 h for MKN-45, and AGS cells, respectively. It was more potent than the parent andrographolide and the clinically used, etoposide with the IC50 values of >50 µM in MKN-45 and 11.3±2.9 µM in AGS cells for andrographolide and 28.5±4.4 µM in MKN-45 and 4.08±0.5 µM in AGS cells for etoposide. Analogue-6 at 2 µM significantly inhibited DNA topoisomerase IIα enzyme in AGS cells, induced DNA damage, activated cleaved PARP-1, and Caspase3 leading to late cellular apoptosis. Interestingly, the expression of tumor suppressor p53 was not activated. These results show the importance of 19-triisopropyl-andrographolide in its emerging selectivity to primary target on topoisomerase IIα enzyme, inducing DNA damage and apoptosis by p53- independent mechanism. Thereby, the results provide insights of the potential of 19-triisopropyl andrographolide as an anticancer agent for gastric cancer. The chemical transformation of andrographolide is a promising strategy in drug discovery of a novel class of anticancer drugs from bioactive natural products.

17.
J Neurosci ; 33(42): 16471-82, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24133252

ABSTRACT

Dendritic spines are actin-rich compartments that protrude from the microtubule-rich dendritic shafts of principal neurons. Spines contain receptors and postsynaptic machinery for receiving the majority of glutamatergic inputs. Recent studies have shown that microtubules polymerize from dendritic shafts into spines and that signaling through synaptic NMDA receptors regulates this process. However, the mechanisms regulating microtubule dynamics in dendrites and spines remain unclear. Here we show that in hippocampal neurons from male and female mice, the majority of microtubules enter spines from highly localized sites at the base of spines. These entries occur in response to synapse-specific calcium transients that promote microtubule entry into active spines. We further document that spine calcium transients promote local actin polymerization, and that F-actin is both necessary and sufficient for microtubule entry. Finally, we show that drebrin, a protein known to mediate interactions between F-actin and microtubules, acts as a positive regulator of microtubule entry into spines. Together these results establish for the first time the essential mechanisms regulating microtubule entry into spines and contribute importantly to our understanding of the role of microtubules in synaptic function and plasticity.


Subject(s)
Actins/metabolism , Calcium/metabolism , Dendritic Spines/metabolism , Microtubules/metabolism , Neuropeptides/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Cytoskeleton/metabolism , Dendrites/metabolism , Female , Hippocampus/metabolism , Male , Mice , Neuronal Plasticity/physiology , Neurons/metabolism
18.
Methods Cell Biol ; 115: 173-89, 2013.
Article in English | MEDLINE | ID: mdl-23973073

ABSTRACT

Microtubules, major components of the cytoskeleton, play important roles in a variety of cellular functions including mitosis, intracellular transport, and the modulation of cell morphology. Several studies have demonstrated that specific G-protein alpha subunits bind to tubulin with a high affinity (~130 nM) and elicit various functional effects on tubulin and microtubules. In this chapter, we present a description of the protocols for several methods that are used to determine the interaction between heterotrimeric G proteins and tubulin, as well as functional consequences of the interactions including protocols for protein purification, binding assays, tubulin GTPase assays, microtubule dynamics assays, and assays for cytoskeletal consequences of G-protein-coupled receptor signaling.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Microtubules/metabolism , Protein Array Analysis/methods , Surface Plasmon Resonance/methods , Tubulin/metabolism , Animals , Brain/cytology , Brain/metabolism , GTP Phosphohydrolases/metabolism , Protein Binding , Sheep , Signal Transduction
19.
Proc Natl Acad Sci U S A ; 110(29): 11857-62, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818590

ABSTRACT

Endocytic protein trafficking is directed by sorting signals on cargo molecules that are recognized by cytosolic adaptor proteins. However, the steps necessary to segregate the variety of cargoes during endocytosis remain poorly defined. Using Caenorhabditis elegans, we demonstrate that multiple plasma membrane endocytic adaptors function redundantly to regulate clathrin-mediated endocytosis and to recruit components of the endosomal sorting complex required for transport (ESCRT) machinery to the cell surface to direct the sorting of ubiquitin-modified substrates. Moreover, our data suggest that preassembly of cargoes with the ESCRT-0 complex at the plasma membrane enhances the efficiency of downstream sorting events in the endolysosomal system. In the absence of a heterooligomeric adaptor complex composed of FCHO, Eps15, and intersectin, ESCRT-0 accumulation at the cell surface is diminished, and the degradation of a ubiquitin-modified cargo slows significantly without affecting the rate of its clathrin-mediated internalization. Consistent with a role for the ESCRT machinery during cargo endocytosis, we further show that the ESCRT-0 complex accumulates at a subset of clathrin-coated pits on the surface of human cells. Our findings suggest a unique mechanism by which ubiquitin-modified cargoes are sequestered into the endolysosomal pathway.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Cell Membrane/metabolism , Endocytosis/physiology , Endosomal Sorting Complexes Required for Transport/metabolism , Animals , Caenorhabditis elegans , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoprecipitation , Mass Spectrometry , RNA Interference , Ubiquitin/metabolism
20.
J Cell Sci ; 126(Pt 11): 2411-23, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23572514

ABSTRACT

Cdc42-interacting protein 4 (CIP4), a member of the F-BAR family of proteins, plays important roles in a variety of cellular events by regulating both membrane and actin dynamics. In many cell types, CIP4 functions in vesicle formation, endocytosis and membrane tubulation. However, recent data indicate that CIP4 is also involved in protrusion in some cell types, including cancer cells (lamellipodia and invadopodia) and neurons (ribbed lamellipodia and veils). In neurons, CIP4 localizes specifically to extending protrusions and functions to limit neurite outgrowth early in development. The mechanism by which CIP4 localizes to the protruding edge membrane and induces lamellipodial/veil protrusion and actin rib formation is not known. Here, we show that CIP4 localization to the protruding edge of neurons is dependent on both the phospholipid content of the plasma membrane and the underlying organization of actin filaments. Inhibiting phosphatidylinositol (3,4,5)-trisphosphate (PIP3) production decreases CIP4 at the membrane. CIP4 localization to the protruding edge is also dependent on Rac1/WAVE1, rather than Cdc42/N-WASP. Capping actin filaments with low concentrations of cytochalasin D or by overexpressing capping protein dramatically decreases CIP4 at the protruding edge, whereas inactivating Arp2/3 drives CIP4 to the protruding edge. We also demonstrate that CIP4 dynamically colocalizes with Ena/VASP and DAAM1, two proteins known to induce unbranched actin filament arrays and play important roles in neuronal development. Together, this is the first study to show that the localization of an F-BAR protein depends on both actin filament architecture and phospholipids at the protruding edge of developing neurons.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Membrane/metabolism , Microtubule-Associated Proteins/metabolism , Neurites/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Minor Histocompatibility Antigens , Neuropeptides/metabolism , Phospholipids/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Transport/physiology , Wiskott-Aldrich Syndrome Protein Family/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...