Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 232
Filter
1.
Molecules ; 26(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34641328

ABSTRACT

This study aims to enhance efficacy and reduce toxicity of the combination treatment of a drug and curcumin (Cur) on leukemic stem cell and leukemic cell lines, including KG-1a and KG-1 (FLT3+ LSCs), EoL-1 (FLT3+ LCs), and U937 (FLT3- LCs). The cytotoxicity of co-treatments of doxorubicin (Dox) or idarubicin (Ida) at concentrations of the IC10-IC80 values and each concentration of Cur at the IC20, IC30, IC40, and IC50 values (conditions 1, 2, 3, and 4) was determined by MTT assays. Dox-Cur increased cytotoxicity in leukemic cells. Dox-Cur co-treatment showed additive and synergistic effects in several conditions. The effect of this co-treatment on FLT3 expression in KG-1a, KG-1, and EoL-1 cells was examined by Western blotting. Dox-Cur decreased FLT3 protein levels and total cell numbers in all the cell lines in a dose-dependent manner. In summary, this study exhibits a novel report of Dox-Cur co-treatment in both enhancing cytotoxicity of Dox and inhibiting cell proliferation via FLT3 protein expression in leukemia stem cells and leukemic cells. This is the option of leukemia treatment with reducing side effects of chemotherapeutic drugs to leukemia patients.


Subject(s)
Curcumin/pharmacology , Doxorubicin/pharmacology , Idarubicin/pharmacology , Leukemia, Myeloid, Acute/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Antigens, Neoplasm/drug effects , Antigens, Neoplasm/metabolism , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Curcuma/chemistry , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Rhizome/chemistry
2.
Cell Death Dis ; 12(11): 978, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675191

ABSTRACT

Colorectal cancers (CRC) can be classified into four consensus molecular subtypes (CMS), among which CMS1 has the best prognosis, contrasting with CMS4 that has the worst outcome. CMS4 CRC is notoriously resistant against therapeutic interventions, as demonstrated by preclinical studies and retrospective clinical observations. Here, we report the finding that two clinically employed agents, everolimus (EVE) and plicamycin (PLI), efficiently target the prototypic CMS4 cell line MDST8. As compared to the prototypic CMS1 cell line LoVo, MDST8 cells treated with EVE or PLI demonstrated stronger cytostatic and cytotoxic effects, increased signs of apoptosis and autophagy, as well as a more pronounced inhibition of DNA-to-RNA transcription and RNA-to-protein translation. Moreover, nontoxic doses of EVE and PLI induced the shrinkage of MDST8 tumors in mice, yet had only minor tumor growth-reducing effects on LoVo tumors. Altogether, these results suggest that EVE and PLI should be evaluated for their clinical activity against CMS4 CRC.


Subject(s)
Adaptor Proteins, Signal Transducing/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Cytoskeletal Proteins/drug effects , Everolimus/therapeutic use , Plicamycin/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Proliferation , Colorectal Neoplasms/pathology , Everolimus/pharmacology , Humans , Mice , Plicamycin/pharmacology
3.
Behav Brain Res ; 409: 113338, 2021 07 09.
Article in English | MEDLINE | ID: mdl-33940049

ABSTRACT

The α7 nicotinic acetylcholine receptor (α7 nAChR) is a potential target for the treatment of cognitive decline in patients with schizophrenia, Alzheimer's disease, and attention-deficit/hyperactivity disorder. Here we examined the promnesic activity of the α7 nAChR agonist (A582941), the type I (CCMI), and the type II (PNU120596) positive allosteric modulators (PAMs) in rats following single and repeated (once daily for seven days) treatment. To determine the neuronal mechanisms underlying the procognitive activity of the tested compounds, levels of the extracellular signal-regulated kinases (Erk1/2) and the activity-regulated cytoskeleton-associated protein (Arc) mRNAs were assessed in the frontal cortical and hippocampal brain regions. Using the novel object recognition test, we demonstrate that the lower doses of A582941 (0.1 mg/kg), CCMI (1 mg/kg), and PNU120596 (0.3 mg/kg) improved recognition memory after repeated but not single administration, suggesting a cumulative effect of repeated dosing. In contrast, the higher doses of A582941 (0.3 mg/kg), CCMI (3 mg/kg) and PNU120596 (1 mg/kg) demonstrated promnesic efficacy following both single and repeated administration. Subsequent in situ hybridization revealed that repeated treatment with A582941 and CCMI, but not PNU120596 enhanced mRNA expression of the Erk1/2 and Arc in the frontal cortex and hippocampus. Present data suggest that both the α7 nAChR agonist and PAMs exhibit procognitive effects after single and repeated administration. The increased level of the Erk1/2 and Arc genes is likely to be at least partially involved in this effect.


Subject(s)
Behavior, Animal/drug effects , Cytoskeletal Proteins/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Hippocampus/drug effects , Nerve Tissue Proteins/drug effects , Nicotinic Agonists/pharmacology , Nootropic Agents/pharmacology , Prefrontal Cortex/drug effects , Recognition, Psychology/drug effects , alpha7 Nicotinic Acetylcholine Receptor/drug effects , Animals , Male , Nicotinic Agonists/administration & dosage , Nootropic Agents/administration & dosage , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley
4.
Cell Rep ; 34(1): 108588, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406435

ABSTRACT

Axonal degeneration is responsible for disease progression and accumulation of disability in many neurodegenerative conditions. The axonal degenerative process can generate a metastable pool of damaged axons that remain structurally and functionally viable but fated to degenerate in the absence of external intervention. SARM1, an NADase that depletes axonal energy stores upon activation, is the central driver of an evolutionarily conserved program of axonal degeneration. We identify a potent and selective small molecule isoquinoline inhibitor of SARM1 NADase that recapitulates the SARM1-/- phenotype and protects axons from degeneration induced by axotomy or mitochondrial dysfunction. SARM1 inhibition post-mitochondrial injury with rotenone allows recovery and rescues axons that already entered the metastable state. We conclude that SARM1 inhibition with small molecules has the potential to treat axonopathies of the central and peripheral nervous systems by preventing axonal degeneration and by allowing functional recovery of a metastable pool of damaged, but viable, axons.


Subject(s)
Armadillo Domain Proteins/drug effects , Armadillo Domain Proteins/physiology , Axons/physiology , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/physiology , Isoquinolines/pharmacology , Animals , Biomarkers/metabolism , Cell Line , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NAD+ Nucleosidase/drug effects , NAD+ Nucleosidase/physiology , Nerve Degeneration/drug therapy , Neurodegenerative Diseases/drug therapy , Phenotype , Recovery of Function
5.
J Alzheimers Dis ; 79(1): 249-265, 2021.
Article in English | MEDLINE | ID: mdl-33252074

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is characterized by accumulation of amyloid-ß (Aß) species and deposition of senile plaques (SPs). Clinical trials with the anti-Aß antibody aducanumab have been completed recently. OBJECTIVE: To characterize the proteomic profile of SPs and surrounding tissue in a mouse model of AD in 10-month-old tgAPPPS1-21 mice after chronic treatment with aducanumab for four months with weekly dosing (10 mg/kg). METHODS: After observing significant reduction of SP numbers in hippocampi of aducanumab-treated mice, we applied a localized proteomic analysis by combining laser microdissection and liquid chromatography-tandem mass spectrometry (LC-MS/MS) of the remaining SPs in hippocampi. We microdissected three subregions, containing SPs, SP penumbra level 1, and an additional penumbra level 2 to follow the proteomic profile as gradient. RESULTS: In the aducanumab-treated mice, we identified 17 significantly regulated proteins that were associated with 1) mitochondria and metabolism (ACAT2, ATP5J, ETFA, EXOG, HK1, NDUFA4, NDUFS7, PLCB1, PPP2R4), 2) cytoskeleton and axons (ADD1, CAPZB, DPYSL3, MAG), 3) stress response (HIST1H1C/HIST1H1D, HSPA12A), and 4) AßPP trafficking/processing (CD81, GDI2). These pathways and some of the identified proteins are implicated in AD pathogenesis. Proteins associated with mitochondria and metabolism were mainly upregulated while proteins associated with AßPP trafficking/processing and stress response pathways were mainly downregulated, suggesting that aducanumab could lead to a beneficial proteomic profile around SPs in tgAPPPS1-21 mice. CONCLUSION: We identified novel proteomic patterns of SPs and surrounding tissue indicating that chronic treatment with aducanumab could inhibit Aß toxicity and increase phagocytosis and cell viability.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/drug effects , Amyloid beta-Protein Precursor/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Brain/drug effects , Plaque, Amyloid/metabolism , Proteome/drug effects , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Chromatography, Liquid , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/metabolism , Presenilin-1/genetics , Protein Transport/drug effects , Proteomics , Stress, Physiological/drug effects , Tandem Mass Spectrometry
6.
Cancer Med ; 9(16): 5999-6008, 2020 08.
Article in English | MEDLINE | ID: mdl-32627938

ABSTRACT

Circular RNAs (circRNAs) play vital roles in the pathogenesis and development of multiple cancers, including hepatocellular carcinoma (HCC). Nevertheless, the regulatory mechanisms of circ-SPECC1 in HCC remain poorly understood. In our study, we found that circ-SPECC1 was apparently downregulated in H2 O2 -treated HCC cells. Additionally, knockdown of circ-SPECC1 inhibited cell proliferation and promoted cell apoptosis of HCC cells under H2 O2 treatment. Moreover, circ-SPECC1 inhibited miR-33a expression by direct interaction, and miR-33a inhibitor partially reversed the effect of circ-SPECC1 knockdown on proliferation and apoptosis of H2 O2 -treated HCC cells. Furthermore, TGFß2 was demonstrated to be a target gene of miR-33a and TGFß2 overexpression rescued the phenotypes of HCC cells attenuated by miR-33a mimics. Meanwhile, autophagy inhibition by 3-methyladenine (3-MA) abrogated the effect of miR-33a mimics on proliferation and apoptosis of H2 O2 -treated HCC cells. Finally, knockdown of circ-SPECC1 hindered tumor growth in vivo. In conclusion, our study demonstrated that circ-SPECC1 regulated TGFß2 and autophagy to promote HCC tumorigenesis under oxidative stress via miR-33a. These findings might provide potential treatment strategies for patients with HCC.


Subject(s)
Autophagy , Carcinoma, Hepatocellular/etiology , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Liver Neoplasms/etiology , MicroRNAs/metabolism , Transforming Growth Factor beta2/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/prevention & control , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/genetics , Down-Regulation , Humans , Hydrogen Peroxide/pharmacology , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/antagonists & inhibitors , Oxidative Stress
7.
Neuropharmacology ; 171: 108084, 2020 07.
Article in English | MEDLINE | ID: mdl-32294462

ABSTRACT

In recent years, with the discovery and research of lactate-specific receptor HCAR1(hydroxycarboxylic acid receptor 1), lactate is not only as a product of Glycolysis in astrocytes, but also as a signaling molecule which has gradually received attention. Studies have found that lactate can be used as an intercellular signaling molecule involved in synaptic plasticity, and so that peripheral administration of lactate can produce antidepressant effects. Here, we focus on HCAR1 on the most widely distributed astrocytes in the brain, found and verified that lactate could cause Arc/arg3.1 protein overexpression in astrocytes through HCAR1. However, the expression of Arc/arg3.1 does not depend on the Gi protein pathway of HCAR1, and we found that lactate enhanced the expression of Arc/arg3.1 protein through the HCAR1-ß-arrestin2 pathway. In summary, lactate acts on HCAR1 of astrocytes. It enhances the expression of MAPK-dependent Arc through ß-arrestin2, thereby reducing the influx of calcium ions when astrocytes are exposed to glutamate damage, achieving the role of protecting astrocytes and indirectly enhancing the absorption of glutamate by astrocytes. These results also demonstrate that HCAR1 in the brain is a potential therapeutic target in an experimental in vitro model of glutamate damage, which is strongly associated with many neurodegenerative diseases.


Subject(s)
Astrocytes/drug effects , Cytoskeletal Proteins/drug effects , Lactic Acid/pharmacology , Nerve Tissue Proteins/drug effects , Receptors, G-Protein-Coupled/drug effects , Signal Transduction/drug effects , beta-Arrestin 2/drug effects , Animals , Calcium Signaling/drug effects , Cells, Cultured , Cytoskeletal Proteins/biosynthesis , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/drug effects , Nerve Tissue Proteins/biosynthesis , Neurodegenerative Diseases/drug therapy , Neuronal Plasticity/drug effects , Receptors, G-Protein-Coupled/biosynthesis
8.
Bosn J Basic Med Sci ; 20(3): 310-318, 2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32020845

ABSTRACT

Nowadays, the emergence of multidrug-resistant bacterial strains initiates the urgent need for the elucidation of the new drug targets for the discovery of antimicrobial drugs. Filamenting temperature-sensitive mutant Z (FtsZ), a eukaryotic tubulin homolog, is a GTP-dependent prokaryotic cytoskeletal protein and is conserved among most bacterial strains. In vitro studies revealed that FtsZ self-assembles into dynamic protofilaments or bundles and forms a dynamic Z-ring at the center of the cell in vivo, leading to septation and consequent cell division. Speculations on the ability of FtsZ in the blockage of cell division make FtsZ a highly attractive target for developing novel antibiotics. Researchers have been working on synthetic molecules and natural products as inhibitors of FtsZ. Accumulating data suggest that FtsZ may provide the platform for the development of novel antibiotics. In this review, we summarize recent advances in the properties of FtsZ protein and bacterial cell division, as well as in the development of FtsZ inhibitors.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , Bacterial Proteins/drug effects , Cytoskeletal Proteins/drug effects , Cell Division/drug effects , Drug Resistance, Bacterial , Humans
9.
Mem Inst Oswaldo Cruz ; 115: e190348, 2020.
Article in English | MEDLINE | ID: mdl-32049098

ABSTRACT

BACKGROUND: It was previously demonstrated that CMC-20, a nitazoxanide and N-methyl-1H-benzimidazole hybrid molecule, had higher in vitro activity against Giardia intestinalis WB strain than metronidazole and albendazole and similar to nitazoxanide. OBJETIVES: To evaluate the in vitro activity of CMC-20 against G. intestinalis strains with different susceptibility/resistance to albendazole and nitazoxanide and evaluate its effect on the distribution of parasite cytoskeletal proteins and its in vivo giardicidal activity. METHODS: CMC-20 activity was tested against two isolates from patients with chronic and acute giardiasis, an experimentally induced albendazole resistant strain and a nitazoxanide resistant clinical isolate. CMC-20 effect on the distribution of parasite cytoskeletal proteins was analysed by indirect immunofluorescence and its activity was evaluated in a murine model of giardiasis. FINDINGS CMC-20: showed broad activity against susceptible and resistant strains to albendazole and nitaxozanide. It affected the parasite microtubule reservoir and triggered the parasite encystation. In this process, alpha-7.2 giardin co-localised with CWP-1 protein. CMC-20 reduced the infection time and cyst load in feces of G. muris infected mice similar to albendazole. MAIN CONCLUSIONS: The in vitro and in vivo giardicidal activity of CMC-20 suggests its potential use in the treatment of giardiasis.


Subject(s)
Albendazole/pharmacology , Antiprotozoal Agents/pharmacology , Cytoskeletal Proteins/drug effects , Giardia lamblia/drug effects , Thiazoles/pharmacology , Albendazole/chemistry , Animals , Antiprotozoal Agents/chemistry , Fluorescent Antibody Technique, Indirect , Humans , Mice , Nitro Compounds , Parasitic Sensitivity Tests , Thiazoles/chemistry , Time Factors
10.
Neuropharmacology ; 162: 107803, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31580838

ABSTRACT

The most common injury of preterm infants is periventricular leukomalacia (PVL) and to date there is still no safe and effective treatment. In our previous studies, leptin has been found to have neuroprotective effects on the preterm ischemia-hypoxia brain damage model rats in animal behavior. To gain insight into the neuroprotective mechanisms of leptin on preterm brain damage model rats, we constructed a comparative peptidomic profiling of hippocampal tissue between leptin-treated after model and preterm ischemia-hypoxia brain damage model rats using a stable isobaric labeling strategy involving tandem mass tag reagents, followed by nano liquid chromatography tandem mass spectrometry. We identified and quantified 4164 peptides, 238 of which were differential expressed in hippocampal tissue in the two groups. A total of 150 peptides were up regulated and 88 peptides were down regulated. These peptides were imported into the Ingenuity Pathway Analysis (IPA) and identified putative roles in nervous system development, function and diseases. We concluded that the preterm ischemia-hypoxia brain damage model with leptin treatment induced peptides changes in hippocampus, and these peptides, especially for the peptides associated "microtubule-associated protein 1b (MAP1b), Elastin (Eln), Piccolo presynaptic cytomatrix protein (Pclo), Zinc finger homeobox 3(Zfhx3), Alpha-kinase 3(Alpk3) and Myosin XVA(Myo15a) ", could be candidate bio-active peptides and participate in neuroprotection of leptin. These may advance our current understanding of the mechanism of leptin's neuroprotective effect on preterm brain damage and may be involved in the etiology of preterm brain damage. Meanwhile, we found that repression of ILK signaling pathway plays a significant role in neuroprotection of leptin. A better understanding of the role of ILK signaling pathway in neuroprotective mechanisms will help scientists and researchers to develop selective, safe and efficacious drug for therapy against human nervous system disorders.


Subject(s)
Hippocampus/metabolism , Hypoxia-Ischemia, Brain/metabolism , Leptin/pharmacology , Neuroprotective Agents/pharmacology , Peptides/metabolism , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , Carotid Artery, Common , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Elastin/drug effects , Elastin/metabolism , Hippocampus/drug effects , Homeodomain Proteins/drug effects , Homeodomain Proteins/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Leukomalacia, Periventricular/metabolism , Leukomalacia, Periventricular/physiopathology , Ligation , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism , Myosins/drug effects , Myosins/metabolism , Neuropeptides/drug effects , Neuropeptides/metabolism , Peptides/drug effects , Protein Serine-Threonine Kinases , Rats , Signal Transduction
11.
Mem. Inst. Oswaldo Cruz ; 115: e190348, 2020. tab, graf
Article in English | LILACS | ID: biblio-1091246

ABSTRACT

BACKGROUND It was previously demonstrated that CMC-20, a nitazoxanide and N-methyl-1H-benzimidazole hybrid molecule, had higher in vitro activity against Giardia intestinalis WB strain than metronidazole and albendazole and similar to nitazoxanide. OBJETIVES To evaluate the in vitro activity of CMC-20 against G. intestinalis strains with different susceptibility/resistance to albendazole and nitazoxanide and evaluate its effect on the distribution of parasite cytoskeletal proteins and its in vivo giardicidal activity. METHODS CMC-20 activity was tested against two isolates from patients with chronic and acute giardiasis, an experimentally induced albendazole resistant strain and a nitazoxanide resistant clinical isolate. CMC-20 effect on the distribution of parasite cytoskeletal proteins was analysed by indirect immunofluorescence and its activity was evaluated in a murine model of giardiasis. FINDINGS CMC-20 showed broad activity against susceptible and resistant strains to albendazole and nitaxozanide. It affected the parasite microtubule reservoir and triggered the parasite encystation. In this process, alpha-7.2 giardin co-localised with CWP-1 protein. CMC-20 reduced the infection time and cyst load in feces of G. muris infected mice similar to albendazole. MAIN CONCLUSIONS The in vitro and in vivo giardicidal activity of CMC-20 suggests its potential use in the treatment of giardiasis.


Subject(s)
Humans , Animals , Mice , Thiazoles/pharmacology , Albendazole/pharmacology , Giardia lamblia/drug effects , Cytoskeletal Proteins/drug effects , Antiprotozoal Agents/pharmacology , Thiazoles/chemistry , Time Factors , Albendazole/chemistry , Fluorescent Antibody Technique, Indirect , Parasitic Sensitivity Tests , Antiprotozoal Agents/chemistry
12.
Mem. Inst. Oswaldo Cruz ; 115: e190348, 2020. tab, graf
Article in English | LILACS | ID: biblio-1056773

ABSTRACT

BACKGROUND It was previously demonstrated that CMC-20, a nitazoxanide and N-methyl-1H-benzimidazole hybrid molecule, had higher in vitro activity against Giardia intestinalis WB strain than metronidazole and albendazole and similar to nitazoxanide. OBJETIVES To evaluate the in vitro activity of CMC-20 against G. intestinalis strains with different susceptibility/resistance to albendazole and nitazoxanide and evaluate its effect on the distribution of parasite cytoskeletal proteins and its in vivo giardicidal activity. METHODS CMC-20 activity was tested against two isolates from patients with chronic and acute giardiasis, an experimentally induced albendazole resistant strain and a nitazoxanide resistant clinical isolate. CMC-20 effect on the distribution of parasite cytoskeletal proteins was analysed by indirect immunofluorescence and its activity was evaluated in a murine model of giardiasis. FINDINGS CMC-20 showed broad activity against susceptible and resistant strains to albendazole and nitaxozanide. It affected the parasite microtubule reservoir and triggered the parasite encystation. In this process, alpha-7.2 giardin co-localised with CWP-1 protein. CMC-20 reduced the infection time and cyst load in feces of G. muris infected mice similar to albendazole. MAIN CONCLUSIONS The in vitro and in vivo giardicidal activity of CMC-20 suggests its potential use in the treatment of giardiasis.


Subject(s)
Humans , Animals , Mice , Thiazoles/pharmacology , Albendazole/pharmacology , Giardia lamblia/drug effects , Cytoskeletal Proteins/drug effects , Antiprotozoal Agents/pharmacology , Thiazoles/chemistry , Time Factors , Albendazole/chemistry , Fluorescent Antibody Technique, Indirect , Parasitic Sensitivity Tests , Antiprotozoal Agents/chemistry
13.
JCI Insight ; 4(17)2019 09 05.
Article in English | MEDLINE | ID: mdl-31484833

ABSTRACT

Chemotherapy-induced peripheral neuropathy is one of the most prevalent dose-limiting toxicities of anticancer therapy. Development of effective therapies to prevent chemotherapy-induced neuropathies could be enabled by a mechanistic understanding of axonal breakdown following exposure to neuropathy-causing agents. Here, we reveal the molecular mechanisms underlying axon degeneration induced by 2 widely used chemotherapeutic agents with distinct mechanisms of action: vincristine and bortezomib. We showed previously that genetic deletion of SARM1 blocks vincristine-induced neuropathy and demonstrate here that it also prevents axon destruction following administration of bortezomib in vitro and in vivo. Using cultured neurons, we found that vincristine and bortezomib converge on a core axon degeneration program consisting of nicotinamide mononucleotide NMNAT2, SARM1, and loss of NAD+ but engage different upstream mechanisms that closely resemble Wallerian degeneration after vincristine and apoptosis after bortezomib. We could inhibit the final common axon destruction pathway by preserving axonal NAD+ levels or expressing a candidate gene therapeutic that inhibits SARM1 in vitro. We suggest that these approaches may lead to therapies for vincristine- and bortezomib-induced neuropathies and possibly other forms of peripheral neuropathy.


Subject(s)
Armadillo Domain Proteins/drug effects , Armadillo Domain Proteins/metabolism , Axons/metabolism , Bortezomib/pharmacology , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Nerve Degeneration/metabolism , Vincristine/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Armadillo Domain Proteins/genetics , Axons/pathology , Cytoskeletal Proteins/genetics , Drug Therapy , Female , Gene Expression Regulation, Neoplastic , Genetic Therapy , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/drug therapy , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurons/metabolism , Nicotinamide Mononucleotide , Nicotinamide-Nucleotide Adenylyltransferase
14.
ACS Chem Neurosci ; 9(9): 2262-2273, 2018 09 19.
Article in English | MEDLINE | ID: mdl-29932631

ABSTRACT

Through epigenetic and other regulatory functions, the histone deacetylase (HDAC) family of enzymes has emerged as a promising therapeutic target for central nervous system and other disorders. Here we report on the synthesis and functional characterization of new HDAC inhibitors based structurally on tianeptine, a drug used primarily to treat major depressive disorder (MDD) that has a poorly understood mechanism of action. Since the chemical structure of tianeptine resembles certain HDAC inhibitors, we profiled the in vitro HDAC inhibitory activity of tianeptine and demonstrated its ability to inhibit the lysine deacetylase activity of a subset of class I HDACs. Consistent with a model of active site Zn2+ chelation by the carboxylic acid present in tianeptine, newly synthesized analogues containing either a hydroxamic acid or ortho-aminoanilide exhibited increased potency and selectivity among the HDAC family. This in vitro potency translated to improved efficacy in a panel of high-content imaging assays designed to assess HDAC target engagement and functional effects on critical pathways involved in neuroplasticity in both primary mouse neurons and, for the first time, human neurons differentiated from pluripotent stem cells. Most notably, tianeptinaline, a class I HDAC-selective analogue of tianeptine, but not tianeptine itself, increased histone acetylation, and enhanced CREB-mediated transcription and the expression of Arc (activity-regulated cytoskeleton-associated protein). Systemic in vivo administration of tianeptinaline to mice confirmed its brain penetration and was found to enhance contextual fear conditioning, a behavioral test of hippocampal-dependent memory. Tianeptinaline and its derivatives provide new pharmacological tools to dissect chromatin-mediated neuroplasticity underlying memory and other epigenetically related processes implicated in health and disease.


Subject(s)
Conditioning, Psychological/drug effects , Histone Deacetylase Inhibitors/pharmacology , Memory/drug effects , Neuronal Plasticity/drug effects , Neurons/drug effects , Animals , Cyclic AMP Response Element-Binding Protein , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/genetics , Epigenesis, Genetic , Fear , Histone Deacetylases , Humans , Mice , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Thiazepines/pharmacology , Transcription, Genetic/drug effects
15.
Neurosci Lett ; 668: 24-30, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29307599

ABSTRACT

Histone deacetylase (HDAC) inhibitors, which have an effect on cell homeostasis, cell cycle progression, and terminal differentiation, can act to promote self-renewal and enhance directed differentiation of several lineages of stem cells. However, the roles of HDAC inhibitors on neurogenic differentiation and the mechanisms of Wnt signaling following treatment with HDAC inhibitors remain unclear in stem cells. We hypothesized that HDAC inhibitors regulate downstream Wnt signaling and neurogenic differentiation of mesenchymal stem cells. Following neural induction with supplementary factors, human adipose tissue-derived mesenchymal stem cells (hADSCs) were differentiated into neurogenic cells in vitro. We examined the neurogenic differentiation induced by the HDAC inhibitors, MS-275, sodium butyrate (NaB), trichostatin A (TSA), and valproic acid (VPA), by RT-PCR and western blot analysis. Based on RT-PCR analysis, the expressions of NEUROG2 and NEFL were highly increased following HDAC inhibitor treatment compared with control medium. Most of the neuronal marker genes were expressed when neural-induced hADSCs (NI-hADSCs) were treated with the HDAC inhibitors individually. Interestingly, expression of most of the Wnt-related genes were highly increased following treatment with the HDAC inhibitors, especially with MS-275 treatment. Further, the protein level of Wnt5 was upregulated after neurogenic induction with MS-275 and VPA treatment, based on western blot analysis. Furthermore, we found that c-Jun expression was increased after treatment with the HDAC inhibitors, except with NaB. The protein levels of phosphor-JNK and phosphor-GSK-3ß were upregulated considerably. In conclusion, the HDAC inhibitors could induce neurogenic differentiation of hADSCs by activating canonical Wnt or non-canonical Wnt signaling pathways.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/drug effects , Cytoskeletal Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Mesenchymal Stem Cells/drug effects , Neurofilament Proteins/metabolism , Neurons/drug effects , Nuclear Proteins/metabolism , Wnt Proteins/drug effects , Wnt Signaling Pathway/drug effects , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Cytoskeletal Proteins/drug effects , Humans , MAP Kinase Signaling System/physiology , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Neurofilament Proteins/drug effects , Nuclear Proteins/drug effects
16.
Microb Pathog ; 114: 393-401, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29233778

ABSTRACT

The hok/sok locus has been shown to enhance the growth of bacteria in adverse growth conditions such as high temperature, low starting-culture densities and antibiotic treatment. This is in addition to their well-established plasmid-stabilization effect via post-segregational killing of plasmid-free daughter cells. It delays the onset of growth by prolonging the lag phase of bacterial culture, and increases the rate of exponential growth when growth eventually begins. This enables the cells adapt to the prevailing growth conditions and enhance their survival in stressful conditions. These effects functionally complement defective SOS response mechanism, and appear analogous to the growth effects of FtsZ in the SOS pathway. In this study, the role of FtsZ in the hok/sok-induced changes in bacterial growth and cell division was investigated. Morphologic studies of early growth-phase cultures and cells growing under temperature stress showed elongated cells typical of FtsZ inhibition/deficiency. Both ftsZ silencing and over-expression produced comparable growth effects in control cells, and altered the growth changes observed otherwise in the hok/sok+ cells. These changes were diminished in SOS-deficient strain containing mutant FtsZ. The involvement of FtsZ in the hok/sok-induced growth changes may be exploited as drug target in host bacteria, which often propagate antibiotic resistance elements.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/drug effects , Bacterial Proteins/physiology , Bacterial Toxins/antagonists & inhibitors , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/physiology , Gene Expression Regulation, Bacterial/drug effects , Phenotype , Anti-Bacterial Agents/pharmacology , Bacteria/cytology , Bacteria/growth & development , Bacterial Physiological Phenomena , Bacterial Proteins/genetics , Bacterial Toxins/analysis , Bacterial Toxins/genetics , Cell Division , Escherichia coli/cytology , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/physiology , Escherichia coli Proteins/analysis , Escherichia coli Proteins/genetics , Gene Silencing , Plasmids , RNA, Bacterial/metabolism
17.
Neurosci Lett ; 661: 23-28, 2017 Nov 20.
Article in English | MEDLINE | ID: mdl-28919534

ABSTRACT

Arc/Arg3.1 is a unique immediate early gene whose expression is highly dynamic and correlated with various forms of synaptic plasticity. Many previous reports highlight the complexity of mechanisms that regulate Arc/Arg3.1 expression in neurons. In the present study, the expression and regulation of Arc/Arg3.1 after glutamate treatment in primary cultured cortical neurons were investigated. We found that both Arc/Arg3.1 mRNA and Arc/Arg3.1 protein dynamically increased within 24h after glutamate treatment. The results of immunostaining showed that abundant amounts of Arc/Arg3.1 protein are presented in both soma and dendrites. The glutamate-induced increase in Arc/Arg3.1 protein levels was partially prevented by the NMDAR inhibitor DL-AP5, but not the AMPAR inhibitor NBQX. The results of calcium imaging showed that glutamate induced significant increases in intracellular calcium levels in a NMDAR-dependent manner. However, the intracellular calcium chelator BAPTA-AM had no effect on glutamate-induced upregulation of Arc/Arg3.1 protein, and alteration of cytosolic calcium ion homeostasis with A23187 and TG did not change Arc/Arg3.1 protein levels. In addition, the phosphorylation of ERK and CREB, two downstream factors of NMDAR signaling, markedly increased after glutamate exposure. Blocking ERK and CREB activation via selective inhibitors partially prevented the glutamate-induced elevation of Arc/Arg3.1 protein levels. Combined observations support a NMDAR-mediated and calcium-independent mechanism by which glutamate increases Arc/Arg3.1 expression in cortical neurons.


Subject(s)
Glutamic Acid/pharmacology , Neuronal Plasticity/drug effects , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Dendrites/drug effects , Dendrites/metabolism , Genes, Immediate-Early/drug effects , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , MAP Kinase Signaling System/drug effects , Neuronal Plasticity/physiology , Neurons/drug effects , Phosphorylation/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
18.
Medicine (Baltimore) ; 96(35): e7602, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28858082

ABSTRACT

The aim of the study was to investigate the effects of arsenic trioxide (As2O3) treatment on the expression of ezrin and serum alpha-fetoprotein (AFP) levels in hepatocellular carcinoma (HCC).A total of 24 patients (20 males and 4 females) with resectable HCC were treated with venous injection of As2O3 for 14 days (10 mg/d) before surgery. The ezrin expression and serum AFP levels were assessed before and after treatment, respectively.The serum AFP levels were 325.5 ng/L before treatment and 278.6 ng/L after treatment, with statistical significant difference (Z = -2.360, P < .05). The expression of ezrin was negative, weak positive, and strong positive in 11, 7, and 6 cases, respectively, before As2O3 treatment, and 17, 5, 2 cases respectively after the treatment. The difference between the 2 groups was statistically significant (χ = 5.619, P < .05). Also, the results showed that there was a significant correlation between the high serum AFP level (AFP ≥ 500 ng/L) and high expression of ezrin (χ = 8.080, P < .05).As2O3 treatment can significantly downregulate the expression of ezrin in HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenicals/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Cytoskeletal Proteins/drug effects , Liver Neoplasms/drug therapy , Oxides/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Arsenic Trioxide , Arsenicals/administration & dosage , Arsenicals/pharmacology , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/surgery , Cytoskeletal Proteins/metabolism , Female , Humans , Immunohistochemistry , Infusions, Intravenous , Liver Neoplasms/blood , Liver Neoplasms/surgery , Male , Middle Aged , Oxides/administration & dosage , Oxides/pharmacology
19.
J Microbiol Biotechnol ; 27(11): 1994-1998, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-28910864

ABSTRACT

FtsZ, a bacterial cell-division protein, is an attractive antibacterial target. In the screening for an inhibitor of Staphylococcus aureus FtsZ, madurahydroxylactone (1) and its related derivatives 2-5 were isolated from Nonomuraea sp. AN100570. Compound 1 inhibited S. aureus FtsZ with an IC50 of 53.4 µM and showed potent antibacterial activity against S. aureus and MRSA with an MIC of 1 µg/ml, whereas 2-5 were weak or inactive. Importantly, 1 induced cell elongation in the cell division phenotype assay, whereas 2-5 did not. It indicates that 1 exhibits its potent antibacterial activity via inhibition of FtsZ, and the hydroxyl group and hydroxylactone ring of 1 are critical for the activity. Thus, madurahydroxylactone is a new type of inhibitor of FtsZ.


Subject(s)
Actinobacteria/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/drug effects , Cytoskeletal Proteins/drug effects , Lactones/antagonists & inhibitors , Staphylococcus aureus/drug effects , Lactones/administration & dosage , Lactones/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Staphylococcal Infections/microbiology , Staphylococcus aureus/cytology
20.
Exp Eye Res ; 155: 91-98, 2017 02.
Article in English | MEDLINE | ID: mdl-28153738

ABSTRACT

Glaucoma is the leading cause of irreversible blindness and is usually classified as angle closure and open angle glaucoma (OAG). Primary open angle glaucoma represents the most frequent clinical presentation leading to ganglion cell death and optic nerve degeneration as a main consequence of an intraocular pressure' (IOP) increase. The mechanisms of this IOP increase in such pathology remain unclear but one protein called Myocilin could be a part of the puzzle in the trabecular meshwork (TM). Previously described to be transcriptionally regulated by glucocorticoids, the comprehension of the trabecular regulation of Myocilin' expression has only weakly progressed since 15 years. Due to the essential molecular and cellular implications of retinoids' pathway in eye development and physiology, we investigate the potential role of the retinoic acid in such regulation and expression. This study demonstrates that the global retinoids signaling machinery is present in immortalized TM cells and that Myocilin (MYOC) expression is upregulated by retinoic acid alone or combined with a glucocorticoid co-treatment. This regulation by retinoic acid acts through the MYOC promoter which contains a critical cluster of four retinoic acid responsive elements (RAREs), with the RARE-DR2 presenting the strongest effect and binding the RARα/RXRα heterodimer. All together, these results open up new perspectives for the molecular understanding glaucoma pathophysiology and provide further actionable clues on Myocilin gene regulation.


Subject(s)
Cytoskeletal Proteins/genetics , Eye Proteins/genetics , Gene Expression Regulation , Glaucoma, Open-Angle/genetics , Glycoproteins/genetics , RNA/genetics , Trabecular Meshwork/metabolism , Tretinoin/pharmacology , Blotting, Western , Cells, Cultured , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/drug effects , Eye Proteins/biosynthesis , Eye Proteins/drug effects , Glaucoma, Open-Angle/drug therapy , Glaucoma, Open-Angle/metabolism , Glycoproteins/biosynthesis , Glycoproteins/drug effects , Humans , Immunohistochemistry , Intraocular Pressure/physiology , Keratolytic Agents/pharmacology , Real-Time Polymerase Chain Reaction , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...