Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
Cardiovasc Drugs Ther ; 35(1): 103-111, 2021 02.
Article En | MEDLINE | ID: mdl-33044585

PURPOSE: Vein graft failure (VGF) is an important limitation for coronary artery bypass graft (CABG) surgery. Inhibition of the excessive proliferation and migration of venous smooth muscle cells (SMCs) is an effective strategy to alleviate VGF during the CABG perioperative period. In the present study, we aimed to explore the role and potential mechanism of all-trans retinoic acid (ATRA) on preventing vein grafts stenosis. METHODS: The autogenous vein grafts model was established in the right jugular artery of rabbits. Immunohistochemistry staining and western blot assays were used to detected the protein expression, while real-time PCR assay was applied for mRNAs expression detection. The interaction between proteins was identified by co-immunoprecipitation assay. The Cell Counting Kit-8 and wound-healing assays were used to investigate the role of ATRA on human umbilical vein smooth muscle cells (HUVSMCs) function. Cell cycle progression was identified by flow cytometry assay. RESULTS: Vein graft stenosis and SMCs hyperproliferation were confirmed in vein grafts by histological and Ki-67 immunohistochemistry assays. Treatment of ATRA (10 mg/kg/day) significantly mitigated the stenosis extent of vein grafts, demonstrated by the decreased thickness of intima-media, and decreased Ki-67 expression. ATRA could repress the PDGF-bb-induced excessive proliferation and migration of HUVSMCs, which was mediated by Rb-E2F dependent cell cycle inhibition. Meanwhile, ATRA could reduce the interaction between KLF5 and RARα, thereby inhibiting the function of cis-elements of KLF5. KLF5-induced inducible nitric oxide synthase (iNOS) expression activation could be significantly inhibited by ATRA. CONCLUSIONS: These results suggested that ATRA treatment may represent an effective prevention and therapy avenue for VGF.


Constriction, Pathologic/drug therapy , Kruppel-Like Transcription Factors/drug effects , Myocytes, Smooth Muscle/drug effects , Tretinoin/pharmacology , Animals , Cell Culture Techniques , Coronary Artery Bypass/adverse effects , Humans , Ki-67 Antigen/immunology , Male , Rabbits
2.
Pharmacol Res ; 158: 104864, 2020 08.
Article En | MEDLINE | ID: mdl-32416217

Acute myeloid leukemia (AML) is a malignant disease originating from hematopoietic stem cells (HSC). Chemotherapy and/or HSC transplantation is unsatisfactory due to serious side effects, multidrug resistance, and high relapse rate. Thus, alternative strategies are urgently needed to develop more effective therapies. Liriope muscari baily saponins C (DT-13) is a novel compound isolated from Liriope muscari (Decne.) Baily, and exhibited a potent cytotoxicity against several solid tumors. However, the anti-AML activity of DT-13 and the potential mechanisms are still unknown. This study is the first to demonstrate that DT-13 had preferential cytotoxicity against AML cells, and remarkably inhibited proliferation and colony forming ability. Moreover, DT-13 induced the death receptor pathway-dependent apoptosis of HL-60 and Kasumi-1 cells by up-regulating Fas, FasL, DR5 and TRAIL as well as promoted the cleavage of caspase 8, caspase 3 and PARP. Meanwhile, DT-13 induced the differentiation with morphological change related to myeloid differentiation, elevated NBT and α-NAE positive cell rates, differentiation markers CD11b and CD14 as well as level of transcription factors C/EBPα and C/EBPß. RNA-sequencing analysis revealed that KLF2 may be one of the potential targets regulated by DT-13. Further studies indicated that KLF2 played a critical role in DT-13-induced apoptosis and differentiation. Moreover, activation of AMPK-FOXO was proved to be the upstream of KLF2 pathway that contributed to the induction of apoptosis and differentiation by DT-13. Additionally, restoration of KLF2 by DT-13 was highly correlated with the AMPK-related histone acetylation mechanisms. Finally, DT-13 exhibited an obvious anti-AML effect in NOD/SCID mice with the engraftment of HL-60 cells. Our study suggests that DT-13 may serve as a novel agent for AML by AMPL-KLF2-mediated apoptosis and differentiation.


AMP-Activated Protein Kinases/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Kruppel-Like Transcription Factors/drug effects , Leukemia, Myeloid, Acute/pathology , Saponins/pharmacology , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Humans , Liriope Plant/chemistry , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Stem Cell Assay
3.
Acta Neuropathol Commun ; 8(1): 41, 2020 04 03.
Article En | MEDLINE | ID: mdl-32245394

Meningioma represents the most common primary brain tumor in adults. Recently several non-NF2 mutations in meningioma have been identified and correlated with certain pathological subtypes, locations and clinical observations. Alterations of cellular pathways due to these mutations, however, have largely remained elusive. Here we report that the Krueppel like factor 4 (KLF4)-K409Q mutation in skull base meningiomas triggers a distinct tumor phenotype. Transcriptomic analysis of 17 meningioma samples revealed that KLF4K409Q mutated tumors harbor an upregulation of hypoxia dependent pathways. Detailed in vitro investigation further showed that the KLF4K409Q mutation induces HIF-1α through the reduction of prolyl hydroxylase activity and causes an upregulation of downstream HIF-1α targets. Finally, we demonstrate that KLF4K409Q mutated tumors are susceptible to mTOR inhibition by Temsirolimus. Taken together, our data link the KLF4K409Q mediated upregulation of HIF pathways to the clinical and biological characteristics of these skull base meningiomas possibly opening new therapeutic avenues for this distinct meningioma subtype.


Gene Expression Regulation, Neoplastic/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kruppel-Like Transcription Factors/genetics , Meningeal Neoplasms/genetics , Meningioma/genetics , Tumor Hypoxia/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/drug effects , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Meningeal Neoplasms/metabolism , Meningioma/metabolism , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Prolyl Hydroxylases , Protein Kinase Inhibitors/pharmacology , RNA-Seq , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Skull Base Neoplasms , Up-Regulation
4.
Theranostics ; 10(4): 1555-1571, 2020.
Article En | MEDLINE | ID: mdl-32042322

As a hallmark of metabolic reprogramming, aerobic glycolysis contributes to tumorigenesis and aggressiveness. However, the mechanisms and therapeutic strategies regulating aerobic glycolysis in neuroblastoma (NB), one of leading causes of cancer-related death in childhood, still remain elusive. Methods: Transcriptional regulators and their downstream glycolytic genes were identified by a comprehensive screening of publicly available datasets. Dual-luciferase, chromatin immunoprecipitation, real-time quantitative RT-PCR, western blot, gene over-expression or silencing, co-immunoprecipitation, mass spectrometry, peptide pull-down assay, sucrose gradient sedimentation, seahorse extracellular flux, MTT colorimetric, soft agar, matrigel invasion, and nude mice assays were undertaken to explore the biological effects and underlying mechanisms of transcriptional regulators in NB cells. Survival analysis was performed by using log-rank test and Cox regression assay. Results: Transcription factor myeloid zinc finger 1 (MZF1) was identified as an independent prognostic factor (hazard ratio=2.330, 95% confidence interval=1.021 to 3.317), and facilitated glycolysis process through increasing expression of hexokinase 2 (HK2) and phosphoglycerate kinase 1 (PGK1). Meanwhile, a 21-amino acid peptide encoded by upstream open reading frame of MZF1, termed as MZF1-uPEP, bound to zinc finger domain of Yin Yang 1 (YY1), resulting in repressed transactivation of YY1 and decreased transcription of MZF1 and downstream genes HK2 and PGK1. Administration of a cell-penetrating MZF1-uPEP or lentivirus over-expressing MZF1-uPEP inhibited the aerobic glycolysis, tumorigenesis and aggressiveness of NB cells. In clinical NB cases, low expression of MZF1-uPEP or high expression of MZF1, YY1, HK2, or PGK1 was associated with poor survival of patients. Conclusions: These results indicate that therapeutic targeting of YY1/MZF1 axis by MZF1-uPEP inhibits aerobic glycolysis and NB progression.


Molecular Targeted Therapy/methods , Neuroblastoma/drug therapy , Warburg Effect, Oncologic/drug effects , Animals , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Proliferation/genetics , Child , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Hexokinase/metabolism , Humans , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Nude , Neuroblastoma/metabolism , Neuroblastoma/pathology , Promoter Regions, Genetic , Survival Analysis , Transcription Factors/drug effects , Transcription Factors/metabolism , YY1 Transcription Factor/drug effects , YY1 Transcription Factor/metabolism
5.
Thyroid ; 29(10): 1518-1520, 2019 10.
Article En | MEDLINE | ID: mdl-31432759

A 13-year-old female with a novel THRB gene mutation (c.1033G>T, p.G345C) presented with 3- to 6-fold higher serum iodothyronine levels and more severe clinical manifestation than 2 other family members carrying the same mutation. The leukocytes of the proband expressed both wild-type and mutant THRB mRNAs, excluding the possibility of a partial deletion of the allele not carrying the mutation. The proband's fibroblasts showed reduced responsiveness to triiodothyronine compared with those of another affected family member. The more severe clinical and biochemical phenotype suggest a modifier-mediated worsening of the resistance to thyroid hormone.


RNA, Messenger/metabolism , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Resistance Syndrome/metabolism , Thyrotropin/metabolism , Thyroxine/metabolism , Triiodothyronine, Reverse/metabolism , Triiodothyronine/metabolism , Adolescent , Family , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , In Vitro Techniques , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/genetics , Mutation , Primary Cell Culture , Severity of Illness Index , Thyroid Hormone Resistance Syndrome/genetics , Triiodothyronine/pharmacology
6.
Clin Res Hepatol Gastroenterol ; 43(3): 292-300, 2019 06.
Article En | MEDLINE | ID: mdl-30528319

Myeloid cell leukemia-1 (Mcl-1) is highly expressed in tumor tissues and cells of hepatocellular carcinoma (HCC), yet the role of Mcl-1 in cancer stem-like cells (CSLCs) remains largely unclear. Herein, we showed that knockdown of Mcl-1 significantly inhibited HCC cells to form spheres under ultra-low attachment condition in serum-free medium, and also attenuated clone formation. Inhibition of Mcl-1 by specific inhibitors S63845 or A-1210477 hindered secondary sphere formation, triggered apoptosis signaling and reduced the level of stem cell transcription factor Nanog, Sox2 and KLF4 in HCC spheroids cells. This study suggests that Mcl-1 is an essential factor for the survival and self-renewal of HCC CSLCs.


Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Self Renewal/drug effects , Liver Neoplasms/pathology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Indoles/pharmacology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/metabolism , Liver Neoplasms/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nanog Homeobox Protein/drug effects , Nanog Homeobox Protein/metabolism , Pyrimidines/pharmacology , SOXB1 Transcription Factors/drug effects , SOXB1 Transcription Factors/metabolism , Sulfonamides/pharmacology , Thiophenes/pharmacology
7.
Yonsei Med J ; 59(3): 416-424, 2018 May.
Article En | MEDLINE | ID: mdl-29611404

PURPOSE: Parkinson's disease (PD) is a common age-dependent neurodegenerative disease. MiR-212 has been demonstrated to exert protective effects in several neurological disorders. The present study aimed to investigate the role and underlying molecular mechanism of miR-212 in PD. MATERIALS AND METHODS: 1-methyl-4-phenylpyridinium (MPP+)-induced SH-SY5Y cells were applied as a PD model in vitro. RT-qPCR was used to measure the expression of miR-212 and Kruppel-like factor 4 (KLF4) mRNA. Western blot analysis was performed to detect the protein levels of KLF4, Notch1 and Jagged1. Cell viability and apoptosis were determined by the Cell Counting Kit-8 and flow cytometry, respectively. Quantitative analysis of caspase-3 activity, lactate dehydrogenase (LDH), reactive oxygen species (ROS), superoxide dismutase (SOD), tumor necrosis factor-α (TNF-α), and interleukin-1 beta (IL-1ß) was conducted with corresponding ELISA kits. Dual-luciferase reporter assay was employed to evaluate the relationship between miR-212 and KLF4. RESULTS: MiR-212 was downregulated in MPP⁺-induced SH-SY5Y cells. Also, miR-212 alleviated MPP⁺-induced SH-SY5Y cell damage, embodied by increased cell viability, decreased caspase-3 activity, LDH release, ROS production, TNF-α, and IL-1ß expression, as well as elevated SOD levels. KLF4 was a direct target of miR-212, and miR-212 repressed KLF4 expression in a post-transcriptional manner. Moreover, miR-212-mediated protection effects were abated following KLF4 expression restoration in MPP⁺-induced SH-SY5Y cells, represented as lowered cell viability and enhanced apoptotic rate. Furthermore, Notch signaling was involved in the regulation of miR-212/KLF4 axis in MPP⁺-induced SH-SY5Y cells. CONCLUSION: miR-212 might attenuate MPP⁺-induced neuronal damage by regulating KLF4/Notch signaling pathway in SH-SY5Y cells, a promising target for PD therapy.


1-Methyl-4-phenylpyridinium/toxicity , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/genetics , Parkinson Disease/genetics , Protective Agents/pharmacology , Signal Transduction/drug effects , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Interleukin-1beta , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/drug effects , MicroRNAs/metabolism , Neurons/drug effects , Neurons/metabolism , Parkinson Disease/diagnosis , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Mol Pain ; 13: 1744806917727917, 2017.
Article En | MEDLINE | ID: mdl-28814144

Background: Nociception is maintained by genome-wide regulation of transcription in the dorsal root ganglia­spinal cord network. Hence, transcription factors constitute a promising class of targets for breakthrough pharmacological interventions to treat chronic pain. DNA decoys are oligonucleotides and specific inhibitors of transcription factor activities. A methodological series of in vivo­in vitro screening cycles was performed with decoy/transcription factor couples to identify targets capable of producing a robust and long-lasting inhibition of established chronic pain. Decoys were injected intrathecally and their efficacy was tested in the spared nerve injury and chronic constriction injury models of chronic pain in rats using repetitive von Frey testing. Results: Results demonstrated that a one-time administration of decoys binding to the Kruppel-like transcription factors (KLFs) 6, 9, and 15 produces a significant and weeks­month long reduction in mechanical hypersensitivity compared to controls. In the spared nerve injury model, decoy efficacy was correlated to its capacity to bind KLF15 and KLF9 at a specific ratio, while in the chronic constriction injury model, efficacy was correlated to the combined binding capacity to KLF6 and KLF9. AYX2, an 18-bp DNA decoy binding KLF6, KLF9, and KLF15, was optimized for clinical development, and it demonstrated significant efficacy in these models. Conclusions: These data highlight KLF6, KLF9, and KLF15 as transcription factors required for the maintenance of chronic pain and illustrate the potential therapeutic benefits of AYX2 for the treatment of chronic pain.


Chronic Pain/drug therapy , Kruppel-Like Factor 6/drug effects , Kruppel-Like Transcription Factors/drug effects , Animals , Chronic Pain/metabolism , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Male , Oligonucleotides/metabolism , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/metabolism
9.
Inflammation ; 40(5): 1589-1598, 2017 Oct.
Article En | MEDLINE | ID: mdl-28578476

KLF2 plays a protective role in antiinflammation and endothelial function, and can be regulated by promoter methylation alteration. Lipopolysaccharide (LPS) is a mediator of inflammatory responses, which causes epigenetic change of certain genes in host cells. We thus aimed to determine whether LPS could control the KLF2 expression by inducing methylation in promoter region. DNA methylation of 16 CpG sites within KLF2 promoter region was detected by bisulfite sequencing PCR. Results showed that methylation at 12 CpG sites were significantly increased in HUVECs after exposure to LPS among the total 16 sites, and the average level was increased by 57%. The KLF2 expressions assessed by reverse transcription quantitative real-time PCR and Western blot were significantly downregulated compared that without LPS simulation. Moreover, both messenger RNA and protein levels of KLF2 in HUVEC co-treated with LPS and DNA methyltransferase (DNMT) 1 small interfering RNA were dramatically higher than that treated with LPS only. Similar result was obtained when the cells were incubated in combination with LPS and 5-aza-2'-deoxycytidine (AZA), suggesting that the reduction of KLF2 expression induced by LPS can be reversed by DNMT1 inhibition. Finally, the presence of AZA changed the expression of genes that depends on KLF2 in LPS-stimulated HUVECs, which downregulated the E-selectin and VCAM and increased the eNOS and thrombomodulin expression. Our data demonstrated that LPS exposure resulted in hypermethylation in KLF2 promoter in HUVECs, which subsequently led to downregulation of the KLF2 expression. The study suggested that epigenetic alteration is involved in LPS-induced inflammatory response and provided a new insight into atherogenesis.


DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/drug effects , Lipopolysaccharides/pharmacology , Cells, Cultured , Down-Regulation/drug effects , Epigenesis, Genetic , Human Umbilical Vein Endothelial Cells , Humans , Inflammation
10.
Biol Psychiatry ; 82(1): 49-61, 2017 07 01.
Article En | MEDLINE | ID: mdl-27837918

BACKGROUND: Variation in the gene encoding zinc finger binding protein 804A (ZNF804A) is associated with schizophrenia and bipolar disorder. Evidence suggests that ZNF804A is a regulator of gene transcription and is present in nuclear and extranuclear compartments. However, a detailed examination of ZNF804A distribution and its neuronal functions has yet to be performed. METHODS: The localization of ZNF804A protein was examined in neurons derived from human neural progenitor cells, human induced pluripotent stem cells, or in primary rat cortical neurons. In addition, small interfering RNA-mediated knockdown of ZNF804A was conducted to determine its role in neurite formation, maintenance of dendritic spine morphology, and responses to activity-dependent stimulations. RESULTS: Endogenous ZNF804A protein localized to somatodendritic compartments and colocalized with the putative synaptic markers in young neurons derived from human neural progenitor cells and human induced pluripotent stem cells. In mature rat neurons, Zfp804A, the homolog of ZNF804A, was present in a subset of dendritic spines and colocalized with synaptic proteins in specific nanodomains, as determined by super-resolution microscopy. Interestingly, knockdown of ZNF804A attenuated neurite outgrowth in young neurons, an effect potentially mediated by reduced neuroligin-4 expression. Furthermore, knockdown of ZNF804A in mature neurons resulted in the loss of dendritic spine density and impaired responses to activity-dependent stimulation. CONCLUSIONS: These data reveal a novel subcellular distribution for ZNF804A within somatodendritic compartments and a nanoscopic organization at excitatory synapses. Moreover, our results suggest that ZNF804A plays an active role in neurite formation, maintenance of dendritic spines, and activity-dependent structural plasticity.


Dendritic Spines/physiology , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/physiology , Neurites/physiology , Synapses/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cells, Cultured , Dendritic Spines/ultrastructure , Humans , Kruppel-Like Transcription Factors/drug effects , Neurites/ultrastructure , Neurons/metabolism , Neurons/physiology , Neurons/ultrastructure , Psychotic Disorders/genetics , RNA, Small Interfering/pharmacology , Rats , Synapses/ultrastructure
11.
Arch Oral Biol ; 74: 69-74, 2017 Feb.
Article En | MEDLINE | ID: mdl-27886571

OBJECTIVE: Cancer stem cells contribute to tumor recurrence, and a hypoxic environment is critical for maintaining cancer stem cells. Apigenin is a natural product with anticancer activity. However, the effect of apigenin on cancer stem cells remains unclear. Our aim was to investigate the effect of apigenin on cancer stem cell marker expression in head and neck squamous cell carcinoma cells under hypoxia. DESIGN: We used three head and neck squamous cell carcinoma cell lines; HN-8, HN-30, and HSC-3. The mRNA expression of cancer stem cell markers was determined by semiquantitative RT-PCR and Real-time PCR. The cytotoxic effect of apigenin was determined by MTT colorimetric assay. Flow cytometry was used to reveal the number of cells expressing cancer stem cell surface markers. RESULTS: HN-30 cells, a cancer cell line from the pharynx, showed the greatest response to hypoxia by increasing their expression of CD44, CD105, NANOG, OCT-4, REX-1, and VEGF. Apigenin significantly decreased HN-30 cell viability in dose- and time-dependent manners. In addition, 40µM apigenin significantly down-regulated the mRNA expression of CD44, NANOG, and CD105. Consistent with these results, the hypoxia-induced increase in CD44+ cells, CD105+ cells, and STRO-1+ cells was significantly abolished by apigenin. CONCLUSION: Apigenin suppresses cancer stem cell marker expression and the number of cells expressing cell surface markers under hypoxia.


Apigenin/pharmacology , Biomarkers/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Hypoxia/genetics , Head and Neck Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation , Endoglin/drug effects , Endoglin/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , Hyaluronan Receptors/drug effects , Hyaluronan Receptors/metabolism , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/metabolism , Nanog Homeobox Protein/drug effects , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/drug effects , Octamer Transcription Factor-3/metabolism , Pharynx , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism
12.
J Am Heart Assoc ; 5(9)2016 09 16.
Article En | MEDLINE | ID: mdl-27638782

BACKGROUND: Hydrogen sulfide (H2S) is a gasotransmitter that regulates multiple cardiovascular functions. Krüppel-like factor 5 (KLF5) exerts diverse functions in the cardiovascular system. Whether and how H2S regulates KLF5 in myocardial hypertrophy is unknown. METHODS AND RESULTS: In our study, hypertrophic myocardial samples in the clinic were collected and underwent histological and molecular biological analysis. Spontaneously hypertensive rats and neonatal rat cardiomyocytes were studied for functional and signaling responses to GYY4137, an H2S-releasing compound. Expression of cystathionine γ-lyase, a principal enzyme for H2S generation in heart, decreased in human hypertrophic myocardium, whereas KLF5 expression increased. After GYY4137 administration for 4 weeks, myocardial hypertrophy was inhibited in spontaneously hypertensive rats, as demonstrated by improvement in cardiac structural parameters, heart mass, size of cardiac myocytes, and expression of atrial natriuretic peptide. H2S diminished expression of KLF5 in myocardium of spontaneously hypertensive rats and in hypertrophic cardiomyocytes. H2S also inhibits platelet-derived growth factor A promoter activity, decreased recruitment of KLF5 to the platelet-derived growth factor A promoter, and reduced atrial natriuretic peptide expression in angiotensin II-stimulated cardiomyocytes, and these effects are suppressed by KLF5 knockdown. KLF5 promoter activity and KLF5 expression was also reversed by H2S. H2S increased the S-sulfhydration on specificity protein 1 in cardiomyocytes. Moreover, H2S decreased KLF5 promoter activity; reduced KLF5 mRNA expression; attenuated specificity protein 1 binding activity with KLF5 promoter; and inhibited hypertrophy after specificity protein 1 mutated at Cys659, Cys689, and Cys692 but not Cys664 overexpression. CONCLUSIONS: These findings suggest that H2S regulates KLF5 transcription activity via specificity protein 1 S-sulfhydration at Cys664 to prevent myocardial hypertrophy.


Cardiomegaly/genetics , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , RNA, Messenger/metabolism , Angiotensin II/blood , Animals , Animals, Newborn , Atrial Natriuretic Factor/drug effects , Atrial Natriuretic Factor/metabolism , Cardiomegaly/metabolism , Case-Control Studies , Gene Expression Regulation/drug effects , Humans , Kruppel-Like Transcription Factors/drug effects , Male , Morpholines/pharmacology , Myocytes, Cardiac/drug effects , Organothiophosphorus Compounds/pharmacology , Platelet-Derived Growth Factor/drug effects , Platelet-Derived Growth Factor/metabolism , RNA, Messenger/drug effects , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Sp1 Transcription Factor/metabolism , Transcriptional Activation/drug effects
13.
Am J Physiol Heart Circ Physiol ; 311(4): H904-H912, 2016 10 01.
Article En | MEDLINE | ID: mdl-27521420

Homeostatic control of vascular smooth muscle cell (VSMC) differentiation is critical for contractile activity and regulation of blood flow. Recently, we reported that precontracted blood vessels are relaxed and the phenotype of VSMC is regulated from a synthetic to contractile state by glucose-6-phosphate dehydrogenase (G6PD) inhibition. In the current study, we investigated whether the increase in the expression of VSMC contractile proteins by inhibition and knockdown of G6PD is mediated through a protein kinase G (PKG)-dependent pathway and whether it regulates blood pressure. We found that the expression of VSMC-restricted contractile proteins, myocardin (MYOCD), and miR-1 and miR-143 are increased by G6PD inhibition or knockdown. Importantly, RNA-sequence analysis of aortic tissue from G6PD-deficient mice revealed uniform increases in VSMC-restricted genes, particularly those regulated by the MYOCD-serum response factor (SRF) switch. Conversely, expression of Krüppel-like factor 4 (KLF4) is decreased by G6PD inhibition. Interestingly, the G6PD inhibition-induced expression of miR-1 and contractile proteins was blocked by Rp-ß-phenyl-1,N2-etheno-8-bromo-guanosine-3',5'-cyclic monophosphorothioate, a PKG inhibitor. On the other hand, MYOCD and miR-143 levels are increased by G6PD inhibition through a PKG-independent manner. Furthermore, blood pressure was lower in the G6PD-deficient compared with wild-type mice. Therefore, our results suggest that the expression of VSMC contractile proteins induced by G6PD inhibition occurs via PKG1α-dependent and -independent pathways.


Aorta/metabolism , Contractile Proteins/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Animals , Aorta/drug effects , Blotting, Western , Cattle , Chromatography, Liquid , Contractile Proteins/drug effects , Contractile Proteins/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/metabolism , Gene Knockdown Techniques , Glucosephosphate Dehydrogenase/genetics , Immunoprecipitation , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , MicroRNAs/drug effects , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polymerase Chain Reaction , Rats , Serum Response Factor/drug effects , Serum Response Factor/genetics , Serum Response Factor/metabolism , Tandem Mass Spectrometry , Trans-Activators/drug effects , Trans-Activators/genetics , Trans-Activators/metabolism
14.
J Pak Med Assoc ; 66(3): 285-91, 2016 Mar.
Article En | MEDLINE | ID: mdl-26968278

OBJECTIVE: To investigate the effects of collagen and growth factors on in vitro proliferation of human spermatogonial stem cells obtained from patients with non-obstructive azoospermia. METHODS: The experimental cross-sectional study was conducted from February 2013 to April 2015 after obtaining approval from the ethics committee of Ahvaz Jundishapur University of Medical Sciences, Iran. Testicular sperm extractions of non-obstructive azoospermic patients were obtained from the Clinical Urology and Embryology, In Vitro Fertilization Department of Imam Khomeini Hospital. Spermatogonial stem cells and Sertoli cells, obtained from human testis biopsies by a two-step enzymatic digestion method, were purified using fluorescence- activated cell-sorting and daturastramonium-lectin, and were cultured separately. To investigate a more direct influential factor on colony formation, one control and two experimental groups were formed. Group 1 acted as the control in which spermatogonial stem cells were co-cultured with Sertoli cells alone. In group 2 they were co-cultured with Sertoli cells and growth factors such as leukaemia inhibitory factor, epidermal growth factor and glial cell-derived neurotrophic factor, and in group 3 with Sertoli cells along with growth factors in the presence of collagen-coated dishes. Number and diameter of the colonies were evaluated after 7 weeks. RESULTS: Specimens obtained related to 21 patients. Number and diameter of the colonies in group 3 (18±2.6 and 276.6±45.5) were significantly more than both groups 1 (3.5±1 and D1:81.6±12) and group 2(11±2.2 and 165.2±32.5) (p<0.05 each). Also, the number and diameter of colony in group 2 were significantly better than the control group (p<0.05).Expression profile of the VASA, promyelocytic leukaemia zinc-finger (PLZF), Octamer-binding transcription factor 4 (OCT4) and integrin a6 (INTGa6) were detected in all groups. Based on cytochemical findings, OCT4 was expressed in the colonies of all three groups. CONCLUSIONS: According to positive effects of collagen and growth factors on the colonisation of spermatogonial stem cells, it seems that using the cells may lead to better colonisation of this type of stem cells.


Adult Germline Stem Cells/drug effects , Cell Proliferation/drug effects , Collagen/pharmacology , Adult Germline Stem Cells/cytology , Azoospermia , Cell Culture Techniques , Coculture Techniques , Cross-Sectional Studies , DEAD-box RNA Helicases/drug effects , DEAD-box RNA Helicases/metabolism , Epidermal Growth Factor/pharmacology , Flow Cytometry , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , Integrin alpha6/drug effects , Integrin alpha6/metabolism , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/metabolism , Leukemia Inhibitory Factor/pharmacology , Male , Octamer Transcription Factor-3/drug effects , Octamer Transcription Factor-3/metabolism , Promyelocytic Leukemia Zinc Finger Protein , Sertoli Cells
15.
Addict Biol ; 21(1): 98-110, 2016 Jan.
Article En | MEDLINE | ID: mdl-25262913

Mitragynine is the major psychoactive alkaloid of the plant kratom/ketum. Kratom is widely used in Southeast Asia as a recreational drug, and increasingly appears as a pure compound or a component of 'herbal high' preparations in the Western world. While mitragynine/kratom may have analgesic, muscle relaxant and anti-inflammatory effects, its addictive properties and effects on cognitive performance are unknown. We isolated mitragynine from the plant and performed a thorough investigation of its behavioural effects in rats and mice. Here we describe an addictive profile and cognitive impairments of acute and chronic mitragynine administration, which closely resembles that of morphine. Acute mitragynine has complex effects on locomotor activity. Repeated administration induces locomotor sensitization, anxiolysis and conditioned place preference, enhances expression of dopamine transporter- and dopamine receptor-regulating factor mRNA in the mesencephalon. While there was no increase in spontaneous locomotor activity during withdrawal, animals showed hypersensitivity towards small challenging doses for up to 14 days. Severe somatic withdrawal signs developed after 12 hours, and increased level of anxiety became evident after 24 hours of withdrawal. Acute mitragynine independently impaired passive avoidance learning, memory consolidation and retrieval, possibly mediated by a disruption of cortical oscillatory activity, including the suppression of low-frequency rhythms (delta and theta) in the electrocorticogram. Chronic mitragynine administration led to impaired passive avoidance and object recognition learning. Altogether, these findings provide evidence for an addiction potential with cognitive impairments for mitragynine, which suggest its classification as a harmful drug.


Behavior, Animal/drug effects , RNA, Messenger/drug effects , Secologanin Tryptamine Alkaloids/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Avoidance Learning/drug effects , Delta Rhythm/drug effects , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/genetics , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/genetics , Locomotion/drug effects , Memory Consolidation/drug effects , Mice , RNA, Messenger/metabolism , Rats , Substance-Related Disorders , Theta Rhythm/drug effects
16.
Endocrinology ; 157(2): 774-87, 2016 Feb.
Article En | MEDLINE | ID: mdl-26606422

Severe thyroid hormone (TH) deficiency during critical phases of brain development results in irreversible neurological and cognitive impairments. The mechanisms accounting for this are likely multifactorial, and are not fully understood. Here we pursue the possibility that one important element is that TH affects basal and activity-dependent neurotrophin expression in brain regions important for neural processing. Graded exposure to propylthiouracil (PTU) during development produced dose-dependent reductions in mRNA expression of nerve growth factor (Ngf) in whole hippocampus of neonates. These changes in basal expression persisted to adulthood despite the return to euthyroid conditions in blood. In contrast to small PTU-induced reductions in basal expression of several genes, developmental PTU treatment dramatically reduced the activity-dependent expression of neurotrophins and related genes (Bdnft, Bdnfiv, Arc, and Klf9) in adulthood and was accompanied by deficits in hippocampal-based learning. These data demonstrate that mild TH insufficiency during development not only reduces expression of important neurotrophins that persists into adulthood but also severely restricts the activity-dependent induction of these genes. Considering the importance of these neurotrophins for sculpting the structural and functional synaptic architecture in the developing and the mature brain, it is likely that TH-mediated deficits in these plasticity mechanisms contribute to the cognitive deficiencies that accompany developmental TH compromise.


Behavior, Animal/physiology , Congenital Hypothyroidism/embryology , Hippocampus/embryology , Neuronal Plasticity/physiology , Prenatal Exposure Delayed Effects/metabolism , Animals , Antithyroid Agents/toxicity , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Congenital Hypothyroidism/chemically induced , Congenital Hypothyroidism/metabolism , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Early Growth Response Protein 1/drug effects , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Fear , Female , Hippocampus/metabolism , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Nerve Growth Factor/drug effects , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/drug effects , Neurotrophin 3/drug effects , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Propylthiouracil/toxicity , Rats , Rats, Long-Evans , Severity of Illness Index , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Thyroxine/deficiency , Thyroxine/drug effects , Thyroxine/metabolism , Triiodothyronine/deficiency , Triiodothyronine/drug effects
17.
Asian J Androl ; 17(6): 985-90, 2015.
Article En | MEDLINE | ID: mdl-26262775

Although paternal ethanol (EtOH) abuse has been shown to affect the growth and behavior of offspring, the exact molecular and mechanistic basis remains largely unclear. Methylation alterations in imprinted genes may be related to well-documented teratogenic effects of ethanol. Here we show that chronic paternal ethanol exposure increases the susceptibility to abnormal behavior in offspring through male game epigenetic alteration. In our study, different doses of ethanol (0, 1.1, 3.3 g kg-1 ) were administered intra-gastrically to male mice and decreased sperm motility was found in the highest ethanol-exposed group compared with the controls. Data also showed a dose-dependent increase in deaf mice of the paternally ethanol-exposed groups. The methylation of H19, Peg3, Ndn and Snrpn was assessed in paternal spermatozoa and in the cerebral cortices of deaf mice. EtOH affected methylation of Peg3 (CpG 3, 7 and 9) in paternal spermatozoa and in the cerebral cortices of deaf mice, but the level of mRNA expression did not change, suggesting that other gene regulation may be involved in these processes. Overall, chronic paternal ethanol exposure could alter the methylation of imprinted genes in sire spermatozoa that could also be passed on to offspring, giving rise to developmental disorders. Our results provide possible epigenetic evidence for a paternal ethanol exposure contribution to Fetal Alcohol Syndrome (FAS).


Anti-Infective Agents, Local/pharmacology , Cerebral Cortex/drug effects , DNA Methylation/drug effects , Ethanol/pharmacology , Genomic Imprinting/drug effects , Hearing/drug effects , Paternal Exposure , RNA, Messenger/drug effects , Spermatozoa/drug effects , Animals , Cerebral Cortex/metabolism , Deafness/genetics , Fetal Alcohol Spectrum Disorders/genetics , Gene Expression/drug effects , Hearing/genetics , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/genetics , Male , Mice , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , RNA, Long Noncoding/drug effects , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism , Spermatozoa/metabolism , snRNP Core Proteins/drug effects , snRNP Core Proteins/genetics
18.
Reproduction ; 149(4): 329-38, 2015 Apr.
Article En | MEDLINE | ID: mdl-25737569

Prospermatogonia transition to type A spermatogonia, which provide the source for the spermatogonial stem cell (SSC) pool. A percentage of these type A spermatogonia then differentiate to enter meiosis as spermatocytes by ∼P10. It is currently unclear as to when these distinct populations are initially formed in the neonatal testis, and when the expression of markers both characteristic of and required for the adult undifferentiated and differentiating states is established. In this study, we compared expression of known spermatogonial cell fate markers during normal development and in response to the differentiation signal provided by retinoic acid (RA). We found that some markers for the undifferentiated state (ZBTB16/PLZF and CDH1) were expressed in nearly all spermatogonia from P1 through P7. In contrast, differentiation markers (STRA8 and KIT) appeared in a subset of spermatogonia at P4, coincident with the onset of RA signaling. GFRA1, which was present in nearly all prospermatogonia at P1, was only retained in STRA8/KIT- spermatogonia. From P4 through P10, there was a great deal of heterogeneity in the male germ cell population in terms of expression of markers, as markers characteristic of the undifferentiated (except GFRA1) and differentiating states were co-expressed through this interval. After P10, these fate markers diverged to mark distinct populations of undifferentiated and differentiating spermatogonia, and this pattern was maintained in juvenile (P18) and adult (P>60) testes. Taken together, these results reveal that the spermatogonia population is heterogeneous during the first wave of spermatogenesis, and indicate that neonatal spermatogonia may not serve as an ideal substitute for studying the function of adult spermatogonia.


Biomarkers/metabolism , Cell Differentiation/physiology , Spermatogenesis/physiology , Spermatogonia/cytology , Testis/cytology , Animals , Animals, Newborn , Antineoplastic Agents/pharmacology , Cdh1 Proteins/drug effects , Cell Differentiation/drug effects , Immunoenzyme Techniques , Kruppel-Like Transcription Factors/drug effects , Male , Mice , Mice, Inbred C57BL , Promyelocytic Leukemia Zinc Finger Protein , Proto-Oncogene Proteins c-kit/drug effects , Spermatogenesis/drug effects , Spermatogonia/drug effects , Spermatogonia/metabolism , Testis/drug effects , Testis/metabolism , Tretinoin/pharmacology
19.
J Gerontol A Biol Sci Med Sci ; 69(3): 245-52, 2014 Mar.
Article En | MEDLINE | ID: mdl-23781099

Sonic hedgehog (Shh) is a morphogen regulating muscle development during embryogenesis. We have shown that the Shh pathway is postnatally recapitulated after injury and during regeneration of the adult skeletal muscle and regulates angiogenesis and myogenesis after muscle injury. Here, we demonstrate that in 18-month-old mice, there is a significant impairment of the upregulation of the Shh pathway that physiologically occurs in the young skeletal muscle after injury. Such impairment is even more pronounced in 24-month-old mice. In old animals, intramuscular therapy with a plasmid encoding the human Shh gene increases the regenerative capacities of the injured muscle, in terms of Myf5-positive cells, regenerating myofibers, and fibrosis. At the molecular level, Shh treatment increases the upregulation of the prototypical growth factors, insulin-like growth factor-1 and vascular endothelial growth factor. These data demonstrate that Shh increases regeneration after injury in the muscle of 24-month-old mice and suggest that the manipulation of the Shh pathway may be useful for the treatment of muscular diseases associated with aging.


Aging/physiology , Hedgehog Proteins/therapeutic use , Muscle, Skeletal/injuries , Regeneration/drug effects , Animals , Cardiotoxins/toxicity , Disease Models, Animal , Fibrosis , Genetic Therapy/methods , Humans , Insulin-Like Growth Factor I/drug effects , Intercellular Signaling Peptides and Proteins/analysis , Kruppel-Like Transcription Factors/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Muscle Development/drug effects , Muscle Development/physiology , Muscle, Skeletal/drug effects , Myofibrils/drug effects , Myogenic Regulatory Factor 5/analysis , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Plasmids/genetics , Signal Transduction/drug effects , Up-Regulation , Vascular Endothelial Growth Factor A/drug effects , Zinc Finger Protein GLI1
20.
Exp Dermatol ; 21(11): 847-52, 2012 Nov.
Article En | MEDLINE | ID: mdl-23163650

Basal cell carcinoma of the skin (BCC) is caused by constitutive activation of the Sonic hedgehog (Shh) pathway, mainly through mutations either in the Shh receptor Patched (PTCH) or in its co-receptor Smoothened (Smo). Inhibitors of this pathway that are currently in clinical trials inhibit Smo. However, mutations in Smo can result in resistance to these inhibitors. To target most BCCs and avoid acquired resistance because of Smo mutations, inhibiting the Shh-pathway downstream of Smo is critical. Attractive downstream targets would be at the level of Gli proteins, the transcriptional activators of this pathway in BCCs. Previously it has been shown that Gli1 and Gli2, when phosphorylated by protein kinase A (PKA), are targeted for proteosomal degradation. Here we show that PKA activation via the cAMP agonist forskolin is sufficient to completely abolish oncogenic Smo activity in vitro. In an inducible BCC mouse model due to a Smo mutation that confers resistance to current Smo inhibitors, topical forskolin treatment significantly reduced Gli1 mRNA levels and resulted in strongly suppressed BCC tumor growth. Our data show that forskolin inhibits the growth of even those BCCs that are resistant to Smo inhibitors and provide a proof-of-principle framework for the development of topically applied human skin-permeable novel pharmacologic inhibitors of oncogenic Shh-signaling through PKA activation.


Carcinoma, Basal Cell/prevention & control , Cyclic AMP-Dependent Protein Kinases/physiology , Hedgehog Proteins/antagonists & inhibitors , Signal Transduction/physiology , Skin Neoplasms/prevention & control , Animals , Carcinoma, Basal Cell/pathology , Carcinoma, Basal Cell/physiopathology , Cell Proliferation/drug effects , Colforsin/pharmacology , Cyclic AMP/agonists , Disease Models, Animal , Female , Hedgehog Proteins/physiology , In Vitro Techniques , Kruppel-Like Transcription Factors/drug effects , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Mutant Strains , Mutation/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Signal Transduction/drug effects , Skin Neoplasms/pathology , Skin Neoplasms/physiopathology , Smoothened Receptor , Zinc Finger Protein GLI1
...