Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Oncol Lett ; 28(6): 577, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39397804

ABSTRACT

Lin28A is an oncoprotein overexpressed in several cancer types such as testicular, ovarian, colon, breast and lung cancers. As a pluripotency factor that promotes tumorigenesis, Lin28A is associated with more undifferentiated and aggressive tumors phenotypes. Moreover, Lin28A is a highly stable protein that is difficult to downregulate. The compound resveratrol (RSV) has anticancer effects. The present study aimed to elucidate the mechanisms underlying the downregulation of Lin28A protein expression by RSV in the NCCIT cell line. NCCIT cells were treated with different concentrations of RSV to investigate its effects on Lin28A expression. The mRNA expression levels of Lin28A and ubiquitin-specific protease 28 (USP28) were assessed using reverse transcription-quantitative PCR. Western blot analysis was employed to evaluate the protein levels of Lin28A, USP28 and phosphorylated Lin28A. In addition, in some experiments, cells were treated with a MAPK/ERK pathway inhibitor, and other experiments involved transfecting cells with small interfering RNAs targeting USP28. The results demonstrated that RSV significantly reduced Lin28A expression by destabilizing the protein; this effect was mediated by the ability of RSV to suppress the expression of USP28, a deubiquitinase that normally protects Lin28A from ubiquitination and degradation. Additionally, RSV inhibited phosphorylation of Lin28A via the MAPK/ERK pathway; this phosphorylation event has previously been shown to enhance the stability of Lin28A by increasing its half-life. This resulted in Lin28A degradation through the proteasomal pathway in NCCIT cells. The results provide further evidence of the anticancer activity of RSV, and identified Lin28A and USP28 as promising therapeutic targets. As a stable oncoprotein, downregulating Lin28A expression is challenging. However, the present study demonstrated that RSV can overcome this hurdle by inhibiting USP28 expression and MAPK/ERK signaling to promote Lin28A degradation. Furthermore, elucidating these mechanisms provides avenues for developing targeted cancer therapies.

2.
Ann Hepatol ; 30(1): 101564, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39276983

ABSTRACT

INTRODUCTION AND OBJECTIVES: To study the effect of eukaryotic initiation factor 3B (EIF3B) on the invasion and migration of hepatocellular carcinoma (HCC) and its potential mechanism. MATERIALS AND METHODS: The clinical significance of EIF3B expression was studied with The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interaction Analysis datasets. Immunohistochemical staining and western blotting were used to examine EIF3B expression in cell lines and tissues from HCC patients. The scratch assay and transwell assay were used to measure the invasion and metastasis of different HCC cell lines in vitro. The molecular mechanism of EIF3B was determined using RNA-seq and identification of dysregulated signaling pathways. Western blotting was used to verify the alterations of EIF3B signaling functioned in the promotion of HCC progression. RESULTS: Elevated expression of EIF3B in HCC correlated significantly with aggressive clinicopathologic characteristics, including advanced tumor grade and poor prognosis. Studies with cultured cells indicated that EIF3B knockdown inhibited HCC cell invasion and metastasis by depressing the epithelial-mesenchymal transition (EMT). EIF3B also activated the TGFBI/MAPK/ERK signaling pathway by increasing the levels of pMEK and pERK. CONCLUSIONS: Our results indicate that EIF3B functions as an oncogene in HCC that accelerates cell invasion, metastasis, and the EMT by stimulation of the TGFBI/MAPK/ERK signaling pathway. EIF3B is a potential target for the treatment of HCC.

3.
Anticancer Agents Med Chem ; 22(18): 3136-3147, 2022.
Article in English | MEDLINE | ID: mdl-35676853

ABSTRACT

INTRODUCTION: Calotropis procera (Aiton) Dryand (Apocynaceae) is an herb that has been commonly used in folk medicine to treat various diseases for more than 1500 years. AIMS: Our goal was to investigate the anti-metastatic effects of phenolics extracted from C. procera (CphE) against 4T1 breast cancer cells and in BALB/c mice. METHODS: 4T1 cells were treated with CphE and quercetin (positive control) at concentrations that inhibited cell viability by 50% (IC50). Levels of reactive oxygen species (ROS), wound healing, and protein expressions were determined following standard protocols. For the in vivo pilot study, the syngeneic BALB/c mouse model was used. 4T1 cells were injected into mammary fat pads. Tumors were allowed to grow for 9 days before gavage treatment with CphE (150 mg GAE/kg/day) or PBS (controls) for one week. Excised tumors, liver, and lungs were analyzed for gene and protein expression and histology. RESULTS: In vitro results showed that CphE suppressed cell viability through apoptosis induction, via caspase-3 cleavage and total PARP reduction. CphE also scavenged ROS and suppressed Akt, mTOR, ERK1/2, CREB, and Src activation contributing to cell motility inhibition. CphE reduced IR, PTEN, TSC2, p70S6, and RPS6, protein levels, which are proteins involved in the PI3K/Akt/mTOR pathway, suggesting this pathway as CphE primary target. In vivo results showed downregulation of ERK1/2 activation by phosphorylation in tumor tissues, accompanied by angiogenesis reduction in tumor and lung tissues. A reduction of Cenpf mRNA levels in liver and lung tissues strongly suggested anti-invasive cancer activity of CphE. CONCLUSION: CphE inhibited 4T1 cell signal pathways that play a key role in cell growth and invasion. The potential for in vitro results to be translated in vivo was confirmed. A complete animal study is a guarantee to confirm the CphE anticancer and antimetastatic activity in vivo.


Subject(s)
Calotropis , Neoplasms , Animals , Apoptosis , Calotropis/genetics , Calotropis/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/metabolism , Pilot Projects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Quercetin/pharmacology , RNA, Messenger , Reactive Oxygen Species , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
4.
Clin Transl Oncol ; 24(1): 34-47, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34255268

ABSTRACT

PURPOSE: Colorectal cancer (CRC) is one most cancer type of high incidence and high mortality rate. Metastasis play an important role in survival rate and life quality of colorectal cancer patients. Nerve growth factor (NGF) has been shown to be involved in the metastasis and deterioration in many cancers, but the detail mechanisms in promoting the metastasis of colorectal cancer remain unknown. In this study, we aimed to explore the mechanism of NGF promoting colorectal cancer metastasis to provide new insights for developing NGF anti-colorectal cancer drugs. METHODS: We examined the expression of NGF in human colorectal cancer by immunohistochemical staining, and Western blot to evaluate the relationship between NGF and colorectal cancer metastasis. Using biochemical experiments including wound healing assay, transwell migration and invasion assay, RT-PCR, Western blot and ELISA to explore the relative mechanism of NGF promoting colorectal cancer cells metastasis in vivo. RESULTS: Our results found that the high expression of NGF was related with high incidence of metastasis. The binding of NGF to TrkA phosphorylated TrkA, which activated MAPK/Erk signaling pathway increasing the expression NGAL to enhance the activity of MMP2 and MMP9, promoted colorectal cancer metastasis. CONCLUSION: Our finding demonstrated that NGF increased NGAL expression to enhance MMPs activity to promoted colorectal cancer cell metastasis by TrkA-MAPK/Erk axis.


Subject(s)
Colorectal Neoplasms/pathology , Lipocalin-2/physiology , Matrix Metalloproteinases/physiology , Nerve Growth Factor/physiology , Female , Humans , Male , Middle Aged , Neoplasm Metastasis
5.
Pharmacol Res Perspect ; 9(4): e00784, 2021 08.
Article in English | MEDLINE | ID: mdl-34176244

ABSTRACT

Myocardial ischemia continues to be the first cause of morbimortality in the world; the definitive treatment is reperfusion; however, this action causes additional damage to ischemic myocardial tissue; this forces to seek therapies of cardioprotection to reduce this additional damage. There are many cardioprotective agents; within these, cannabinoids have shown to have beneficial effects, mainly cannabidiol (CBD). CBD is a non psychoactive cannabinoid. To evaluate the effect in experimental models of CBD in myocardial ischemia reperfusion in rats, twelve-week-old male rats have been used. The animals were divides in 3 groups: control(C), ischemia reperfusion (IR) and CBD pretreatment (1/day/5mg/kg /10days). Langendorff organ isolate studies were performed, and the area of infarction was assessed with triphenyl tetrazolium, in addition to molecular analysis of AT1 and AT2 receptors and Akt and Erk proteins and their phosphorylated forms related to RISK pathways. It was observed that there is an improvement with the use of CBD increasing inotropism and cardiac lusitropism, improving considerably the cardiovascular functionality. These could be related to the reduction of the area of infarction and activation of the AT2 receptor and the RISK pathway with absence of activation of the AT2 receptor (these could relate the reduction of the infarct area and the restoration of cardiovascular function with the activation of the AT2 receptor and the RISK pathway with the absence of activation of the AT2 receptor). The use of cannabinoids was shown to have beneficial effects when used as a treatment for myocardial reperfusion damage.


Subject(s)
Cannabidiol/therapeutic use , Cardiotonic Agents/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Animals , Cannabidiol/pharmacology , Cardiotonic Agents/pharmacology , Heart/physiology , Hemodynamics , In Vitro Techniques , MAP Kinase Signaling System/drug effects , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Ventricular Function, Left/drug effects
6.
J Oral Pathol Med ; 49(8): 796-802, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32492752

ABSTRACT

BACKGROUND: Brown tumors are giant cell-rich lesions that result from abnormal bone metabolism in hyperparathyroidism, one of the most common endocrine disorders worldwide. Brown tumors occasionally affect the jaws and, despite well-known clinical and microscopic features, their molecular pathogenesis remains unclear. We investigated the presence of pathogenic activating mutations in TRPV4, FGFR1, and KRAS in a cohort of brown tumors since these have recently been reported in giant-cell lesions of the jaws and non-ossifying fibromas of the bones (FGFR1 and KRAS), which are histologic mimics of brown tumors. METHODS: We target sequenced 13 brown tumors of the jaws associated with primary or secondary hyperparathyroidism. As mutations in these genes are known to activate the MAPK/ERK signaling pathway, we also assessed the immunostaining of the phosphorylated form of ERK1/2 (pERK1/2) in these lesions. RESULTS: KRAS pathogenic mutations were detected in seven cases (p.G12V n = 4, p.G12D n = 1, p.G13D n = 1, p.A146T n = 1). KRAS variants of unknown significance (VUS), p.A134T and p.E37K, were also detected. All samples showed wild-type sequences for FGFR1 and TRPV4 genes. The activation of the MAPK/ERK signaling pathway was demonstrated by pERK1/2 immunohistochemical positivity of the brown tumors´ mononuclear cells. CONCLUSION: Mutations in KRAS and activation of the MAPK/ERK signaling pathway were detected in brown tumors of hyperparathyroidism of the jaws, expanding the spectrum of giant cell lesions whose molecular pathogenesis involve RAS signaling.


Subject(s)
Hyperparathyroidism , Jaw Neoplasms , Humans , Hyperparathyroidism/genetics , Jaw , Jaw Neoplasms/genetics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics
7.
Mol Oncol ; 13(2): 290-306, 2019 02.
Article in English | MEDLINE | ID: mdl-30422399

ABSTRACT

In malignant transformation, cellular stress-response pathways are dynamically mobilized to counterbalance oncogenic activity, keeping cancer cells viable. Therapeutic disruption of this vulnerable homeostasis might change the outcome of many human cancers, particularly those for which no effective therapy is available. Here, we report the use of fibroblast growth factor 2 (FGF2) to demonstrate that further mitogenic activation disrupts cellular homeostasis and strongly sensitizes cancer cells to stress-targeted therapeutic inhibitors. We show that FGF2 enhanced replication and proteotoxic stresses in a K-Ras-driven murine cancer cell model, and combinations of FGF2 and proteasome or DNA damage response-checkpoint inhibitors triggered cell death. CRISPR/Cas9-mediated K-Ras depletion suppressed the malignant phenotype and prevented these synergic toxicities in these murine cells. Moreover, in a panel of human Ewing's sarcoma family tumor cells, sublethal concentrations of bortezomib (proteasome inhibitor) or VE-821 (ATR inhibitor) induced cell death when combined with FGF2. Sustained MAPK-ERK1/2 overactivation induced by FGF2 appears to underlie these synthetic lethalities, as late pharmacological inhibition of this pathway restored cell homeostasis and prevented these described synergies. Our results highlight how mitotic signaling pathways which are frequently overridden in malignant transformation might be exploited to disrupt the robustness of cancer cells, ultimately sensitizing them to stress-targeted therapies. This approach provides a new therapeutic rationale for human cancers, with important implications for tumors still lacking effective treatment, and for those that frequently relapse after treatment with available therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Fibroblast Growth Factor 2/pharmacology , Stress, Physiological , Animals , Bortezomib/pharmacology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Proteasome Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/metabolism
8.
Mol Oncol, v. 13, n.2, p. 290-306, dez. 2019
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-2670

ABSTRACT

In malignant transformation, cellular stress-response pathways are dynami-cally mobilized to counterbalance oncogenic activity, keeping cancer cellsviable. Therapeutic disruption of this vulnerable homeostasis might changethe outcome of many human cancers, particularly those for which no effec-tive therapy is available. Here, we report the use of fibroblast growth factor2 (FGF2) to demonstrate that further mitogenic activation disrupts cellularhomeostasis and strongly sensitizes cancer cells to stress-targeted therapeu-tic inhibitors. We show that FGF2 enhanced replication and proteotoxicstresses in a K-Ras-driven murine cancer cell model, and combinations ofFGF2 and proteasome or DNA damage response-checkpoint inhibitorstriggered cell death. CRISPR/Cas9-mediated K-Ras depletion suppressedthe malignant phenotype and prevented these synergic toxicities in thesemurine cells. Moreover, in a panel of human Ewing’s sarcoma family tumorcells, sublethal concentrations of bortezomib (proteasome inhibitor) or VE-821 (ATR inhibitor) induced cell death when combined with FGF2. Sus-tained MAPK-ERK1/2 overactivation induced by FGF2 appears to under-lie these synthetic lethalities, as late pharmacological inhibition of thispathway restored cell homeostasis and prevented these described synergies.Our results highlight how mitotic signaling pathways which are frequentlyoverridden in malignant transformation might be exploited to disrupt therobustness of cancer cells, ultimately sensitizing them to stress-targeted ther-apies. This approach provides a new therapeutic rationale for human can-cers, with important implications for tumors still lacking effectivetreatment, and for those that frequently relapse after treatment with avail-able therapies.

9.
Belo Horizonte; s.n; 2019. 77 p. ilus, tab.
Thesis in Portuguese | BBO - Dentistry | ID: biblio-1150890

ABSTRACT

As lesões de células gigantes (LCG) compõem um grupo de doenças que acometem os maxilares e que compartilham do mesmo quadro histopatológico. Existem dois subtipos de LCG, a periférica e a central. A lesão central de células gigantes (LCCG) é uma lesão intraóssea, usualmente assintomática, que afeta com maior frequência a região anterior da mandíbula de indivíduos jovens. A lesão periférica de células gigantes (LPCG) se manifesta clinicamente como um nódulo usualmente de coloração vermelha-arroxeada, que afeta com frequência a gengiva ou mucosa alveolar de indivíduos entre a quarta e quinta décadas de vida. Em alguns casos, a LPCG pode se desenvolver adjacente ao implante dentário, apresentando características clínicas e histopatológicas muito semelhantes a forma convencional não associada ao implante. Recentemente, nosso grupo de pesquisa reportou mutações recorrentes, mutuamente exclusivas e com ganho de função nos genes TRPV4, KRAS e FGFR1, com consequente ativação constitutiva da via de sinalização intracelular MAPK/ERK nas LCG dos maxilares. No entanto, o perfil molecular da LPCG associada aos implantes dentários é ainda desconhecido. Assim, o objetivo deste estudo foi avaliar o perfil de alterações moleculares na LPCG associada aos implantes dentários, investigando mutações nos genes KRAS, FGFR1 e TRPV4 previamente descritas na lesão convencional. Além disso, a ativação da via MAPK por meio da reação imuno-histoquímica para a forma fosforilada das proteínas ERK1/2 (fosfo-ERK1/2) foi também avaliada. Para isso, foram utilizadas 15 amostras de LPCG associada ao implante dentário incluídas em blocos de parafina. Mutações ativadoras no gene KRAS foram encontradas em 8 das 15 amostras analisadas, afetando os códons 12 (p.G12A/D), 14 (p.V14L), 37 (p.E37K), 127 (p.T127I) e 146 (p.A146V). Não foram detectadas mutações afetando os genes FGFR1 e TRPV4. As células mononucleares mostraram uma forte marcação nuclear e citoplasmática para a proteína fosfo-ERK1/2 na análise imuno-histoquímica, o que sugere ativação da via MAPK/ERK na LPCG associada ao implante dentário. Concluindo, este estudo mostra ativação da via MAPK/ERK na LPCG associada aos implantes. Nossos achados também demonstram que as lesões relacionadas aos implantes apresentam perfil molecular semelhante a LPCG convencional.


Giant cell lesions (GLC) are a group of jaw diseases that share the same histopathological features. The GCL of the jaws have two distinct clinical subtypes: central and peripheral. The central giant cell lesion (CGCL) is an intraosseous disease, often asymptomatic that most commonly affects young individuals in the anterior region of the jaw. Peripheral giant cell lesion (PGCL) is clinically characterized by nodule with a reddish-purple color, mainly presented in the gingiva or alveolar mucosa of female individuals between the fourth and fifth decades of life. In some cases, PGCL may develop adjacent to a dental implant. The clinical and histopathological features of these lesions are very similar to those of non-implant-associated lesions. Recently, our research group reported recurrent, mutually exclusive and activating mutations in the TRPV4, KRAS and FGFR1 genes and a consequent constitutive activation of the MAPK/ERK intracellular signaling pathway in the GCL of the jaws. However, the molecular profile of PGCL associated with dental implants has not been determined. Thus, the objective of this study was to evaluate the molecular profile of the PGCL associated with dental implants by the investigation of KRAS, FGFR1 and TRPV4 mutations previously reported in the conventional lesions. MAPK activation was also evaluated through the immunohistochemical expression of the phosphorylated form of ERK1/2 proteins (phosphoERK1/2). For this purpose, 15 samples of PGCL associated with dental implant were used. Activating mutations in the KRAS gene were found in 7 of the 15 samples analyzed, affecting codons 12 (p.G12A / D), 14 (p.V14L), 37 (p.E37K) and 146 (p.A146V). Mutations in FGFR1 and TRPV4 genes were not detected. Mononuclear cells were strong staining by phospho-ERK1/2 protein in the immunohistochemical analysis, which confirmed the activation of the MAPK/ERK pathway in the PGCL associated with dental implants. In conclusion, the present study shows MAPK/ERK pathway activation in PGCL associated with dental implants. Our findings also demonstrate that the lesions associated with dental implants present a similar molecular profile with the conventional PGCL


Subject(s)
Granuloma, Giant Cell , Jaw Diseases , Maxillary Diseases , Giant Cells , Dental Implantation , Wounds and Injuries , Pathology, Molecular
10.
Int J Mol Sci ; 19(3)2018 Mar 13.
Article in English | MEDLINE | ID: mdl-29534015

ABSTRACT

Classical Hodgkin lymphoma (cHL) cells overexpress heat-shock protein 90 (HSP90), an important intracellular signaling hub regulating cell survival, which is emerging as a promising therapeutic target. Here, we report the antitumor effect of celastrol, an anti-inflammatory compound and a recognized HSP90 inhibitor, in Hodgkin and Reed-Sternberg cell lines. Two disparate responses were recorded. In KM-H2 cells, celastrol inhibited cell proliferation, induced G0/G1 arrest, and triggered apoptosis through the activation of caspase-3/7. Conversely, L428 cells exhibited resistance to the compound. A proteomic screening identified a total of 262 differentially expressed proteins in sensitive KM-H2 cells and revealed that celastrol's toxicity involved the suppression of the MAPK/ERK (extracellular signal regulated kinase/mitogen activated protein kinase) pathway. The apoptotic effects were preceded by a decrease in RAS (proto-oncogene protein Ras), p-ERK1/2 (phospho-extracellular signal-regulated Kinase-1/2), and c-Fos (proto-oncogene protein c-Fos) protein levels, as validated by immunoblot analysis. The L428 resistant cells exhibited a marked induction of HSP27 mRNA and protein after celastrol treatment. Our results provide the first evidence that celastrol has antitumor effects in cHL cells through the suppression of the MAPK/ERK pathway. Resistance to celastrol has rarely been described, and our results suggest that in cHL it may be mediated by the upregulation of HSP27. The antitumor properties of celastrol against cHL and whether the disparate responses observed in vitro have clinical correlates deserve further research.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hodgkin Disease/metabolism , Reed-Sternberg Cells/metabolism , Triterpenes/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pentacyclic Triterpenes , Proteome , Proto-Oncogene Mas , Reed-Sternberg Cells/drug effects , ras Proteins/metabolism
11.
Skelet Muscle ; 7(1): 20, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29017538

ABSTRACT

BACKGROUND: The hepatocyte growth factor (HGF) is required for the activation of muscle progenitor cells called satellite cells (SC), plays a role in the migration of proliferating SC (myoblasts), and is present as a soluble factor during muscle regeneration, along with extracellular matrix (ECM) molecules. In this study, we aimed at determining whether HGF is able to interact with ECM proteins, particularly laminin 111 and fibronectin, and to modulate human myoblast migration. METHODS: We evaluated the expression of the HGF-receptor c-Met, laminin, and fibronectin receptors by immunoblotting, flow cytometry, or immunofluorescence and used Transwell assays to analyze myoblast migration on laminin 111 and fibronectin in the absence or presence of HGF. Zymography was used to check whether HGF could modulate the production of matrix metalloproteinases by human myoblasts, and the activation of MAPK/ERK pathways was evaluated by immunoblotting. RESULTS: We demonstrated that human myoblasts express c-Met, together with laminin and fibronectin receptors. We observed that human laminin 111 and fibronectin have a chemotactic effect on myoblast migration, and this was synergistically increased when low doses of HGF were added. We detected an increase in MMP-2 activity in myoblasts treated with HGF. Conversely, MMP-2 inhibition decreased the HGF-associated stimulation of cell migration triggered by laminin or fibronectin. HGF treatment also induced in human myoblasts activation of MAPK/ERK pathways, whose specific inhibition decreased the HGF-associated stimulus of cell migration triggered by laminin 111 or fibronectin. CONCLUSIONS: We demonstrate that HGF induces ERK phosphorylation and MMP production, thus stimulating human myoblast migration on ECM molecules. Conceptually, these data state that the mechanisms involved in the migration of human myoblasts comprise both soluble and insoluble moieties. This should be taken into account to optimize the design of therapeutic cell transplantation strategies by improving the migration of donor cells within the host tissue, a main issue regarding this approach.


Subject(s)
Cell Movement , Extracellular Matrix/metabolism , Hepatocyte Growth Factor/pharmacology , MAP Kinase Signaling System , Matrix Metalloproteinases/metabolism , Myoblasts/metabolism , Cells, Cultured , Humans , Integrin alpha5beta1/metabolism , Matrix Metalloproteinases/genetics , Myoblasts/drug effects , Myoblasts/physiology , Proto-Oncogene Proteins c-met/metabolism , Receptors, Laminin/metabolism
12.
J Cell Biochem ; 117(3): 741-50, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26335579

ABSTRACT

cAMP is a second messenger well documented to be involved in the phosphorylation of PKA, MAP kinase, and histone H3 (H3). Early, we reported that cAMP also induced H3 dephosphorylation in a variety of proliferating cell lines. Herein, it is shown that cAMP elicits a biphasic H3 dephosphorylation independent of PKA activation in cycling cells. H89, a potent inhibitor of PKA catalytic sub-unite, could not abolish this effect. Additionally, H89 induces a rapid and biphasic H3 serine 10 dephosphorylation, while a decline in the basal phosphorylation of CREB/ATF-1 is observed. Rp-cAMPS, an analog of cAMP and specific inhibitor of PKA, is unable to suppress cAMP-mediated H3 dephosphorylation, whereas Rp-cAMPS effectively blocks CREB/ATF-1 hyper-phosphorylation by cAMP and its inducers. Interestingly, cAMP exerts a rapid and profound H3 dephosphorylation at much lower concentration (50-fold lower, 0.125 mM) than the concentration required for maximal CREB/ATF-1 phosphorylation (5 mM). Much higher cAMP concentration is required to fully induce CREB/ATF-1 gain in phosphate (5 mM), which correlates with the inhibition of H3 dephosphorylation. Also, the dephosphorylation of H3 does not overlap at onset of MAP kinase phosphorylation pathways, p38 and ERK. Surprisingly, rapamycin (an mTOR inhibitor), cAMP, and its natural inducer isoproterenol, elicit identical dephosphorylation kinetics on both S6K1 ribosomal kinase (a downstream mTOR target) and H3. Finally, cAMP-induced H3 dephosphorylation is PP1/2-dependent. The results suggest that a pathway, requiring much lower cAMP concentration to that required for CREB/ATF-1 hyper-phosphorylation, is responsible for histone H3 dephosphorylation and may be linked to mTOR down regulation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/physiology , Histones/metabolism , Protein Processing, Post-Translational , TOR Serine-Threonine Kinases/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Enzyme Activation , Humans , Isoquinolines/pharmacology , MAP Kinase Signaling System , Okadaic Acid/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Sirolimus/pharmacology , Sulfonamides/pharmacology
13.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;42(11): 1044-1049, Nov. 2009. ilus
Article in English | LILACS | ID: lil-529096

ABSTRACT

Neuronal apoptosis occurs in the diabetic brain due to insulin deficiency or insulin resistance, both of which reduce the expression of stem cell factor (SCF). We investigated the possible involvement of the activation of the MAPK/ERK and/or AKT pathways in neuroprotection by SCF in diabetes. Male C57/B6 mice (20-25 g) were randomly divided into four groups of 10 animals each. The morphology of the diabetic brain in mice treated or not with insulin or SCF was evaluated by H&E staining and TUNEL. SCF, ERK1/2 and AKT were measured by Western blotting. In diabetic mice treated with insulin or SCF, there was fewer structural change and apoptosis in the cortex compared to untreated mice. The apoptosis rate of the normal group, the diabetic group receiving vehicle, the diabetic group treated with insulin, and the diabetic group treated with SCF was 0.54 ± 0.077 percent, 2.83 ± 0.156 percent, 1.86 ± 0.094 percent, and 1.78 ± 0.095 percent (mean ± SEM), respectively. SCF expression was lower in the diabetic cortex than in the normal cortex; however, insulin increased the expression of SCF in the diabetic cortex. Furthermore, expression of phosphorylated ERK1/2 and AKT was decreased in the diabetic cortex compared to the normal cortex. However, insulin or SCF could activate the phosphorylation of ERK1/2 and AKT in the diabetic cortex. The results suggest that SCF may protect the brain from apoptosis in diabetes and that the mechanism of this protection may, at least in part, involve activation of the ERK1/2 and AKT pathways. These results provide insight into the mechanisms by which SCF and insulin exert their neuroprotective effects in the diabetic brain.


Subject(s)
Animals , Male , Mice , Apoptosis/drug effects , Brain/pathology , Diabetes Mellitus, Experimental/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-akt/metabolism , Stem Cell Factor/therapeutic use , Apoptosis/physiology , Blotting, Western , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/enzymology , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Mice, Inbred BALB C , Signal Transduction , Streptozocin
SELECTION OF CITATIONS
SEARCH DETAIL