Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
J Cell Biochem ; 125(7): e30610, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38860517

ABSTRACT

17ß-estradiol is a hormone that plays a vital role in human physiology. It acts through estrogen receptors, specifically estrogen receptor α and estrogen receptor ß, and its action is determined by the pulsatile secretion in the bloodstream. 17ß-estradiol affects cell proliferation, and dysregulation of 17ß-estradiol:estrogen receptor α signaling contribute to the development of breast cancer. Previous research on 17ß-estradiol:estrogen receptor α signaling has primarily used two-dimensional cell cultures, which do not fully recapitulate the complexity of tumors that exist in a three-dimensional environment and do not consider the pulsatile nature of this hormone. To address these limitations, we studied 17ß-estradiol:estrogen receptor α signaling in cell proliferation using both two-dimensional and three-dimensional breast cancer cell culture models under continuous and pulsatile stimulation conditions. Results revealed that breast cancer cells grown in an alginate-based three-dimensional matrix exhibited similar responsiveness to 17ß-estradiol compared with cells grown in conventional two-dimensional culture plates. 17ß-estradiol induced the expression of proteins containing estrogen response element in the three-dimensional model. The efficacy of the antiestrogen drugs fulvestrant (ICI182,280) and 4OH-tamoxifen was also demonstrated in the three-dimensional model. These results support the use of the three-dimensional culture model for studying tumor response to drugs and provide a more realistic microenvironment for such studies. Furthermore, the study revealed that a brief 5-min exposure to 17ß-estradiol triggered a physiological response comparable with continuous hormone exposure, suggesting that the cellular response to 17ß-estradiol is more important than the continuous presence of the hormone. In conclusion, the study demonstrates that the alginate-based three-dimensional culture model is suitable for studying the effects of 17ß-estradiol and antiestrogen drugs on breast cancer cells, offering a more realistic representation of tumor-microenvironment interactions. The results also highlight the importance of considering the physiological importance of the temporal dynamics in studying 17ß-estradiol signaling and cellular responses.


Subject(s)
Cell Proliferation , Estradiol , Estrogen Receptor alpha , Signal Transduction , Humans , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Cell Proliferation/drug effects , Signal Transduction/drug effects , Female , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , MCF-7 Cells , Cell Culture Techniques, Three Dimensional/methods , Cell Culture Techniques/methods , Fulvestrant/pharmacology
2.
Int J Mol Sci ; 24(6)2023 Mar 21.
Article in English | MEDLINE | ID: mdl-36982977

ABSTRACT

A promising therapeutic strategy to delay and/or prevent the onset of neurodegenerative diseases (NDs) could be to restore neuroprotective pathways physiologically triggered by neurons against stress injury. Recently, we identified the accumulation of neuroglobin (NGB) in neuronal cells, induced by the 17ß-estradiol (E2)/estrogen receptor ß (ERß) axis, as a protective response that increases mitochondria functionality and prevents the activation of apoptosis, increasing neuron resilience against oxidative stress. Here, we would verify if resveratrol (Res), an ERß ligand, could reactivate NGB accumulation and its protective effects against oxidative stress in neuronal-derived cells (i.e., SH-SY5Y cells). Our results demonstrate that ERß/NGB is a novel pathway triggered by low Res concentrations that lead to rapid and persistent NGB accumulation in the cytosol and in mitochondria, where the protein contributes to reducing the apoptotic death induced by hydrogen peroxide (H2O2). Intriguingly, Res conjugation with gold nanoparticles increases the stilbene efficacy in enhancing neuron resilience against oxidative stress. As a whole, ERß/NGB axis regulation is a novel mechanism triggered by low concentration of Res to regulate, specifically, the neuronal cell resilience against oxidative stress reducing the triggering of the apoptotic cascade.


Subject(s)
Metal Nanoparticles , Neuroblastoma , Humans , Resveratrol/pharmacology , Globins/metabolism , Nerve Tissue Proteins/metabolism , Estrogen Receptor beta/metabolism , Hydrogen Peroxide/pharmacology , Gold/pharmacology , Neuroglobin/pharmacology , Oxidative Stress , Apoptosis , Neurons/metabolism
3.
Int J Mol Sci ; 24(3)2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36768470

ABSTRACT

Breast cancer is the first leading tumor in women in terms of incidence worldwide. Seventy percent of cases are estrogen receptor (ER) α-positive. In these malignancies, 17ß-estradiol (E2) via ERα increases the levels of neuroglobin (NGB), a compensatory protein that protects cancer cells from stress-induced apoptosis, including chemotherapeutic drug treatment. Our previous data indicate that resveratrol (RSV), a plant-derived polyphenol, prevents E2/ERα-induced NGB accumulation in this cellular context, making E2-dependent breast cancer cells more prone to apoptosis. Unfortunately, RSV is readily metabolized, thus preventing its effectiveness. Here, four different RSV analogs have been developed, and their effect on the ERα/NGB pathway has been compared with RSV conjugated with highly hydrophilic gold nanoparticles as prodrug to evaluate if RSV derivatives maintain the breast cancer cells' susceptibility to the chemotherapeutic drug paclitaxel as the original compound. Results demonstrate that RSV conjugation with gold nanoparticles increases RSV efficacy, with respect to RSV analogues, reducing NGB levels and enhancing the pro-apoptotic action of paclitaxel, even preventing the anti-apoptotic action exerted by E2 treatment on these cells. Overall, RSV conjugation with gold nanoparticles makes this complex a promising agent for medical application in breast cancer treatment.


Subject(s)
Breast Neoplasms , Metal Nanoparticles , Prodrugs , Female , Humans , Neuroglobin/pharmacology , Breast Neoplasms/metabolism , Resveratrol/pharmacology , Resveratrol/therapeutic use , Estrogen Receptor alpha/metabolism , Prodrugs/pharmacology , Prodrugs/therapeutic use , Globins/metabolism , Nerve Tissue Proteins/metabolism , Gold/pharmacology , Estradiol/pharmacology , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Cell Line, Tumor , Apoptosis , Estrogens/pharmacology
4.
Int J Mol Sci ; 23(24)2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36555447

ABSTRACT

Naringenin (Nar) is one of major citrus flavonoids predominantly found in grapefruit and orange. In vivo studies have demonstrated Nar potential as a normolipidemic agent capable to reduce circulating cholesterol in hypercholesterolemic rabbits, rats, and patients, suggesting a new role for this molecule in cardiovascular disease prevention. Although Nar cholesterol-lowering effects are known, the underlying mechanisms have not yet been elucidated. Interestingly, Nar binds to the estrogen receptors (ERs), modulating both transcriptional and membrane-initiating signals. Although estrogen and ERs are deeply involved in lipid metabolism, no data are available regarding a putative role of these nuclear receptors as mediators of the hypocholesterolemic effect exerted by Nar. Thus, the aim of this work was to study the involvement of ERs in Nar-induced modulation of cholesterol metabolism. Results obtained in HepG2 cell line demonstrate that Nar can modulate the molecular network of cholesterol homeostasis. However, these effects were only partially dependent on the activity of estrogen receptor α. As a whole, our data highlight new molecular mechanisms by which Nar influences cholesterol metabolism, opening a new scenery about dietary impact on human health.


Subject(s)
Estrogen Receptor alpha , Flavanones , Humans , Rats , Animals , Rabbits , Estrogen Receptor alpha/metabolism , Flavanones/pharmacology , Flavanones/therapeutic use , Flavonoids , Cholesterol
5.
Cells ; 10(11)2021 11 17.
Article in English | MEDLINE | ID: mdl-34831423

ABSTRACT

Retinal neurodegeneration affects an increasing number of people worldwide causing vision impairments and blindness, reducing quality of life, and generating a great economic challenge. Due to the complexity of the tissue, and the diversity of retinal neurodegenerative diseases in terms of etiology and clinical presentation, so far, there are no cures and only a few early pathological markers have been identified. Increasing efforts have been made to identify and potentiate endogenous protective mechanisms or to abolish detrimental stress responses to preserve retinal structure and function. The discovering of the intracellular monomeric globin neuroglobin (NGB), found at high concentration in the retina, has opened new possibilities for the treatment of retinal disease. Indeed, the NGB capability to reversibly bind oxygen and its neuroprotective function against several types of insults including oxidative stress, ischemia, and neurodegenerative conditions have raised the interest in the possible role of the globin as oxygen supplier in the retina and as a target for retinal neurodegeneration. Here, we provide the undercurrent knowledge on NGB distribution in retinal layers and the evidence about the connection between NGB level modulation and the functional outcome in terms of retinal neuroprotection to provide a novel therapeutic/preventive target for visual pathway degenerative disease.


Subject(s)
Molecular Targeted Therapy , Neuroglobin/antagonists & inhibitors , Retinal Degeneration/drug therapy , Animals , Humans , Models, Biological , Neuroglobin/genetics , Neuroglobin/metabolism , Neuroprotective Agents/pharmacology , Retina/drug effects , Retina/pathology
6.
Toxics ; 9(10)2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34678933

ABSTRACT

Phthalates comprise a group of synthetic chemicals present in the environment because of their wide use as plasticizers and as additives in products for personal care. Among others, diethyl phthalate (DEP) is largely used in products for infants, children, and adults, in which its exposure has been correlated with an increased risk of breast cancer. The adverse health outcomes deriving from phthalate exposure have been associated with their activity as endocrine disruptors (EDCs) of the steroid and thyroid hormone signaling by affecting developmental and reproductive health, and even carcinogenicity. However, the estrogen disruptor activities of DEP are still controversial, and the mechanism at the root of the estrogenic-disrupting action of DEP remains to be clarified. Here, we evaluated the DEP mechanism of action on the activation status of estrogen receptor α (ERα) by analyzing the receptor's phosphorylation as well as both nuclear and extra-nuclear pathways triggered by the receptor to modulate the proliferation of breast cancer cells. Although DEP does not bind to ERα, our results suggest that this phthalate ester exerts multiple parallel interactions with ERα signaling and emphasize the importance to determine an appropriate battery of in vitro methods that will include specific molecular mechanisms involved in the endocrine disruption.

7.
Mol Cell Endocrinol ; 538: 111452, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34500041

ABSTRACT

Over the last decades, a great body of evidence has defined a novel view of the cellular mechanism of action of the steroid hormone 17ß-estradiol (E2) through its estrogen receptors (i.e., ERα and ERß). It is now clear that the E2-activated ERs work both as transcription factors and extra-nuclear plasma membrane-localized receptors. The activation of a plethora of signal transduction cascades follows the E2-dependent engagement of plasma membrane-localized ERs and is required for the coordination of gene expression, which ultimately controls the occurrence of the pleiotropic effects of E2. The definition of the molecular mechanisms by which the ERs locate at the cell surface (i.e., palmitoylation and protein association) determined the quest for understanding the specificity of the extra-nuclear E2 signaling. The use of mice models lacking the plasma membrane ERα localization unveiled that the extra-nuclear E2 signaling is operational in vivo but tissue-specific. However, the underlying molecular details for such ERs signaling diversity in the perspective of the E2 physiological functions in the different cellular contexts are still not understood. Therefore, to gain insights into the tissue specificity of the extra-nuclear E2 signaling to physiological functions, here we reviewed the known ERs extra-nuclear interactors and tried to extrapolate from available databases the ERα and ERß extra-nuclear interactomes. Based on literature data, it is possible to conclude that by specifically binding to extra-nuclear localized proteins in different sub-cellular compartments, the ERs fine-tune their molecular activities. Moreover, we report that the context-dependent diversity of the ERs-mediated extra-nuclear E2 actions can be ascribed to the great flexibility of the physical structures of ERs and the spatial-temporal organization of the logistics of the cells (i.e., the endocytic compartments). Finally, we provide lists of proteins belonging to the potential ERα and ERß extra-nuclear interactomes and propose that the systematic experimental definition of the ERs extra-nuclear interactomes in different tissues represents the next step for the research in the ERs field. Such characterization will be fundamental for the identification of novel druggable targets for the innovative treatment of ERs-related diseases.


Subject(s)
Cell Membrane/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Animals , Estradiol/metabolism , Mice , Organ Specificity , Signal Transduction
8.
Cells ; 10(8)2021 08 05.
Article in English | MEDLINE | ID: mdl-34440755

ABSTRACT

The expression of the α-subtype of Estrogen Receptor (ERα) characterizes most breast cancers (more than 75%), for which endocrine therapy is the mainstay for their treatment. However, a high percentage of ERα+ breast cancers are de novo or acquired resistance to endocrine therapy, and the definition of new targets for improving therapeutic interventions and the prediction of treatment response is demanding. Our previous data identified the ERα/AKT/neuroglobin (NGB) pathway as a common pro-survival process activated in different ERα breast cancer cell lines. However, no in vivo association between the globin and the malignity of breast cancer has yet been done. Here, we evaluated the levels and localization of NGB in ERα+ breast ductal carcinoma tissue of different grades derived from pre-and post-menopausal patients. The results indicate a strong association between NGB accumulation, ERα, AKT activation, and the G3 grade, while no association with the menopausal state has been evidenced. Analyses of the data set (e.g., GOBO) strengthen the idea that NGB accumulation could be linked to tumor cell aggressiveness (high grade) and resistance to treatment. These data support the view that NGB accumulation, mainly related to ER expression and tumor grade, represents a compensatory process, which allows cancer cells to survive in an unfavorable environment.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Carcinoma, Ductal, Breast/chemistry , Estrogen Receptor alpha/analysis , Neuroglobin/analysis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/therapy , Case-Control Studies , Disease Progression , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Progression-Free Survival , Proto-Oncogene Proteins c-akt/analysis , Signal Transduction , Tumor Microenvironment
9.
Am J Physiol Cell Physiol ; 321(1): C72-C81, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34010067

ABSTRACT

Estradiol (E2) and selective estrogen receptor modulators (SERMs) have broad-ranging cellular effects that include mitochondrial respiration and reactive oxygen species (ROS) metabolism. Many of these effects have been studied using cell culture models. Recent advances have revealed the extent to which cellular metabolism is affected by the culture environment. Cell culture media with metabolite composition similar to blood plasma [e.g., Plasmax, Human Plasma-Like Medium (HPLM)] alter cellular behaviors including responses to drugs. Similar effects have been observed with respect to O2 levels in cell culture. Given these observations, we investigated whether the effects of E2 and SERMs are also influenced by media composition and O2 level during cell culture experiments. We analyzed mitochondrial network characteristics, cellular oxidative metabolism, and H2O2 production in C2C12 myoblasts growing in physiological (5%) or standard cell culture (18%)O2 and in physiological (Plasmax) or standard cell culture [Dulbecco's modified Eagle's medium (DMEM)] media. Although E2 significantly lowered H2O2 production from cells growing in 18% O2/DMEM (standard cell culture), it had no effect on cells growing in Plasmax. Moreover, culture conditions significantly altered the effects of E2 and SERMs on mitochondrial abundance and network characteristics. These results indicate that the effects of E2 and SERMs on various aspects of cell physiology strongly depends on growth conditions, which in turn emphasizes the need to consider this carefully in cell culture experiments.


Subject(s)
Culture Media/pharmacology , Estradiol/pharmacology , Mitochondria/drug effects , Myoblasts/drug effects , Oxygen/pharmacology , Reactive Oxygen Species/metabolism , Animals , Apoptosis , Cell Culture Techniques , Cell Line , Culture Media/chemistry , Fluorescence , Genes, Reporter , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism
10.
Molecules ; 26(8)2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33924212

ABSTRACT

Neuroglobin (NGB) is a myoglobin-like monomeric globin that is involved in several processes, displaying a pivotal redox-dependent protective role in neuronal and extra-neuronal cells. NGB remarkably exerts its function upon upregulation by NGB inducers, such as 17ß-estradiol (E2) and H2O2. However, the molecular bases of NGB's functions remain undefined, mainly in non-neuronal cancer cells. Human MCF-7 breast cancer cells with a knocked-out (KO) NGB gene obtained using CRISPR/Cas9 technology were analyzed using shotgun label-free quantitative proteomics in comparison with control cells. The differential proteomics experiments were also performed after treatment with E2, H2O2, and E2 + H2O2. All the runs acquired using liquid chromatography-tandem mass spectrometry were elaborated within the same MaxQuant analysis, leading to the quantification of 1872 proteins in the global proteomic dataset. Then, a differentially regulated protein dataset was obtained for each specific treatment. After the proteomic study, multiple bioinformatics analyses were performed to highlight unbalanced pathways and processes. Here, we report the proteomic and bioinformatic investigations concerning the effects on cellular processes of NGB deficiency and cell treatments. Globally, the main processes that were affected were related to the response to stress, cytoskeleton dynamics, apoptosis, and mitochondria-driven pathways.


Subject(s)
Breast Neoplasms/genetics , Neuroglobin/genetics , Oxidative Stress/genetics , Proteomics , Apoptosis/genetics , Breast Neoplasms/pathology , Computational Biology , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Knockout Techniques , Humans , MCF-7 Cells , Neoplasm Proteins/genetics
11.
Arch Biochem Biophys ; 701: 108823, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33675812

ABSTRACT

Dysfunctional mitochondria have severe consequences on cell functions including Reactive Oxygen Specie (ROS) generation, alteration of mitochondrial signaling, Ca2+ buffering, and activation of apoptotic pathway. These dysfunctions are closely linked with degenerative diseases including neurodegeneration. The discovery of neuroglobin (NGB) as an endogenous neuroprotective protein, which effects seem to depend on its mitochondrial localization, could drive new therapeutic strategies against aged-related neurodegenerative diseases. Indeed, high levels of NGB are active against several brain injuries, including neurodegeneration, hypoxia, ischemia, toxicity, and nutrient deprivation opening a new scenario in the comprehension of the relationship between neural pathologies and mitochondrial homeostasis. In this review, we provide the current understanding of the role of mitochondria in neurodegeneration and discuss structural and functional connection between NGB and mitochondria with the purpose of defining a novel mitochondrial-based neuroprotective mechanism(s).


Subject(s)
Calcium Signaling , Calcium/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Neuroglobin/metabolism , Neuroprotection , Animals , Humans , Mitochondria/pathology , Neurodegenerative Diseases/pathology
12.
Cancers (Basel) ; 12(12)2020 Dec 19.
Article in English | MEDLINE | ID: mdl-33352737

ABSTRACT

Estrogen receptor α expressing breast cancers (BC) are classically treated with endocrine therapy. Prolonged endocrine therapy often results in a metastatic disease (MBC), for which a standardized effective therapy is still lacking. Thus, new drugs are required for primary and metastatic BC treatment. Here, we report that the Food and Drug Administration (FDA)-approved drugs, ouabain and digoxin, induce ERα degradation and prevent proliferation in cells modeling primary and metastatic BC. Ouabain and digoxin activate the cellular proteasome, instigating ERα degradation, which causes the inhibition of 17ß-estradiol signaling, induces the cell cycle blockade in the G2 phase, and triggers apoptosis. Remarkably, these effects are independent of the inhibition of the Na/K pump. The antiproliferative effects of ouabain and digoxin occur also in diverse cancer models (i.e., tumor spheroids and xenografts). Additionally, gene profiling analysis reveals that these drugs downregulate the expression of genes related to endocrine therapy resistance. Therefore, ouabain and digoxin behave as 'anti-estrogen'-like drugs, and are appealing candidates for the treatment of primary and metastatic BCs.

13.
Cancers (Basel) ; 12(9)2020 Aug 29.
Article in English | MEDLINE | ID: mdl-32872414

ABSTRACT

Components of tumor microenvironment, including tumor and/or stromal cells-derived factors, exert a critical role in breast cancer (BC) progression. Here we evaluated the possible role of neuroglobin (NGB), a monomeric globin that acts as a compensatory protein against oxidative and apoptotic processes, as part of BC microenvironment. The extracellular NGB levels were evaluated by immunofluorescence of BC tissue sections and by Western blot of the culture media of BC cell lines. Moreover, reactive oxygen species (ROS) generation, cell apoptosis, and cell migration were evaluated in different BC cells and non-tumorigenic epithelial mammary cells treated with BC cells (i.e., Michigan Cancer Foundation-7, MCF-7) conditioned culture media and extracellular NGB. Results demonstrate that NGB is a component of BC microenvironment. NGB is released in tumor microenvironment by BC cells only under oxidative stress conditions where it can act as autocrine/paracrine factor able to communicate cell resilience against oxidative stress and chemotherapeutic treatment.

14.
Cancers (Basel) ; 12(1)2020 Jan 09.
Article in English | MEDLINE | ID: mdl-31936631

ABSTRACT

Although soy consumption is associated with breast cancer prevention, the low bioavailability and the extensive metabolism of soy-active components limit their clinical application. Here, the impact of daidzein (D) and its metabolites on estrogen-dependent anti-apoptotic pathway has been evaluated in breast cancer cells. In estrogen receptor α-positive breast cancer cells treated with D and its metabolites, single or in mixture, ERα activation and Neuroglobin (NGB) levels, an anti-apoptotic estrogen/ERα-inducible protein, were evaluated. Moreover, the apoptotic cascade activation, as well as the cell number after stimulation was assessed in the absence/presence of paclitaxel to determine the compound effects on cell susceptibility to a chemotherapeutic agent. Among the metabolites, only D-4'-sulfate maintains the anti-estrogenic effect of D, reducing the NGB levels and rendering breast cancer cells more prone to the paclitaxel treatment, whereas other metabolites showed estrogen mimetic effects, or even estrogen independent effects. Intriguingly, the co-stimulation of D and gut metabolites strongly reduced D effects. The results highlight the important and complex influence of metabolic transformation on isoflavones physiological effects and demonstrate the need to take biotransformation into account when assessing the potential health benefits of consumption of soy isoflavones in cancer.

15.
Antioxid Redox Signal ; 32(4): 217-227, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31686530

ABSTRACT

Aims: Nuclear factor (erythroid-derived 2)-like-2 factor (NRF-2) is a transcription factor well known to provide an advantage for cancer growth and survival regulating the cellular redox pathway. In breast cancer cells, we recently identified the monomeric heme-globin neuroglobin (NGB) as part of a new mechanism induced by the steroid hormone 17ß-estradiol (E2) against oxidative stress. While there is mounting evidence suggesting a critical role of NGB as a sensor of oxidative stress, scarce information is available about its involvement in NRF-2 pathway activation in breast cancer cells. Results: Although NGB is not involved in the rapid E2-induced NRF-2 stability, E2 loses the capacity to regulate the expression of NRF-2-dependent genes in NGB-depleted MCF-7 cells. These data strongly sustain a role of NGB as a compensatory protein in the E2-activated intracellular pathway devoted to the increase of cancer cells tolerance to reactive oxygen species (ROS) generation in stressing conditions acting as key regulator of NRF-2 pathway activity in a time-dependent manner. Innovation: In this study, we identified a new role of NGB in the cell response to oxidative stress. Conclusion: Altogether, reported results open new insights on the NGB effect in regulating intracellular pathways related to cell adaptive response to stress and, as consequence, to cell survival, beyond its direct effect as ROS scavenger, opening new prospective in cancer therapeutic intervention.


Subject(s)
Antioxidants/pharmacology , Breast Neoplasms/metabolism , Estradiol/pharmacology , NF-E2-Related Factor 2/metabolism , Neuroglobin/genetics , Breast Neoplasms/genetics , CRISPR-Cas Systems , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , MCF-7 Cells , Neuroglobin/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
16.
Oxid Med Cell Longev ; 2019: 6315034, 2019.
Article in English | MEDLINE | ID: mdl-31354909

ABSTRACT

Reactive oxygen species (ROS) result from intracellular aerobic metabolism and/or extracellular stimuli. Although endogenous antioxidant systems exquisitely balance ROS production, an excess of ROS production, commonly found in diverse human degenerative pathologies including cancer, gives rise to the oxidative stress. Increased oxidative stress in cancer is related to the sustained proliferation and metabolism of cancer cells. However, cancer cells show an intrinsic higher antioxidant capacity with respect to the normal counterpart as well as an ability to cope with oxidative stress-induced cell death by establishing mechanisms of adaptation, which define a selective advantage against the adverse oxidative stress environment. The identification of survival factors and adaptive pathways, set up by cancer cells against oxidative stress, provides multiple targets for the therapeutic intervention against cancer. Neuroglobin (NGB), a globin primarily described in neurons as an oxidative stress sensor and cytoprotective factor against redox imbalance, has been recently recognized as a novel tumor-associated protein. In this review, the involvement of NGB in the cancer cell adaptation and resistance to oxidative stress will be discussed highlighting the globin role in the regulation of both the stress-induced apoptotic pathway and antioxidant systems activated by cancer cells.


Subject(s)
Neoplasms/drug therapy , Neuroglobin/therapeutic use , Cell Death , Globins , Humans , Neuroglobin/pharmacology , Oxidative Stress , Reactive Oxygen Species
17.
J Cell Physiol ; 234(4): 3147-3157, 2019 04.
Article in English | MEDLINE | ID: mdl-30421506

ABSTRACT

Neuroglobin (NGB), an antiapoptotic protein upregulated by 17ß-estradiol (E2), is part of E2/estrogen receptor α (ERα) pathway pointed to preserve cancer cell survival in presence of microenvironmental stressors including chemotherapeutic drugs. Here, the possibility that resveratrol (Res), an anticancer plant polyphenol, could increase the susceptibility of breast cancer cells to paclitaxel (Pacl) by affecting E2/ERα/NGB pathway has been evaluated. In MCF-7 and T47D (ERα-positive), but not in MDA-MB 231 (ERα-negative) nor in SK-N-BE (ERα and ERß positive), Res decreases NGB levels interfering with E2/ERα-induced NGB upregulation and with E2-induced ERα and protein kinase B phosphorylation. Although Res treatment does not reduce cell viability by itself, this compound potentiates Pacl proapoptotic effects. Notably, the increase of NGB levels by NGB expression vector transfection prevents Pacl or Res/Pacl effects. Taken together, these findings indicate a new Res-based mechanism that acts on tumor cells impairing the E2/ERα/NGB signaling pathways and increasing cancer cell susceptibility to chemotherapeutic agent.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Neuroglobin/metabolism , Paclitaxel/pharmacology , Resveratrol/pharmacology , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Synergism , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neuroglobin/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
18.
Int J Mol Sci ; 19(9)2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30189583

ABSTRACT

The potential "health benefits" of dietary polyphenols have been ascribed to their direct antioxidant activity and their impact on the regulation of cell and tissue redox balance. However, because of the relative poor bioavailability of many of these compounds, their effects could not be easily explained by the antioxidant action, which may occur only at high circulating and tissue concentrations. Therefore, many efforts have been put forward to clarify the molecular mechanisms underlining the biological effect of polyphenols in physiological and pathological conditions. Polyphenols' bioavailability, metabolism, and their effects on enzyme, membrane, and/or nuclear receptors and intracellular transduction mechanisms may define the overall impact of these compounds on cancer risk and progression, which is still debated and not yet clarified. Polyphenols are able to bind to estrogen receptor α (ERα) and ß (ERß), and therefore induce biological effects in human cells through mimicking or inhibiting the action of endogenous estrogens, even at low concentrations. In this work, the role and effects of food-contained polyphenols in hormone-related cancers will be reviewed, mainly focusing on the different polyphenols' mechanisms of action with particular attention on their estrogen receptor-based effects, and on the consequences of such processes on tumor progression and development.


Subject(s)
Antioxidants/pharmacology , Dietary Supplements , Neoplasms/metabolism , Polyphenols/pharmacology , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Animals , Antioxidants/chemistry , Antioxidants/therapeutic use , Biological Availability , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/pathology , Polyphenols/chemistry , Polyphenols/therapeutic use , Receptors, Estrogen/chemistry , Structure-Activity Relationship
19.
J Cell Physiol ; 233(7): 5087-5103, 2018 07.
Article in English | MEDLINE | ID: mdl-29219195

ABSTRACT

Neuroglobin (NGB) is a relatively recent discovered monomeric heme-protein, which behave in neurons as a sensor of injuring stimuli including oxidative stress, hypoxia, and neurotoxicity. In addition, the anti-apoptotic activity of overexpressed NGB has been reported both in neurons and in cancer cell lines. We recently demonstrated that, NGB functions as a compensatory protein of the steroid hormone 17ß-estradiol (E2) protecting cancer cells against the apoptotic death induced by oxidative stress. However, the E2-induced signaling pathways at the root of NGB over-expression and mitochondrial re-localization in breast cancer cells is still elusive. By using a kinase screening library, here, we report that: i) There is a strong positive correlation between NGB and ERα expression and activity in breast cancer cells; ii) The E2-activated phosphatidyl-inositol 3 kinase (PI3K)/protein kinase B (AKT) and protein kinase C (PKC) pathways are necessary to modulate the NGB protein levels; iii) The E2-induced persistent activation of AKT drive NGB to mitochondria; iv) Reactive oxygen species (ROS)-inducing compounds activating rapidly and transiently AKT does not affect the NGB mitochondrial level; and v) High level of NGB into mitochondria are necessary for the pro-survival and anti-apoptotic effect of this globin in cancer cells. As a whole, these results underline the E2 triggered pathways in E2-responsive breast cancer cells that involve NGB as a compensatory protein devoted to cancer cell survival.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Neuroglobin/genetics , Oxidative Stress/genetics , Animals , Apoptosis/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estradiol/genetics , Estradiol/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mitochondria/genetics , Mitochondria/metabolism , Neuroglobin/metabolism , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase C/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
20.
PLoS One ; 12(12): e0189179, 2017.
Article in English | MEDLINE | ID: mdl-29216269

ABSTRACT

Environmental factors or adverse growth conditions that may reduce cell function or viability are considered stress. The cell ability to sense and respond to environmental stresses determine its function and survival destiny. We recently defined Neuroglobin (NGB), a heme-protein, as a compensatory protein in the 17ß-Estradiol (E2) anti-apoptotic activity and as a sensor of oxidative stress in both neurons and breast cancer cells. Here, the possibility that NGB levels could represent a pivotal regulator of integrated response of cancer cells to stress has been evaluated. Data obtained in neuroblastoma and in breast cancer cell lines evidence that nutrient deprivation significantly up-regulated NGB levels at different time points. However, the analysis of autophagy activation led to exclude any possible role of stress- or E2-induced NGB in the upstream regulation of general autophagy. However, the over-expression of Flag-NGB in ERα stable transfected HEK-293 cells completely affects nutrient deprivation-induced decrease in cell number. In addition, reported results indicate that modulation of the anti-apoptotic Bcl-2 level may play a key role in the protective NGB function against energetic stress. Overall, these data define a role of NGB as compensatory protein in the cell machinery activated in response to stress and as general stress adaptation marker of cancer cells susceptible to oxidative stress, oxygen and, as demonstrated here for the first time, even to nutrient willingness. Despite the lacking of any direct NGB role on autophagic flux activated by energetic stress, NGB upregulation appears functional in delaying stress-related cell death allowing an appropriate cell response and adaptation to the changing extracellular conditions.


Subject(s)
Breast Neoplasms/pathology , Globins/physiology , Nerve Tissue Proteins/physiology , Neuroblastoma/pathology , Neurons/pathology , Autophagy , Breast Neoplasms/metabolism , Cell Line, Tumor , Culture Media , Globins/metabolism , HEK293 Cells , Humans , Nerve Tissue Proteins/metabolism , Neuroblastoma/metabolism , Neuroglobin , Neurons/metabolism , Oxidative Stress
SELECTION OF CITATIONS
SEARCH DETAIL