Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Cell Mol Med ; 28(7): e18192, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38506079

ABSTRACT

In the last decade, extensive attention has been paid to the uremic toxin indoxyl sulphate (IS) as an inducer of cardiac fibroblast (cFib) activation and cardiac fibrosis in chronic kidney disease. At cellular level, IS engages aryl hydrocarbon receptor (AhR) and regulates many biological functions. We analysed how AhR inhibition by CH-223191 (CH) and overexpression of non-functional (dominant negative, DN) nuclear factor-erythroid-2-related factor 2 (NRF2), a transcription factor recruited by AhR, modulate the response of neonatal mouse (nm) cFib to IS. We also evaluated nm-cardiomyocytes after incubation with the conditioned medium (CM) of IS±CH-treated nm-cFib. IS induced activation, collagen synthesis, TLR4 and-downstream-MCP-1, and the genes encoding angiotensinogen, angiotensin-converting enzyme, angiotensin type 1 receptor (AT1r) and neprilysin (Nepr) in nm-cFib. CH antagonized IS-initiated nm-cFib activation, but did not affect or even magnified the other features. IS promoted NRF2 nuclear translocation and expression the NRF2 target Nqo1. Both pre-incubation with CH and transfection of DN-NRF2 resulted in loss of NRF2 nuclear localization. Moreover, DN-NRF2 overexpression led to greater TLR4 and MCP-1 levels following exposure to IS. The CM of IS-primed nm-cFib and to a larger extent the CM of IS+CH-treated nm-cFib upregulated AT1r, Nepr and TNFα and myostatin genes in nm-cardiomyocytes. Hence, IS triggers pro-inflammatory activation of nm-cFib partly via AhR, and AhR-NRF2 counteract it. Strategies other than AhR inhibition are needed to target IS detrimental actions on cardiac cells.


Subject(s)
Indican , Signal Transduction , Mice , Animals , Indican/pharmacology , Indican/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Toll-Like Receptor 4/genetics , Fibroblasts/metabolism
2.
Sci Rep ; 13(1): 597, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36631513

ABSTRACT

Classical cadherins, including vascular endothelial (VE)-cadherin, are targeted by matrix metalloproteinases (MMPs) and γ-secretase during adherens junction (AJ) disassembly, a mechanism that might have relevance for endothelial cell (EC) integrity and vascular homeostasis. Here, we show that oxidative stress triggered by H2O2 exposure induced efficient VE-cadherin proteolysis by MMPs and γ-secretase in human umbilical endothelial cells (HUVECs). The cytoplasmic domain of VE-cadherin produced by γ-secretase, VE-Cad/CTF2-a fragment that has eluded identification so far-could readily be detected after H2O2 treatment. VE-Cad/CTF2, released into the cytosol, was tightly regulated by proteasomal degradation and was sequentially produced from an ADAM10/17-generated C-terminal fragment, VE-Cad/CTF1. Interestingly, BMP9 and BMP10, two circulating ligands critically involved in vascular maintenance, significantly reduced VE-Cad/CTF2 levels during H2O2 challenge, as well as mitigated H2O2-mediated actin cytoskeleton disassembly during VE-cadherin processing. Notably, BMP9/10 pretreatments efficiently reduced apoptosis induced by H2O2, favoring endothelial cell recovery. Thus, oxidative stress is a trigger of MMP- and γ-secretase-mediated endoproteolysis of VE-cadherin and AJ disassembly from the cytoskeleton in ECs, a mechanism that is negatively controlled by the EC quiescence factors, BMP9 and BMP10.


Subject(s)
Amyloid Precursor Protein Secretases , Proteasome Endopeptidase Complex , Humans , Amyloid Precursor Protein Secretases/metabolism , Proteasome Endopeptidase Complex/metabolism , Endothelial Cells/metabolism , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Cadherins/metabolism , Oxidative Stress , Matrix Metalloproteinases/metabolism , Cells, Cultured , Adherens Junctions/metabolism , Bone Morphogenetic Proteins/metabolism
3.
Antioxidants (Basel) ; 11(8)2022 Aug 19.
Article in English | MEDLINE | ID: mdl-36009331

ABSTRACT

Alterations of redox homeostasis leads to a condition of resilience known as hormesis that is due to the activation of redox-sensitive pathways stimulating cell proliferation, growth, differentiation, and angiogenesis. Instead, supraphysiological production of reactive oxygen species (ROS) exceeds antioxidant defence and leads to oxidative distress. This condition induces damage to biomolecules and is responsible or co-responsible for the onset of several chronic pathologies. Thus, a dietary antioxidant supplementation has been proposed in order to prevent aging, cardiovascular and degenerative diseases as well as carcinogenesis. However, this approach has failed to demonstrate efficacy, often leading to harmful side effects, in particular in patients affected by cancer. In this latter case, an approach based on endogenous antioxidant depletion, leading to ROS overproduction, has shown an interesting potential for enhancing susceptibility of patients to anticancer therapies. Therefore, a deep investigation of molecular pathways involved in redox balance is crucial in order to identify new molecular targets useful for the development of more effective therapeutic approaches. The review herein provides an overview of the pathophysiological role of ROS and focuses the attention on positive and negative aspects of antioxidant modulation with the intent to find new insights for a successful clinical application.

4.
Antioxidants (Basel) ; 11(6)2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35740068

ABSTRACT

Induction of heme oxygenase 1 (HO-1) favors immune-escape in BRAFV600 melanoma cells treated with Vemurafenib/PLX4032 under standard cell culture conditions. However, the oxygen tension under standard culture conditions (~18 kPa O2) is significantly higher than the physiological oxygen levels encountered in vivo. In addition, cancer cells in vivo are often modified by hypoxia. In this study, MeOV-1 primary melanoma cells bearing the BRAFV600E mutation, were adapted to either 5 kPa O2 (physiological normoxia) or 1 kPa O2 (hypoxia) and then exposed to 10 µM PLX4032. PLX4032 abolished ERK phosphorylation, reduced Bach1 expression and increased HO-1 levels independent of pericellular O2 tension. Moreover, cell viability was significantly reduced further in cells exposed to PLX4032 plus Tin mesoporphyrin IX, a HO-1 inhibitor. Notably, our findings provide the first evidence that HO-1 inhibition in combination with PLX4032 under physiological oxygen tension and hypoxia restores and increases the expression of the NK ligands ULBP3 and B7H6 compared to cells exposed to PLX4032 alone. Interestingly, although silencing NRF2 prevented PLX4032 induction of HO-1, other NRF2 targeted genes were unaffected, highlighting a pivotal role of HO-1 in melanoma resistance and immune escape. The present findings may enhance translation and highlight the potential of the HO-1 inhibitors in the therapy of BRAFV600 melanomas.

5.
Antioxidants (Basel) ; 10(5)2021 May 17.
Article in English | MEDLINE | ID: mdl-34067625

ABSTRACT

Heme oxygenase 1 (HO-1) plays a key role in cell adaptation to stressors through the antioxidant, antiapoptotic, and anti-inflammatory properties of its metabolic products. For these reasons, in cancer cells, HO-1 can favor aggressiveness and resistance to therapies, leading to poor prognosis/outcome. Genetic polymorphisms of HO-1 promoter have been associated with an increased risk of cancer progression and a high degree of therapy failure. Moreover, evidence from cancer biopsies highlights the possible correlation between HO-1 expression, pathological features, and clinical outcome. Indeed, high levels of HO-1 in tumor specimens often correlate with reduced survival rates. Furthermore, HO-1 modulation has been proposed in order to improve the efficacy of antitumor therapies. However, contrasting evidence on the role of HO-1 in tumor biology has been reported. This review focuses on the role of HO-1 as a promising biomarker of cancer progression; understanding the correlation between HO-1 and clinical data might guide the therapeutic choice and improve the outcome of patients in terms of prognosis and life quality.

6.
J Cell Sci ; 134(3)2021 02 04.
Article in English | MEDLINE | ID: mdl-33443102

ABSTRACT

KRIT1 is a scaffolding protein that regulates multiple molecular mechanisms, including cell-cell and cell-matrix adhesion, and redox homeostasis and signaling. However, rather little is known about how KRIT1 is itself regulated. KRIT1 is found in both the cytoplasm and the nucleus, yet the upstream signaling proteins and mechanisms that regulate KRIT1 nucleocytoplasmic shuttling are not well understood. Here, we identify a key role for protein kinase C (PKC) in this process. In particular, we found that PKC activation promotes the redox-dependent cytoplasmic localization of KRIT1, whereas inhibition of PKC or treatment with the antioxidant N-acetylcysteine leads to KRIT1 nuclear accumulation. Moreover, we demonstrated that the N-terminal region of KRIT1 is crucial for the ability of PKC to regulate KRIT1 nucleocytoplasmic shuttling, and may be a target for PKC-dependent regulatory phosphorylation events. Finally, we found that silencing of PKCα, but not PKCδ, inhibits phorbol 12-myristate 13-acetate (PMA)-induced cytoplasmic enrichment of KRIT1, suggesting a major role for PKCα in regulating KRIT1 nucleocytoplasmic shuttling. Overall, our findings identify PKCα as a novel regulator of KRIT1 subcellular compartmentalization, thus shedding new light on the physiopathological functions of this protein.


Subject(s)
Active Transport, Cell Nucleus , KRIT1 Protein/metabolism , Protein Kinase C-alpha , HeLa Cells , Humans , Phosphorylation , Protein Kinase C-alpha/genetics , Tetradecanoylphorbol Acetate
7.
Front Physiol ; 11: 23, 2020.
Article in English | MEDLINE | ID: mdl-32082188

ABSTRACT

Among antioxidants in the human body, bilirubin has been recognized over the past 20 years to afford protection against different chronic conditions, including inflammation and cardiovascular disease. Moderate increases in plasma concentration and cellular bilirubin generation from metabolism of heme via heme oxygenase (HMOX) in virtually all tissues can modulate endothelial and vascular function and exert antioxidant and anti-inflammatory roles. This review aims to provide an up-to-date and critical overview of the molecular mechanisms by which bilirubin derived from plasma or from HMOX1 activation in vascular cells affects endothelial function. Understanding the molecular actions of bilirubin may critically improve the management not only of key cardiovascular diseases, but also provide insights into a broad spectrum of pathologies driven by endothelial dysfunction. In this context, therapeutic interventions aimed at mildly increasing serum bilirubin as well as bilirubin generated endogenously by endothelial HMOX1 should be considered.

8.
Int J Cancer ; 146(7): 1950-1962, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31376303

ABSTRACT

Heme oxygenase 1 (HO-1) plays a pivotal role in preventing cell damage. Indeed, through the antioxidant, antiapoptotic and anti-inflammatory properties of its metabolic products, it favors cell adaptation against different stressors. However, HO-1 induction has also been related to the gain of resistance to therapy in different types of cancers and its involvement in cancer immune-escape has been hypothesized. We have investigated the role of HO-1 expression in Vemurafenib-treated BRAFV600 melanoma cells in modulating their susceptibility to NK cell-mediated recognition. Different cell lines, isolated in house from melanoma patients, have been exposed to 1-10 µM PLX4032, which efficiently reduced ERK phosphorylation. In three lines, Vemurafenib was able to induce only a limited decrease in cell viability, while HO-1 expression was upregulated. HO-1 silencing/inhibition was able to induce a further significant reduction of Vemurafenib-treated melanoma viability. Moreover, while NK cell degranulation and killing activity were decreased upon interaction with melanoma exposed to Vemurafenib, HO-1 silencing was able to completely restore NK cell ability to degranulate and kill. Furthermore, melanoma cell treatment with Vemurafenib downregulated the expression of ligands of NKp30 and NKG2D activating receptors, and HO-1 silencing/inhibition was able to restore their expression. Our results indicate that HO-1 downregulation can both improve the efficacy of Vemurafenib on melanoma cells and favor melanoma susceptibility to NK cell-mediated recognition and killing.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Heme Oxygenase-1/genetics , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Mutation , Proto-Oncogene Proteins B-raf/genetics , Vemurafenib/pharmacology , Biomarkers , Cell Line, Tumor , Cytotoxicity, Immunologic , Gene Silencing , Heme Oxygenase-1/metabolism , Humans , Immunophenotyping , Killer Cells, Natural/immunology , Receptors, Natural Killer Cell/metabolism
9.
Int J Mol Sci ; 19(8)2018 Aug 01.
Article in English | MEDLINE | ID: mdl-30071692

ABSTRACT

Heme oxygenase 1 (HO-1) up-regulation is recognized as a pivotal mechanism of cell adaptation to stress. Under control of different transcription factors but with a prominent role played by Nrf2, HO-1 induction is crucial also in nervous system response to damage. However, several lines of evidence have highlighted that HO-1 expression is associated to neuronal damage and neurodegeneration especially in Alzheimer's and Parkinson's diseases. In this review, we summarize the current literature regarding the role of HO-1 in nervous system pointing out different molecular mechanisms possibly responsible for HO-1 up-regulation in nervous system homeostasis and neurodegeneration.


Subject(s)
Alzheimer Disease/enzymology , Gene Expression Regulation, Fungal , Heme Oxygenase-1/biosynthesis , Neurons/enzymology , Parkinson Disease/enzymology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Cell Survival , Heme Oxygenase-1/genetics , Humans , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology
10.
Front Oncol ; 8: 199, 2018.
Article in English | MEDLINE | ID: mdl-29951371

ABSTRACT

Heme oxygenase 1 (HO-1) is crucially involved in cell adaptation to oxidative stress and has been demonstrated to play an important role in cancer progression and resistance to therapies. We recently highlighted that undifferentiated neuroblastoma (NB) cells are prone to counteract oxidative stress through the induction of HO-1. Conversely, differentiated NB cells were more sensitive to oxidative stress since HO-1 was scarcely upregulated. In this work, we investigated the role played by miR-494, which has been proved to be involved in cancer biology and in the modulation of oxidative stress, in the upregulation of HO-1. We showed that NB differentiation downregulates miR-494 level. In addition, endogenous miR-494 inhibition in undifferentiated cells impairs HO-1 induction in response to exposure to 500 µM H2O2, reducing the number of viable cells. The analysis of Bach1 expression did not reveal any significant modifications in any experimental conditions tested, proving that the impairment of HO-1 induction observed in cells treated with miR-494 inhibitor and exposed to H2O2 is independent from Bach1. Our results underline the role played by miR-494 in favoring HO-1 induction and cell adaptation to oxidative stress and contribute to the discovery of new potential pharmacological targets to improve anticancer therapies.

11.
Sci Rep ; 7(1): 7568, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28790431

ABSTRACT

Neuronal adaptation to oxidative stress is crucially important in order to prevent degenerative diseases. The role played by the Nrf2/HO-1 system in favoring cell survival of neuroblastoma (NB) cells exposed to hydrogen peroxide (H2O2) has been investigated using undifferentiated or all-trans retinoic acid (ATRA) differentiated SH-SY5Y cells. While undifferentiated cells were basically resistant to the oxidative stimulus, ATRA treatment progressively decreased cell viability in response to H2O2. HO-1 silencing decreased undifferentiated cell viability when exposed to H2O2, proving the role of HO-1 in cell survival. Conversely, ATRA differentiated cells exposed to H2O2 showed a significantly lower induction of HO-1, and only the supplementation with low doses of bilirubin (0,5-1 µM) restored viability. Moreover, the nuclear level of Bach1, repressor of HO-1 transcription, strongly decreased in undifferentiated cells exposed to oxidative stress, while did not change in ATRA differentiated cells. Furthermore, Bach1 was displaced from HO-1 promoter in undifferentiated cells exposed to H2O2, enabling the binding of Nrf2. On the contrary, in ATRA differentiated cells treated with H2O2, Bach1 displacement was impaired, preventing Nrf2 binding and limiting HO-1 transcription. In conclusion, our findings highlight the central role of Bach1 in HO-1-dependent neuronal response to oxidative stress.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Heme Oxygenase-1/metabolism , Hydrogen Peroxide/toxicity , Neurons/physiology , Oxidants/toxicity , Oxidative Stress , Cell Line, Tumor , Cell Survival/drug effects , Humans , Neurons/drug effects
12.
Antioxidants (Basel) ; 6(2)2017 May 05.
Article in English | MEDLINE | ID: mdl-28475131

ABSTRACT

The upregulation of heme oxygenase-1 (HO-1) is one of the most important mechanisms of cell adaptation to stress. Indeed, the redox sensitive transcription factor Nrf2 is the pivotal regulator of HO-1 induction. Through the antioxidant, antiapoptotic, and antinflammatory properties of its metabolic products, HO-1 plays a key role in healthy cells in maintaining redox homeostasis and in preventing carcinogenesis. Nevertheless, several lines of evidence have highlighted the role of HO-1 in cancer progression and its expression correlates with tumor growth, aggressiveness, metastatic and angiogenetic potential, resistance to therapy, tumor escape, and poor prognosis, even though a tumor- and tissue-specific activity has been observed. In this review, we summarize the current literature regarding the pro-tumorigenic role of HO-1 dependent tumor progression as a promising target in anticancer strategy.

13.
Oxid Med Cell Longev ; 2016: 6235641, 2016.
Article in English | MEDLINE | ID: mdl-27418953

ABSTRACT

Reactive oxygen species (ROS) and their products are components of cell signaling pathways and play important roles in cellular physiology and pathophysiology. Under physiological conditions, cells control ROS levels by the use of scavenging systems such as superoxide dismutases, peroxiredoxins, and glutathione that balance ROS generation and elimination. Under oxidative stress conditions, excessive ROS can damage cellular proteins, lipids, and DNA, leading to cell damage that may contribute to carcinogenesis. Several studies have shown that cancer cells display an adaptive response to oxidative stress by increasing expression of antioxidant enzymes and molecules. As a double-edged sword, ROS influence signaling pathways determining beneficial or detrimental outcomes in cancer therapy. In this review, we address the role of redox homeostasis in cancer growth and therapy and examine the current literature regarding the redox regulatory systems that become upregulated in cancer and their role in promoting tumor progression and resistance to chemotherapy.


Subject(s)
Antioxidants/metabolism , Homeostasis , Neoplasms/pathology , Neoplasms/therapy , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Clinical Trials as Topic , Humans , Oxidation-Reduction , Reactive Oxygen Species/metabolism
14.
PLoS One ; 11(2): e0149276, 2016.
Article in English | MEDLINE | ID: mdl-26925780

ABSTRACT

OBJECTIVE: The uremic toxin Indoxyl-3-sulphate (IS), a ligand of Aryl hydrocarbon Receptor (AhR), raises in blood during early renal dysfunction as a consequence of tubular damage, which may be present even when eGFR is normal or only moderately reduced, and promotes cardiovascular damage and monocyte-macrophage activation. We previously found that patients with abdominal aortic aneurysms (AAAs) have higher CD14+CD16+ monocyte frequency and prevalence of moderate chronic kidney disease (CKD) than age-matched control subjects. Here we aimed to evaluate the IS levels in plasma from AAA patients and to investigate in vitro the effects of IS concentrations corresponding to mild-to-moderate CKD on monocyte polarization and macrophage differentiation. METHODS: Free IS plasma levels, monocyte subsets and laboratory parameters were evaluated on blood from AAA patients and eGFR-matched controls. THP-1 monocytes, treated with IS 1, 10, 20 µM were evaluated for CD163 expression, AhR signaling and then induced to differentiate into macrophages by PMA. Their phenotype was evaluated both at the stage of semi-differentiated and fully differentiated macrophages. AAA and control sera were similarly used to treat THP-1 monocytes and the resulting macrophage phenotype was analyzed. RESULTS: IS plasma concentration correlated positively with CD14+CD16+ monocytes and was increased in AAA patients. In THP-1 cells, IS promoted CD163 expression and transition to macrophages with hallmarks of classical (IL-6, CCL2, COX2) and alternative phenotype (IL-10, PPARγ, TGF-ß, TIMP-1), via AhR/Nrf2 activation. Analogously, AAA sera induced differentiation of macrophages with enhanced IL-6, MCP1, TGF-ß, PPARγ and TIMP-1 expression. CONCLUSION: IS skews monocyte differentiation toward low-inflammatory, profibrotic macrophages and may contribute to sustain chronic inflammation and maladaptive vascular remodeling.


Subject(s)
Cell Transdifferentiation , Indican/metabolism , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aortic Aneurysm, Abdominal/blood , Aortic Aneurysm, Abdominal/metabolism , Apoptosis , Biomarkers , Case-Control Studies , Cell Line , Cell Proliferation , Cell Transdifferentiation/genetics , Chemotaxis, Leukocyte/immunology , Gene Expression , Glomerular Filtration Rate , Humans , Immunophenotyping , Indican/blood , Indican/urine , Macrophages/immunology , Monocytes/immunology , Phenotype , Reactive Oxygen Species/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Cell Surface/metabolism
15.
Free Radic Biol Med ; 89: 91-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26391462

ABSTRACT

Hyperglycemia and diabetes are associated with endothelial cell dysfunction arising from enhanced oxidative injury, leading to the progression of diabetic vascular pathologies. The redox-sensitive transcription factor nuclear factor-E2-related factor 2 (Nrf2) is a master regulator of antioxidant genes, such as heme oxygenase-1 (HO-1), involved in cellular defenses against oxidative stress. We have investigated the pathways involved in high glucose-induced activation of HO-1 in endothelial cells and examined the molecular mechanisms underlying cytoprotection. Elevated d-glucose increased intracellular generation of reactive oxygen species (ROS), leading to nuclear translocation of Nrf2 and HO-1 expression in bovine aortic endothelial cells, with no changes in cell viability. Superoxide scavenging and inhibition of endothelial nitric oxide synthase (eNOS) abrogated upregulation of HO-1 expression by elevated glucose. Inhibition of HO-1 increased the sensitivity of endothelial cells to high glucose-mediated damage, while addition of bilirubin restored cell viability. Our findings establish that exposure of endothelial cells to high glucose leads to activation of endogenous antioxidant defense genes via the Nrf2/ARE pathway. Upregulation of HO-1 provides cytoprotection against high glucose-induced oxidative stress through the antioxidant properties of bilirubin. Modulation of the Nrf2 pathway in the early stages of diabetes may thus protect against sustained damage by hyperglycemia during progression of the disease.


Subject(s)
Aorta/cytology , Bilirubin/pharmacology , Cytoprotection , Endothelium, Vascular/cytology , Glucose/toxicity , Heme Oxygenase-1/metabolism , Animals , Antioxidants/pharmacology , Aorta/drug effects , Aorta/metabolism , Apoptosis/drug effects , Blotting, Western , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Electrophoretic Mobility Shift Assay , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , RNA, Messenger/genetics , Reactive Oxygen Species , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sweetening Agents/toxicity
16.
Neurobiol Aging ; 35(6): 1301-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24484607

ABSTRACT

The aggregation of amyloid-ß (Aß) peptides plays a crucial role in the onset and progression of Alzheimer's disease. Monomeric form of Aß, indeed, could exert a physiological role. Considering the anti-oligomerization property of all-trans retinoic acid (ATRA), the involvement of monomeric Aß1-42 in ATRA-induced neuronal differentiation has been investigated. Four-day ATRA treatment increases ß-secretase 1 (BACE1) level, Aß1-42 production, and receptor for advanced glycation end-products (RAGE) expression. RAGE is a well-recognized receptor for Aß, and the block of both RAGE and Aß1-42 with specific antibodies strongly impairs neurite formation in ATRA-treated cells. The involvement of Aß1-42 and RAGE in ATRA-induced morphologic changes has been confirmed treating undifferentiated cells with different molecular assemblies of peptide: 1 µM monomeric, but not oligomeric, Aß1-42 increases RAGE expression and favors neurite elongation. The block of RAGE completely prevents this effect. Furthermore, our data underline the involvement of the RAGE-dependent adhesion molecule amphoterin-induced gene and open reading frame-1 as downstream effector of both ATRA and Aß1-42. In conclusion, our findings identify a novel physiological role for monomeric Aß1-42 and RAGE in neuronal differentiation.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/physiology , Cell Differentiation/genetics , Neurons/cytology , Peptide Fragments/chemistry , Peptide Fragments/physiology , Receptors, Immunologic/physiology , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Antibodies/pharmacology , Aspartic Acid Endopeptidases/metabolism , Aspartic Acid Endopeptidases/physiology , Cell Differentiation/drug effects , Disease Progression , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neurites/physiology , Neurons/physiology , Open Reading Frames , Peptide Fragments/immunology , Peptide Fragments/metabolism , Polymerization/drug effects , Receptor for Advanced Glycation End Products , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Tretinoin/pharmacology , Tumor Cells, Cultured , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Biochim Biophys Acta ; 1842(4): 613-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24380881

ABSTRACT

High-risk neuroblastoma (NB) is characterized by the development of chemoresistance, and bortezomib (BTZ), a selective inhibitor of proteasome, has been proposed in order to overcome drug resistance. Considering the involvement of the nuclear factor-erythroid-derived 2-like 2 (Nrf2) and heme oxygenase-1 (HO-1) in the antioxidant and detoxifying ability of cancer cells, in this study we have investigated their role in differently aggressive NB cell lines treated with BTZ, focusing on the modulation of HO-1 to improve sensitivity to therapy. We have shown that MYCN amplified HTLA-230 cells were slightly sensitive to BTZ treatment, due to the activation of Nrf2 that led to an impressive up-regulation of HO-1. BTZ-treated HTLA-230 cells down-regulated p53 and up-regulated p21, favoring cell survival. The inhibition of HO-1 activity obtained by Zinc (II) protoprophyrin IX (ZnPPIX) was able to significantly increase the pro-apoptotic effect of BTZ in a p53- and p21-independent way. However, MYCN non-amplified SH-SY5Y cells showed a greater sensitivity to BTZ in relation to their inability to up-regulate HO-1. Therefore, we have shown that HO-1 inhibition improves the sensitivity of aggressive NB to proteasome inhibition-based therapy, suggesting that HO-1 up-regulation can be used as a marker of chemoresistance in NB. These results open up a new scenario in developing a combined therapy to overcome chemoresistance in high-risk neuroblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Drug Resistance, Neoplasm , Heme Oxygenase-1/physiology , Neuroblastoma/drug therapy , Pyrazines/pharmacology , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21/analysis , Heme Oxygenase-1/analysis , Heme Oxygenase-1/antagonists & inhibitors , Humans , N-Myc Proto-Oncogene Protein , NF-E2-Related Factor 2/physiology , Neuroblastoma/enzymology , Neuroblastoma/pathology , Nuclear Proteins/analysis , Oncogene Proteins/analysis , Risk , Up-Regulation
18.
Oxid Med Cell Longev ; 2013: 972913, 2013.
Article in English | MEDLINE | ID: mdl-23766865

ABSTRACT

Glutathione (GSH) plays an important role in a multitude of cellular processes, including cell differentiation, proliferation, and apoptosis, and disturbances in GSH homeostasis are involved in the etiology and progression of many human diseases including cancer. While GSH deficiency, or a decrease in the GSH/glutathione disulphide (GSSG) ratio, leads to an increased susceptibility to oxidative stress implicated in the progression of cancer, elevated GSH levels increase the antioxidant capacity and the resistance to oxidative stress as observed in many cancer cells. The present review highlights the role of GSH and related cytoprotective effects in the susceptibility to carcinogenesis and in the sensitivity of tumors to the cytotoxic effects of anticancer agents.


Subject(s)
Disease Progression , Drug Resistance, Neoplasm , Glutathione/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , Glutathione/biosynthesis , Humans , Neoplasms/drug therapy
19.
Exp Mol Pathol ; 94(1): 270-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22771309

ABSTRACT

Malondialdehyde (MDA), a major lipid peroxidation product, spontaneously binds to, and modifies proteins. In vivo, proteins are physiologically exposed to micromolar MDA concentrations for long periods. In order to mimic this process in vitro, protein modification is often performed by short exposure to millimolar MDA concentrations, also in order to generate antigenic structures for antibody production. However, in our study, spectrophotometric and fluorimetric characteristics, electrophoretic migration, susceptibility to trypsin digestion and reactivity to antibodies indicated substantial differences between albumin incubated with millimolar MDA concentrations for a short period of time and albumin incubated with micromolar MDA concentrations for a long period of time. Therefore, our study showed that short incubation of albumin with millimolar MDA concentrations does not mimic the consequences of albumin exposure to long incubation with micromolar MDA concentrations. This casts doubts on the real possibility that antibodies, elicited with proteins modified with millimolar MDA concentrations for a short period, could detect all MDA-modified proteins in vivo. Moreover, natural antibodies against albumin, modified with micromolar MDA concentrations, have been detected in the serum of healthy blood donors, which appears to justify the existence of these kinds of modified proteins in vivo.


Subject(s)
Antibodies/immunology , Malondialdehyde/chemistry , Proteins/metabolism , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/immunology , Blotting, Western , Electrophoresis , Enzyme-Linked Immunosorbent Assay , Lipid Peroxidation , Malondialdehyde/metabolism , Proteins/immunology , Serum Albumin, Bovine/metabolism , Spectrophotometry , Tryptases
20.
Int J Mol Med ; 29(5): 899-905, 2012 May.
Article in English | MEDLINE | ID: mdl-22344537

ABSTRACT

Diabetes-induced glutathione (GSH) decrease is usually ascribed to GSH oxidation. Here we investigate, in streptozotocin-treated rats, if impairment of GSH synthesis contributes to GSH decrease in diabetic liver, and if antioxidant treatments can provide protection. Diabetic rats were divided into 3 groups: untreated diabetic rats (UD); N-acetyl-cysteine (NAC)-treated diabetic rats; taurine (TAU)-treated diabetic rats; a group of non-streptozotocin-treated rats was used as control (CTR). All rats were sacrificed at 40 weeks of age. Diabetes induced hepatic glutathione decrease, but oxidized glutathione (GSSG) did not increase significantly. Accumulations of cysteine and cysteinyl-glycine in UD suggest respectively decreased glutathione synthesis and increased loss through the plasma membrane with subsequent degradation. Decreased expression of γ-glutamyl-cysteine synthetase in UD is consistent with repressed GSH synthesis. Moreover, diabetes caused increase of GSSG/GSH ratio and induction of heme oxygenase-1, both signs of oxidative stress. Supplementation with NAC or TAU resulted in amelioration of glutathione levels, probably depending on antioxidant activity, more efficient glutathione synthesis and decreased GSH loss and degradation. In conclusion, impaired synthesis and increased loss and degradation of GSH appear to contribute to a decrease in GSH levels in diabetic liver. NAC and TAU are able to partially protect from oxidative stress and GSH decrease, while enhancing GSH synthesis and restricting GSH loss.


Subject(s)
Acetylcysteine/therapeutic use , Antioxidants/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Glutathione/metabolism , Liver/metabolism , Taurine/therapeutic use , Animals , Diabetes Mellitus, Experimental/chemically induced , Glutathione Disulfide/metabolism , Heme Oxygenase-1/metabolism , Liver/drug effects , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar , Streptozocin
SELECTION OF CITATIONS
SEARCH DETAIL
...