Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Mol Life Sci ; 80(12): 373, 2023 Nov 25.
Article in English | MEDLINE | ID: mdl-38007410

ABSTRACT

Mitofusin-2 (MFN2) is an outer mitochondrial membrane protein essential for mitochondrial networking in most cells. Autosomal dominant mutations in the MFN2 gene cause Charcot-Marie-Tooth type 2A disease (CMT2A), a severe and disabling sensory-motor neuropathy that impacts the entire nervous system. Here, we propose a novel therapeutic strategy tailored to correcting the root genetic defect of CMT2A. Though mutant and wild-type MFN2 mRNA are inhibited by RNA interference (RNAi), the wild-type protein is restored by overexpressing cDNA encoding functional MFN2 modified to be resistant to RNAi. We tested this strategy in CMT2A patient-specific human induced pluripotent stem cell (iPSC)-differentiated motor neurons (MNs), demonstrating the correct silencing of endogenous MFN2 and replacement with an exogenous copy of the functional wild-type gene. This approach significantly rescues the CMT2A MN phenotype in vitro, stabilizing the altered axonal mitochondrial distribution and correcting abnormal mitophagic processes. The MFN2 molecular correction was also properly confirmed in vivo in the MitoCharc1 CMT2A transgenic mouse model after cerebrospinal fluid (CSF) delivery of the constructs into newborn mice using adeno-associated virus 9 (AAV9). Altogether, our data support the feasibility of a combined RNAi and gene therapy strategy for treating the broad spectrum of human diseases associated with MFN2 mutations.


Subject(s)
Charcot-Marie-Tooth Disease , Induced Pluripotent Stem Cells , Humans , Mice , Animals , RNA Interference , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Induced Pluripotent Stem Cells/metabolism , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/therapy , Charcot-Marie-Tooth Disease/metabolism , Mutation , Hydrolases/genetics , Mice, Transgenic
2.
Mol Neurodegener ; 16(1): 71, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663413

ABSTRACT

BACKGROUND: Oxidative stress (OS) is an imbalance between oxidant and antioxidant species and, together with other numerous pathological mechanisms, leads to the degeneration and death of motor neurons (MNs) in amyotrophic lateral sclerosis (ALS). MAIN BODY: Two of the main players in the molecular and cellular response to OS are NRF2, the transcription nuclear factor erythroid 2-related factor 2, and its principal negative regulator, KEAP1, Kelch-like ECH (erythroid cell-derived protein with CNC homology)-associated protein 1. Here we first provide an overview of the structural organization, regulation, and critical role of the KEAP1-NRF2 system in counteracting OS, with a focus on its alteration in ALS. We then examine several compounds capable of promoting NRF2 activity thereby inducing cytoprotective effects, and which are currently in different stages of clinical development for many pathologies, including neurodegenerative diseases. CONCLUSIONS: Although challenges associated with some of these compounds remain, important advances have been made in the development of safer and more effective drugs that could actually represent a breakthrough for fatal degenerative diseases such as ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , NF-E2-Related Factor 2 , Oxidative Stress , Amyotrophic Lateral Sclerosis/metabolism , Antioxidants/metabolism , Antioxidants/pharmacology , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology
3.
Data Brief ; 20: 1901-1904, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30294641

ABSTRACT

The data presented here are related to the original research article entitled "Imatinib enhances the maintenance of Chronic Myeloid Leukemia (CML) stem cell potential in the absence of glucose" (Bono et al., 2018). The sensitivity to the tyrosine kinase inhibitor imatinib-mesylate (IM) of KCL22 CML cells cultured under glucose shortage have been determined by scoring cell survival/growth via trypan blue exclusion and stem cell potential via Culture Repopulation Ability (CRA) assay. Discussion of the data can be found in Bono et al. (2018).

4.
Stem Cell Res ; 28: 33-38, 2018 04.
Article in English | MEDLINE | ID: mdl-29414416

ABSTRACT

The introduction of BCR/Abl tyrosine kinase inhibitors (TKI), such as imatinib-mesylate (IM), has revolutioned the treatment of chronic myeloid leukemia (CML). However, although extremely effective in inducing CML remission, IM is unable to eliminate leukemia stem cells (LSC). This is largely due to the suppression of BCR/Abl protein, driven by the reduction of energy supply due to oxygen or glucose shortage, in stem cell niches of bone marrow. Here, we investigated whether, in K562 and KCL22 CML cell cultures, glucose shortage induces refractoriness of stem cell potential to IM. In the absence of glucose, IM, while maintaining its detrimental effect on CML cell bulk, actually enhanced colony formation ability and stem cell potential. This was paralleled by an increased expression of the Nanog and Sox-2 stem cell markers. These evidences stress further the importance of developing strategies alternative to TKI capable to target LSC of CML.


Subject(s)
Glucose/deficiency , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Fusion Proteins, bcr-abl/metabolism , Humans , K562 Cells , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Tumor Stem Cell Assay
5.
Stem Cells Int ; 2017: 4979474, 2017.
Article in English | MEDLINE | ID: mdl-29118813

ABSTRACT

Previous studies based on low oxygen concentrations in the incubation atmosphere revealed that metabolic factors govern the maintenance of normal hematopoietic or leukemic stem cells (HSC and LSC). The physiological oxygen concentration in tissues ranges between 0.1 and 5.0%. Stem cell niches (SCN) are placed in tissue areas at the lower end of this range ("hypoxic" SCN), to which stem cells are metabolically adapted and where they are selectively hosted. The data reported here indicated that driver oncogenic proteins of several leukemias are suppressed following cell incubation at oxygen concentration compatible with SCN physiology. This suppression is likely to represent a key positive regulator of LSC survival and maintenance (self-renewal) within the SCN. On the other hand, LSC committed to differentiation, unable to stand suppression because of addiction to oncogenic signalling, would be unfit to home in SCN. The loss of oncogene addiction in SCN-adapted LSC has a consequence of crucial practical relevance: the refractoriness to inhibitors of the biological activity of oncogenic protein due to the lack of their molecular target. Thus, LSC hosted in SCN are suited to sustain the long-term maintenance of therapy-resistant minimal residual disease.

6.
Biochem Biophys Res Commun ; 490(3): 977-983, 2017 08 26.
Article in English | MEDLINE | ID: mdl-28666875

ABSTRACT

Neoangiogenesis is the main pathogenic event involved in a variety of retinal diseases. It has been recently demonstrated that inhibiting the urokinase-type plasminogen activator receptor (uPAR) results in reduced angiogenesis in a mouse model of oxygen-induced retinopathy (OIR), establishing uPAR as a therapeutic target in proliferative retinopathies. Here, we evaluated in cultured human retinal endothelial cells (HRECs) and in OIR mice the potential of a specific antisense oligodeoxyribonucleotide (ASO) in blocking the synthesis of uPAR and in providing antiangiogenic effects. uPAR expression in HRECs was inhibited by lipofection with the phosphorotioated 5'-CGGCGGGTGACCCATGTG-3' ASO-uPAR, complementary to the initial translation site of uPAR mRNA. Inhibition of uPAR expression via ASO-uPAR was evaluated in HRECs by analyzing VEGF-induced tube formation and migration. In addition, the well-established and reproducible murine OIR model was used to induce retinal neovascularization in vivo. OIR mice were injected intraperitoneally with ASO-uPAR and retinopathy was evaluated considering the extent of the avascular area in the central retina and neovascular tuft formation. The ASO-uPAR specifically decreased uPAR mRNA and protein levels in HRECs and mitigated VEGF-induced tube formation and cell migration. Noteworthy, in OIR mice ASO-uPAR administration reduced both the avascular area and the formation of neovascular tufts. In conclusion, although the extrapolation of these experimental findings to the clinic is not straightforward, ASO-uPAR may be considered a potential therapeutic tool for treatment of proliferative retinal diseases.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Oligodeoxyribonucleotides, Antisense/therapeutic use , Receptors, Urokinase Plasminogen Activator/genetics , Retina/pathology , Retinal Neovascularization/genetics , Retinal Neovascularization/therapy , Angiogenesis Inhibitors/genetics , Animals , Cell Line , Cell Movement/drug effects , Disease Models, Animal , Genetic Therapy , Humans , Mice , Oligodeoxyribonucleotides, Antisense/genetics , RNA, Messenger/genetics , Receptors, Urokinase Plasminogen Activator/analysis , Receptors, Urokinase Plasminogen Activator/metabolism , Retina/cytology , Retina/metabolism , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Vascular Endothelial Growth Factor A/metabolism
7.
Oncotarget ; 7(51): 84810-84825, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27852045

ABSTRACT

BCR/Abl protein drives the onset and progression of Chronic Myeloid Leukemia (CML). We previously showed that BCR/Abl protein is suppressed in low oxygen, where viable cells retain stem cell potential. This study addressed the regulation of BCR/Abl protein expression under oxygen or glucose shortage, characteristic of the in vivo environment where cells resistant to tyrosine kinase inhibitors (TKi) persist. We investigated, at transcriptional, translational and post-translational level, the mechanisms involved in BCR/Abl suppression in K562 and KCL22 CML cells. BCR/abl mRNA steady-state analysis and ChIP-qPCR on BCR promoter revealed that BCR/abl transcriptional activity is reduced in K562 cells under oxygen shortage. The SUnSET assay showed an overall reduction of protein synthesis under oxygen/glucose shortage in both cell lines. However, only low oxygen decreased polysome-associated BCR/abl mRNA significantly in KCL22 cells, suggesting a decreased BCR/Abl translation. The proteasome inhibitor MG132 or the pan-caspase inhibitor z-VAD-fmk extended BCR/Abl expression under oxygen/glucose shortage in K562 cells. Glucose shortage induced autophagy-dependent BCR/Abl protein degradation in KCL22 cells. Overall, our results showed that energy restriction induces different cell-specific BCR/Abl protein suppression patterns, which represent a converging route to TKi-resistance of CML cells. Thus, the interference with BCR/Abl expression in environment-adapted CML cells may become a useful implement to current therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Glucose/metabolism , Hypoxia/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/therapeutic use , Carcinogenesis , Drug Resistance, Neoplasm , Energy Metabolism , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Oncogene Proteins v-abl/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins c-bcr/genetics , Tumor Microenvironment
8.
Cell Cycle ; 13(20): 3169-75, 2014.
Article in English | MEDLINE | ID: mdl-25485495

ABSTRACT

This Perspective addresses the interactions of cancer stem cells (CSC) with environment which result in the modulation of CSC metabolism, and thereby of CSC phenotype and resistance to therapy. We considered first as a model disease chronic myeloid leukemia (CML), which is triggered by a well-identified oncogenetic protein (BCR/Abl) and brilliantly treated with tyrosine kinase inhibitors (TKi). However, TKi are extremely effective in inducing remission of disease, but unable, in most cases, to prevent relapse. We demonstrated that the interference with cell metabolism (oxygen/glucose shortage) enriches cells exhibiting the leukemia stem cell (LSC) phenotype and, at the same time, suppresses BCR/Abl protein expression. These LSC are therefore refractory to the TKi Imatinib-mesylate, pointing to cell metabolism as an important factor controlling the onset of TKi-resistant minimal residual disease (MRD) of CML and the related relapse. Studies of solid neoplasias brought another player into the control of MRD, low tissue pH, which often parallels cancer growth and progression. Thus, a 3-party scenario emerged for the regulation of CSC/LSC maintenance, MRD induction and disease relapse: the "hypoxic" versus the "ischemic" vs. the "acidic" environment. As these environments are unlikely constrained within rigid borders, we named this model the "metabolically-modulated stem cell niche."


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Neoplastic Stem Cells/cytology , Stem Cell Niche/physiology , Drug Resistance, Neoplasm/physiology , Fusion Proteins, bcr-abl/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...