Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Nat Rev Mol Cell Biol ; 24(12): 857-875, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37612414

ABSTRACT

The Ser/Thr kinase mechanistic target of rapamycin (mTOR) is a central regulator of cellular metabolism. As part of mTOR complex 1 (mTORC1), mTOR integrates signals such as the levels of nutrients, growth factors, energy sources and oxygen, and triggers responses that either boost anabolism or suppress catabolism. mTORC1 signalling has wide-ranging consequences for the growth and homeostasis of key tissues and organs, and its dysregulated activity promotes cancer, type 2 diabetes, neurodegeneration and other age-related disorders. How mTORC1 integrates numerous upstream cues and translates them into specific downstream responses is an outstanding question with major implications for our understanding of physiology and disease mechanisms. In this Review, we discuss recent structural and functional insights into the molecular architecture of mTORC1 and its lysosomal partners, which have greatly increased our mechanistic understanding of nutrient-dependent mTORC1 regulation. We also discuss the emerging involvement of aberrant nutrient-mTORC1 signalling in multiple diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Multiprotein Complexes , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Nutrients
3.
Arch Toxicol ; 97(7): 2035-2049, 2023 07.
Article in English | MEDLINE | ID: mdl-37258688

ABSTRACT

To transfer toxicological findings from model systems, e.g. animals, to humans, standardized safety factors are applied to account for intra-species and inter-species variabilities. An alternative approach would be to measure and model the actual compound-specific uncertainties. This biological concept assumes that all observed toxicities depend not only on the exposure situation (environment = E), but also on the genetic (G) background of the model (G × E). As a quantitative discipline, toxicology needs to move beyond merely qualitative G × E concepts. Research programs are required that determine the major biological variabilities affecting toxicity and categorize their relative weights and contributions. In a complementary approach, detailed case studies need to explore the role of genetic backgrounds in the adverse effects of defined chemicals. In addition, current understanding of the selection and propagation of adverse outcome pathways (AOP) in different biological environments is very limited. To improve understanding, a particular focus is required on modulatory and counter-regulatory steps. For quantitative approaches to address uncertainties, the concept of "genetic" influence needs a more precise definition. What is usually meant by this term in the context of G × E are the protein functions encoded by the genes. Besides the gene sequence, the regulation of the gene expression and function should also be accounted for. The widened concept of past and present "gene expression" influences is summarized here as Ge. Also, the concept of "environment" needs some re-consideration in situations where exposure timing (Et) is pivotal: prolonged or repeated exposure to the insult (chemical, physical, life style) affects Ge. This implies that it changes the model system. The interaction of Ge with Et might be denoted as Ge × Et. We provide here general explanations and specific examples for this concept and show how it could be applied in the context of New Approach Methodologies (NAM).


Subject(s)
Adverse Outcome Pathways , Humans , Animals , Uncertainty , Models, Biological
4.
Cell Death Dis ; 13(12): 1055, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36539400

ABSTRACT

Ion channels are non-conventional, druggable oncological targets. The intermediate-conductance calcium-dependent potassium channel (KCa3.1) is highly expressed in the plasma membrane and in the inner mitochondrial membrane (mitoKCa3.1) of various cancer cell lines. The role mitoKCa3.1 plays in cancer cells is still undefined. Here we report the synthesis and characterization of two mitochondria-targeted novel derivatives of a high-affinity KCa3.1 antagonist, TRAM-34, which retain the ability to block channel activity. The effects of these drugs were tested in melanoma, pancreatic ductal adenocarcinoma and breast cancer lines, as well as in vivo in two orthotopic models. We show that the mitochondria-targeted TRAM-34 derivatives induce release of mitochondrial reactive oxygen species, rapid depolarization of the mitochondrial membrane, fragmentation of the mitochondrial network. They trigger cancer cell death with an EC50 in the µM range, depending on channel expression. In contrast, inhibition of the plasma membrane KCa3.1 by membrane-impermeant Maurotoxin is without effect, indicating a specific role of mitoKCa3.1 in determining cell fate. At sub-lethal concentrations, pharmacological targeting of mitoKCa3.1 significantly reduced cancer cell migration by enhancing production of mitochondrial reactive oxygen species and nuclear factor-κB (NF-κB) activation, and by downregulating expression of Bcl-2 Nineteen kD-Interacting Protein (BNIP-3) and of Rho GTPase CDC-42. This signaling cascade finally leads to cytoskeletal reorganization and impaired migration. Overexpression of BNIP-3 or pharmacological modulation of NF-κB and CDC-42 prevented the migration-reducing effect of mitoTRAM-34. In orthotopic models of melanoma and pancreatic ductal adenocarcinoma, the tumors at sacrifice were 60% smaller in treated versus untreated animals. Metastasis of melanoma cells to lymph nodes was also drastically reduced. No signs of toxicity were observed. In summary, our results identify mitochondrial KCa3.1 as an unexpected player in cancer cell migration and show that its pharmacological targeting is efficient against both tumor growth and metastatic spread in vivo.


Subject(s)
Carcinoma, Pancreatic Ductal , Melanoma , Pancreatic Neoplasms , Potassium Channels, Calcium-Activated , Animals , NF-kappa B/metabolism , Calcium/metabolism , Calcium Channels , Potassium Channels , Reactive Oxygen Species/metabolism , Cell Death , Mitochondria/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Pancreatic Neoplasms
5.
Sci Adv ; 8(37): eadd2926, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36103527

ABSTRACT

The mechanistic target of rapamycin complex 1 (mTORC1) regulates cell growth and catabolism in response to nutrients through phosphorylation of key substrates. The tumor suppressor folliculin (FLCN) is a RagC/D guanosine triphosphatase (GTPase)-activating protein (GAP) that regulates mTORC1 phosphorylation of MiT-TFE transcription factors, controlling lysosome biogenesis and autophagy. We determined the cryo-electron microscopy structure of the active FLCN complex (AFC) containing FLCN, FNIP2, the N-terminal tail of SLC38A9, the RagAGDP:RagCGDP.BeFx- GTPase dimer, and the Ragulator scaffold. Relative to the inactive lysosomal FLCN complex structure, FLCN reorients by 90°, breaks contact with RagA, and makes previously unseen contacts with RagC that position its Arg164 finger for catalysis. Disruption of the AFC-specific interfaces of FLCN and FNIP2 with RagC eliminated GAP activity and led to nuclear retention of TFE3, with no effect on mTORC1 substrates S6K or 4E-BP1. The structure provides a basis for regulation of an mTORC1 substrate-specific pathway and a roadmap to discover MiT-TFE family selective mTORC1 antagonists.

7.
Elife ; 102021 07 01.
Article in English | MEDLINE | ID: mdl-34196606

ABSTRACT

The voltage-gated potassium channel Kv1.3 plays an apparent dual physiological role by participating in activation and proliferation of leukocytes as well as promoting apoptosis in several types of tumor cells. Therefore, Kv1.3 is considered a potential pharmacological target for immunodeficiency and cancer. Different cellular locations of Kv1.3, at the plasma membrane or the mitochondria, could be responsible for such duality. While plasma membrane Kv1.3 facilitates proliferation, the mitochondrial channel modulates apoptotic signaling. Several molecular determinants of Kv1.3 drive the channel to the cell surface, but no information is available about its mitochondrial targeting. Caveolins, which are able to modulate cell survival, participate in the plasma membrane targeting of Kv1.3. The channel, via a caveolin-binding domain (CDB), associates with caveolin 1 (Cav1), which localizes Kv1.3 to lipid raft membrane microdomains. The aim of our study was to understand the role of such interactions not only for channel targeting but also for cell survival in mammalian cells. By using a caveolin association-deficient channel (Kv1.3 CDBless), we demonstrate here that while the Kv1.3-Cav1 interaction is responsible for the channel localization in the plasma membrane, a lack of such interaction accumulates Kv1.3 in the mitochondria. Kv1.3 CDBless severely affects mitochondrial physiology and cell survival, indicating that a functional link of Kv1.3 with Cav1 within the mitochondria modulates the pro-apoptotic effects of the channel. Therefore, the balance exerted by these two complementary mechanisms fine-tune the physiological role of Kv1.3 during cell survival or apoptosis. Our data highlight an unexpected role for the mitochondrial caveolin-Kv1.3 axis during cell survival and apoptosis.


Subject(s)
Apoptosis/genetics , Caveolin 1/genetics , Cell Survival/genetics , Kv1.3 Potassium Channel/genetics , Caveolin 1/metabolism , HEK293 Cells , Humans , Kv1.3 Potassium Channel/metabolism , Mitochondria/metabolism
8.
Redox Biol ; 45: 102030, 2021 09.
Article in English | MEDLINE | ID: mdl-34147842

ABSTRACT

Potassium channels are important regulators of cellular homeostasis and targeting these proteins pharmacologically is unveiling important mechanisms in cancer cell biology. Here we demonstrate that pharmacological stimulation of the Kv11.1 potassium channel activity results in mitochondrial reactive oxygen species (ROS) production and fragmentation in breast cancer cell lines and patient-derived organoids independent of breast cancer subtype. mRNA expression profiling revealed that Kv11.1 activity significantly altered expression of genes controlling the production of ROS and endoplasmic-reticulum (ER) stress. Characterization of the transcriptional signature of breast cancer cells treated with Kv11.1 potassium channel activators strikingly revealed an adaptive response to the potentially lethal augmentation of ROS by increasing Nrf2-dependent transcription of antioxidant genes. Nrf2 in this context was shown to promote survival in breast cancer, whereas knockdown of Nrf2 lead to Kv11.1-induced cell death. In conclusion, we found that the Kv11.1 channel activity promotes oxidative stress in breast cancer cells and that suppression of the Nrf2-mediated anti-oxidant survival mechanism strongly sensitized breast cancer cells to a lethal effect of pharmacological activation of Kv11.1.


Subject(s)
Antioxidants , Breast Neoplasms , Antioxidants/pharmacology , Breast Neoplasms/genetics , Endoplasmic Reticulum Stress , Female , Humans , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species
9.
Nat Commun ; 12(1): 2103, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33833234

ABSTRACT

Mitochondrial diseases impair oxidative phosphorylation and ATP production, while effective treatment is still lacking. Defective complex III is associated with a highly variable clinical spectrum. We show that pyocyanin, a bacterial redox cycler, can replace the redox functions of complex III, acting as an electron shunt. Sub-µM pyocyanin was harmless, restored respiration and increased ATP production in fibroblasts from five patients harboring pathogenic mutations in TTC19, BCS1L or LYRM7, involved in assembly/stabilization of complex III. Pyocyanin normalized the mitochondrial membrane potential, and mildly increased ROS production and biogenesis. These in vitro effects were confirmed in both DrosophilaTTC19KO and in Danio rerioTTC19KD, as administration of low concentrations of pyocyanin significantly ameliorated movement proficiency. Importantly, daily administration of pyocyanin for two months was not toxic in control mice. Our results point to utilization of redox cyclers for therapy of complex III disorders.


Subject(s)
Adenosine Triphosphate/biosynthesis , Electron Transport Complex III/metabolism , Membrane Proteins/genetics , Mitochondrial Diseases/drug therapy , Mitochondrial Proteins/genetics , Pyocyanine/pharmacology , ATPases Associated with Diverse Cellular Activities/genetics , Animals , Animals, Genetically Modified , Cell Line , Drosophila melanogaster , Electron Transport Complex III/genetics , Humans , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Mitochondrial Diseases/pathology , Molecular Chaperones/genetics , Oxidation-Reduction/drug effects , Pyocyanine/metabolism , Reactive Oxygen Species/metabolism , Zebrafish
10.
FASEB J ; 35(3): e21362, 2021 03.
Article in English | MEDLINE | ID: mdl-33629768

ABSTRACT

Friedreich ataxia (FRDA) is a neurodegenerative disease resulting from a severe decrease of frataxin (FXN). Most patients carry a GAA repeat expansion in both alleles of the FXN gene, whereas a small fraction of them are compound heterozygous for the expansion and a point mutation in the other allele. FXN is involved in the mitochondrial biogenesis of the FeS-clusters. Distinctive feature of FRDA patient cells is an impaired cellular respiration, likely due to a deficit of key redox cofactors working as electrons shuttles through the respiratory chain. However, a definite relationship between FXN levels, FeS-clusters assembly dysregulation and bioenergetics failure has not been established. In this work, we performed a comparative analysis of the mitochondrial phenotype of cell lines from FRDA patients, either homozygous for the expansion or compound heterozygotes for the G130V mutation. We found that, in healthy cells, FXN and two key proteins of the FeS-cluster assembly machinery are enriched in mitochondrial cristae, the dynamic subcompartment housing the respiratory chain. On the contrary, FXN widely redistributes to the matrix in FRDA cells with defects in respiratory supercomplexes assembly and altered respiratory function. We propose that this could be relevant for the early mitochondrial defects afflicting FRDA cells and that perturbation of mitochondrial morphodynamics could in turn be critical in terms of disease mechanisms.


Subject(s)
Electron Transport Chain Complex Proteins/biosynthesis , Energy Metabolism , Friedreich Ataxia/metabolism , Iron-Binding Proteins/physiology , Mitochondrial Membranes/metabolism , Cell Line , Friedreich Ataxia/pathology , Humans , Iron-Binding Proteins/genetics , Mitochondrial Membranes/pathology , Frataxin
11.
Redox Biol ; 37: 101705, 2020 10.
Article in English | MEDLINE | ID: mdl-33007503

ABSTRACT

The potassium channel Kv1.3, involved in several important pathologies, is the target of a family of psoralen-based drugs whose mechanism of action is not fully understood. Here we provide evidence for a physical interaction of the mitochondria-located Kv1.3 (mtKv1.3) and Complex I of the respiratory chain and show that this proximity underlies the death-inducing ability of psoralenic Kv1.3 inhibitors. The effects of PAP-1-MHEG (PAP-1, a Kv1.3 inhibitor, with six monomeric ethylene glycol units attached to the phenyl ring of PAP-1), a more soluble novel derivative of PAP-1 and of its various portions on mitochondrial physiology indicate that the psoralenic moiety of PAP-1 bound to mtKv1.3 facilitates the diversion of electrons from Complex I to molecular oxygen. The resulting massive production of toxic Reactive Oxygen Species leads to death of cancer cells expressing Kv1.3. In vivo, PAP-1-MHEG significantly decreased melanoma volume. In summary, PAP-1-MHEG offers insights into the mechanisms of cytotoxicity of this family of compounds and may represent a valuable clinical tool.


Subject(s)
Kv1.3 Potassium Channel , Mitochondria , Animals , Cell Line, Tumor , Dissection , Humans , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Mice, Inbred C57BL , Reactive Oxygen Species
12.
Cancers (Basel) ; 12(9)2020 Sep 10.
Article in English | MEDLINE | ID: mdl-32927611

ABSTRACT

Mitochondria are organelles that are mainly involved in the generation of ATP by cellular respiration. In addition, they modulate several intracellular functions, ranging from cell proliferation and differentiation to cell death. Importantly, mitochondria are social and can interact with other organelles, such as the Endoplasmic Reticulum, lysosomes and peroxisomes. This symbiotic relationship gives advantages to both partners in regulating some of their functions related to several aspects of cell survival, metabolism, sensitivity to cell death and metastasis, which can all finally contribute to tumorigenesis. Moreover, growing evidence indicates that modulation of the length and/or numbers of these contacts, as well as of the distance between the two engaged organelles, impacts both on their function as well as on cellular signaling. In this review, we discuss recent advances in the field of contacts and communication between mitochondria and other intracellular organelles, focusing on how the tuning of mitochondrial function might impact on both the interaction with other organelles as well as on intracellular signaling in cancer development and progression, with a special focus on calcium signaling.

13.
Cell Rep ; 28(8): 1949-1960.e6, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433973

ABSTRACT

Wnt signaling affects fundamental development pathways and, if aberrantly activated, promotes the development of cancers. Wnt signaling is modulated by different factors, but whether the mitochondrial energetic state affects Wnt signaling is unknown. Here, we show that sublethal concentrations of different compounds that decrease mitochondrial ATP production specifically downregulate Wnt/ß-catenin signaling in vitro in colon cancer cells and in vivo in zebrafish reporter lines. Accordingly, fibroblasts from a GRACILE syndrome patient and a generated zebrafish model lead to reduced Wnt signaling. We identify a mitochondria-Wnt signaling axis whereby a decrease in mitochondrial ATP reduces calcium uptake into the endoplasmic reticulum (ER), leading to endoplasmic reticulum stress and to impaired Wnt signaling. In turn, the recovery of the ATP level or the inhibition of endoplasmic reticulum stress restores Wnt activity. These findings reveal a mechanism that links mitochondrial energetic metabolism to the control of the Wnt pathway that may be beneficial against several pathologies.


Subject(s)
Adenosine Triphosphate/biosynthesis , Down-Regulation , Endoplasmic Reticulum Stress , Mitochondria/metabolism , Wnt Signaling Pathway , Animals , Cell Line , Fibroblasts/metabolism , Humans , Zebrafish
14.
Cancers (Basel) ; 11(6)2019 May 31.
Article in English | MEDLINE | ID: mdl-31159324

ABSTRACT

Mitochondrial ion channels are emerging oncological targets, as modulation of these ion-transporting proteins may impact on mitochondrial membrane potential, efficiency of oxidative phosphorylation and reactive oxygen production. In turn, these factors affect the release of cytochrome c, which is the point of no return during mitochondrial apoptosis. Many of the currently used chemotherapeutics induce programmed cell death causing damage to DNA and subsequent activation of p53-dependent pathways that finally leads to cytochrome c release from the mitochondrial inter-membrane space. The view is emerging, as summarized in the present review, that ion channels located in this organelle may account in several cases for the resistance that cancer cells can develop against classical chemotherapeutics, by preventing drug-induced apoptosis. Thus, pharmacological modulation of these channel activities might be beneficial to fight chemo-resistance of different types of cancer cells.

15.
Cell Physiol Biochem ; 52(2): 232-239, 2019.
Article in English | MEDLINE | ID: mdl-30816671

ABSTRACT

BACKGROUND/AIMS: Pterostilbene (Pt; trans-3,5-dimethoxy-4'-hydroxystilbene) is a natural phenol found in blueberries and grapevines. It shows remarkable biomedical activities similar to those of resveratrol. Its high bioavailability is a major advantage for possible biomedical applications. The goal of the study was to evaluate the effects of chronic pterostilbene administration on cognitive performance in aged rats with mild cognitive impairment. METHODS: 18-month-old animals were subjected to behavioral tests to establish the "baseline", then divided into treatment and control groups. The former were chronically fed Pt (22.5 mg/kg-day) for 20 consecutive days. At the end of this period all animals were tested again and sacrificed. The dentate gyrus, the hippocampus and the prefrontal and perirhinal cortices were then collected, and RT-qPCR and/or Western blot analyses were performed on a few transcripts/proteins involved in synaptic remodeling. Mitochondrial content was also assessed. RESULTS: Pt administration improved performance in behavioral tests and positively affected memory consolidation. We found increased levels of REST, PSD-95 and mitochondrial porin1 in the dentate gyrus and a positive correlation between T-maze test score and levels of cAMP responsive element binding protein (CREB) phosphorylation. CONCLUSION: These results underscore the therapeutic potential of Pt supplementation for age-related cognitive decline.


Subject(s)
Aging/metabolism , Behavior, Animal/drug effects , Cognition/drug effects , Maze Learning/drug effects , Stilbenes/pharmacology , Animals , CREB-Binding Protein/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Dentate Gyrus/metabolism , Disks Large Homolog 4 Protein/biosynthesis , Rats , Repressor Proteins/biosynthesis
16.
Leukemia ; 33(5): 1148-1160, 2019 05.
Article in English | MEDLINE | ID: mdl-30353029

ABSTRACT

Protein phosphatase 2 A (PP2A) is a tumour suppressor whose strong inhibition underlies the phosphorylation-dependent, anti-apoptotic mechanisms in Chronic Lymphocytic Leukemia (CLL). Inactivation of PP2A is due to the cooperative action of the phosphorylation of Y307 of its catalytic subunit by the aberrant cytosolic pool of the Src Family Kinase Lyn and the interaction with its protein inhibitor SET, which is overexpressed in CLL. In this study, we developed a library of compounds, the most potent being the one named CC11, which restores PP2A activity by disrupting the PP2A/SET complex, thereby triggering the mitochondrial pathway of apoptosis. This process involves the recruitment of the pro-apoptotic BH3-only proteins Bad and Bim to mitochondria, the former upon direct dephosphorylation and the latter being newly expressed upon dephosphorylation and activation of its transcription factor FoxO3a. These findings highlight that PP2A antagonizes the prosurvival pathways controlled by Akt, which phosphorylates and thereby suppresses a variety of pro-apoptotic factors and tumour suppressors including Bad and FoxO3a. Furthermore, the PP2A-mediated pro-apoptotic effect of CC11 is synergistically potentiated by the abrogation of Lyn's activity. Our results show that CC11 represents a promising lead compound for a new therapeutic rationale aimed at abrogating the aberrant oncogenic signals in CLL.


Subject(s)
Apoptosis/drug effects , Forkhead Box Protein O3/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Protein Phosphatase 2/metabolism , bcl-Associated Death Protein/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cytochromes c/genetics , Cytochromes c/metabolism , Enzyme Activation , Gene Expression , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Models, Biological , Phosphorylation
17.
Int J Mol Sci ; 19(7)2018 Jul 15.
Article in English | MEDLINE | ID: mdl-30011966

ABSTRACT

In recent years, several experimental evidences have underlined a new role of ion channels in cancer development and progression. In particular, mitochondrial ion channels are arising as new oncological targets, since it has been proved that most of them show an altered expression during tumor development and the pharmacological targeting of some of them have been demonstrated to be able to modulate cancer growth and progression, both in vitro as well as in vivo in pre-clinical mouse models. In this scenario, pharmacology of mitochondrial ion channels would be in the near future a new frontier for the treatment of tumors. In this review, we discuss the new advances in the field, by focusing our attention on the improvements in new drug developments to target mitochondrial ion channels.


Subject(s)
Antineoplastic Agents/therapeutic use , Ion Channels/antagonists & inhibitors , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Animals , Disease Progression , Humans , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Models, Biological , Molecular Targeted Therapy/trends , Neoplasms/metabolism , Neoplasms/pathology
18.
Biochem Biophys Res Commun ; 500(1): 51-58, 2018 05 27.
Article in English | MEDLINE | ID: mdl-28642134

ABSTRACT

Mitochondria are intracellular organelles involved in several processes from bioenergetics to cell death. In the latest years, ion channels are arising as new possible targets in controlling several cellular functions. The discovery that several plasma membrane located ion channels have intracellular counterparts, has now implemented this consideration and the number of studies enforcing the understanding of their role in different metabolic pathways. In this review, we will discuss the recent updates in the field, focusing our attention on the involvement of potassium channels during mitochondrial mediated apoptotic cell death. Since mitochondria are one of the key organelles involved in this process, it is not surprising that potassium channels located in their inner membrane could be involved in modulating mitochondrial membrane potential, ROS production, and respiratory chain complexes functions. Eventually, these events lead to changes in the mitochondrial fitness that prelude to the cytochrome c release and apoptosis. In this scenario, both the inhibition and the activation of mitochondrial potassium channels could cause cell death, and their targeting could be a novel pharmacological way to treat different human diseases.


Subject(s)
Apoptosis/genetics , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Neoplasms/metabolism , Potassium Channels/metabolism , Animals , Cytochromes c/metabolism , Energy Metabolism/genetics , Gene Expression Regulation , Humans , Ion Transport , Membrane Potential, Mitochondrial/physiology , Mitochondria/ultrastructure , Mitochondrial Membranes/ultrastructure , Neoplasms/genetics , Neoplasms/pathology , Potassium Channels/classification , Potassium Channels/genetics , Reactive Oxygen Species/metabolism , Signal Transduction
19.
Front Oncol ; 7: 239, 2017.
Article in English | MEDLINE | ID: mdl-29034212

ABSTRACT

Previous results link the mitochondrial potassium channel Kv1.3 (mitoKv1.3) to the regulation of apoptosis. By synthesizing new, mitochondria-targeted derivatives (PAPTP and PCARBTP) of PAP-1, a specific membrane-permeant Kv1.3 inhibitor, we have recently provided evidence that both drugs acting on mitoKv1.3 are able to induce apoptosis and reduce tumor growth in vivo without affecting healthy tissues and cells. In the present article, by exploiting these new drugs, we addressed the question whether mitoKv1.3 contributes to the regulation of cell proliferation as well. When used at low concentrations, which do not compromise cell survival, both drugs slightly increased the percentage of cells in S phase while decreased the population at G0/G1 stage of cells from two different pancreatic ductal adenocarcinoma lines. Our data suggest that the observed modulation is related to ROS levels within the cells, opening the way to link mitochondrial ion channel function to downstream, ROS-related signaling events that might be important for cell cycle progression.

20.
Cancer Cell ; 31(4): 516-531.e10, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28399409

ABSTRACT

The potassium channel Kv1.3 is highly expressed in the mitochondria of various cancerous cells. Here we show that direct inhibition of Kv1.3 using two mitochondria-targeted inhibitors alters mitochondrial function and leads to reactive oxygen species (ROS)-mediated death of even chemoresistant cells independently of p53 status. These inhibitors killed 98% of ex vivo primary chronic B-lymphocytic leukemia tumor cells while sparing healthy B cells. In orthotopic mouse models of melanoma and pancreatic ductal adenocarcinoma, the compounds reduced tumor size by more than 90% and 60%, respectively, while sparing immune and cardiac functions. Our work provides direct evidence that specific pharmacological targeting of a mitochondrial potassium channel can lead to ROS-mediated selective apoptosis of cancer cells in vivo, without causing significant side effects.


Subject(s)
Antineoplastic Agents/pharmacology , Kv1.3 Potassium Channel/antagonists & inhibitors , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Potassium Channel Blockers/pharmacology , Aged , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Case-Control Studies , Coumarins/pharmacology , Drug Stability , Female , Humans , Kv1.3 Potassium Channel/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Melanoma/drug therapy , Melanoma/pathology , Mice, Inbred C57BL , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Targeted Therapy , Organophosphorus Compounds/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Potassium Channel Blockers/chemical synthesis , Potassium Channel Blockers/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...