Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
EMBO J ; 40(10): e106214, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33932034

ABSTRACT

BNIP3 is a mitophagy receptor with context-dependent roles in cancer, but whether and how it modulates melanoma growth in vivo remains unknown. Here, we found that elevated BNIP3 levels correlated with poorer melanoma patient's survival and depletion of BNIP3 in B16-F10 melanoma cells compromised tumor growth in vivo. BNIP3 depletion halted mitophagy and enforced a PHD2-mediated downregulation of HIF-1α and its glycolytic program both in vitro and in vivo. Mechanistically, we found that BNIP3-deprived melanoma cells displayed increased intracellular iron levels caused by heightened NCOA4-mediated ferritinophagy, which fostered PHD2-mediated HIF-1α destabilization. These effects were not phenocopied by ATG5 or NIX silencing. Restoring HIF-1α levels in BNIP3-depleted melanoma cells rescued their metabolic phenotype and tumor growth in vivo, but did not affect NCOA4 turnover, underscoring that these BNIP3 effects are not secondary to HIF-1α. These results unravel an unexpected role of BNIP3 as upstream regulator of the pro-tumorigenic HIF-1α glycolytic program in melanoma cells.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melanoma/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Computational Biology , Female , Gas Chromatography-Mass Spectrometry , Humans , Immunoblotting , Immunohistochemistry , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Signal Transduction/genetics , Signal Transduction/physiology
2.
Biol Chem ; 400(2): 187-193, 2019 01 28.
Article in English | MEDLINE | ID: mdl-29924728

ABSTRACT

Aerobic glycolysis ('Warburg effect') is used by cancer cells to fuel tumor growth. Interestingly, metastatic melanoma cells rely on glutaminolysis rather than aerobic glycolysis for their bioenergetic needs through the tricarboxylic acid (TCA) cycle. Here, we compared the effects of glucose or glutamine on melanoma cell proliferation, migration and oxidative phosphorylation in vitro. We found that glutamine-driven melanoma cell's aggressive traits positively correlated with increased expression of HIF1α and its pro-autophagic target BNIP3. BNIP3 silencing reduced glutamine-mediated effects on melanoma cell growth, migration and bioenergetics. Hence, BNIP3 is a vital component of the mitochondria quality control required for glutamine-driven melanoma aggressiveness.


Subject(s)
Glutamine/metabolism , Melanoma/pathology , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Skin Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Energy Metabolism , Gene Silencing , Humans , Melanoma/metabolism , Membrane Proteins/genetics , Neoplasm Metastasis , Proto-Oncogene Proteins/genetics , Skin Neoplasms/metabolism
3.
Photochem Photobiol Sci ; 14(8): 1410-24, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25666525

ABSTRACT

Autophagy is a major catabolic pathway in a eukaryotic cell, employed for cellular self-degradation of obsolete or damaged cytoplasmic components serving as a major quality control and recycling mechanism that supports cell survival. Autophagy is fundamentally a cytoprotective and pro-survival process yet in general, it has become clear through a number of studies that autophagy has an exceedingly contextual role in cancer biology; conditional on which phase, location or type of oncogenic trigger and/or therapy is under consideration, the role of autophagy could end up fluctuating from pro- to anti-tumourigenic. Numerous studies have revealed that, contingent on the photosensitiser under consideration, autophagy triggered by PDT either adds to therapy resistance (by suppressing cell death) or vulnerability (by enabling autophagic cell death). Beyond cell death regulation, cancer cell-associated autophagy also supports resistance against PDT by reducing anticancer immune effector mechanisms. In this review, we have concisely described the state-of-the-art and the prevailing gap-in-knowledge vis-à-vis the role of PDT-triggered autophagy in cancer therapy resistance or susceptibility.


Subject(s)
Autophagy , Neoplasms/drug therapy , Neoplasms/immunology , Photochemotherapy , Animals , Autophagy/drug effects , Autophagy/radiation effects , Humans , Photosensitizing Agents/pharmacology
4.
Exp Dermatol ; 23(2): 101-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24313465

ABSTRACT

Despite advances in cancer diagnosis and therapy, metastatic melanoma remains untreatable, due to its notorious resistance to apoptosis, deeming traditional therapies obsolete. Deregulated PI3K/Akt signalling is a common oncogenic event enabling melanocyte transformation and represents a significant and 'druggable' pathway in melanoma. Emerging data show that the ability of cancer cells to survive is also facilitated by alteration of vital homoeostatic mechanisms, such as autophagy. Although the role of autophagy in melanoma is still controversial, recent studies suggest that basal autophagy is down-modulated in primary melanomas. However, the dynamic connection between pro-tumorigenic PI3K/Akt and autophagy during melanoma progression has not been systematically studied. By using human primary melanocytes, incipient melanoma and metastatic melanoma cell lines, we show that early in melanomagenesis, increased Akt activity is associated with a low baseline autophagic flux. However, during melanoma progression, metastatic melanoma cells regain the ability to stimulate autophagic flux, supporting survival. Heightened autophagy is associated with an attenuated Akt activation status and can be suppressed by overexpressing a constitutive active mutant of Akt. On the other hand, blocking the higher Akt activity of primary melanoma is sufficient to incite autophagy. Interestingly, we found that although Akt supports survival of melanocytes and all melanoma cell lines, autophagy inhibition specifically targeted the metastatic melanoma cells, thus indicating a stage-specific requirement for Akt and autophagic flux, throughout melanoma progression. Therefore, this study highlights a dynamic interplay between Akt signalling and autophagic rescue in melanoma, which should be considered in the design of therapeutic strategies targeting these pathways.


Subject(s)
Autophagy/physiology , Melanoma/pathology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-akt/physiology , Cell Line, Tumor , Chloroquine/pharmacology , Disease Progression , Humans , In Vitro Techniques , Melanocytes/metabolism , Melanocytes/pathology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins B-raf/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Signal Transduction/physiology , Tumor Stem Cell Assay
5.
Biochim Biophys Acta ; 1816(1): 1-12, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21338656

ABSTRACT

The hypoxia inducible factor-1α (HIF-1α) is a pleiotropic transcription factor typically activated in response to low oxygen tension as well as other stress factors in normoxic conditions. Upon activation HIF-1α mediates the transcriptional activation of target genes involved in a variety of processes comprising stress adaptation, metabolism, growth and invasion, but also apoptotic cell death. The molecular mechanisms, signaling pathways and downstream targets evoked by the activation of HIF-1α in epidermal cells are becoming increasingly understood and underscore the participation of HIF-1α in crucial processes including malignant transformation and cancer progression. Recent studies have implicated HIF-1α as an integral part of the multifaceted signal transduction initiated by the exposure of keratinocytes to ultraviolet radiation B (UVB), which represents the most ubiquitous hazard for human skin and the principal risk factor for skin cancer. HIF-1α activation by UVB exposure contributes to either repair or the removal of UVB-damaged keratinocytes by inducing apoptosis, thus revealing a tumor suppressor role for HIF-1α in these cells. On the other hand, the constitutive expression of HIF-1α evoked by the mild hypoxic state of the skin has been implicated as a positive factor in the transformation of normal melanocytes into malignant melanoma, one of the most aggressive types of human cancers. Here we review the uncovered and complex role of HIF-1α in skin carcinogenesis.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Skin Neoplasms/etiology , Animals , Cell Hypoxia , Humans , Neoplasms, Radiation-Induced/etiology , Skin/pathology , Ultraviolet Rays/adverse effects
6.
Nat Commun ; 13(1): 2760, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35589749

ABSTRACT

Autophagy has vasculoprotective roles, but whether and how it regulates lymphatic endothelial cells (LEC) homeostasis and lymphangiogenesis is unknown. Here, we show that genetic deficiency of autophagy in LEC impairs responses to VEGF-C and injury-driven corneal lymphangiogenesis. Autophagy loss in LEC compromises the expression of main effectors of LEC identity, like VEGFR3, affects mitochondrial dynamics and causes an accumulation of lipid droplets (LDs) in vitro and in vivo. When lipophagy is impaired, mitochondrial ATP production, fatty acid oxidation, acetyl-CoA/CoA ratio and expression of lymphangiogenic PROX1 target genes are dwindled. Enforcing mitochondria fusion by silencing dynamin-related-protein 1 (DRP1) in autophagy-deficient LEC fails to restore LDs turnover and lymphatic gene expression, whereas supplementing the fatty acid precursor acetate rescues VEGFR3 levels and signaling, and lymphangiogenesis in LEC-Atg5-/- mice. Our findings reveal that lipophagy in LEC by supporting FAO, preserves a mitochondrial-PROX1 gene expression circuit that safeguards LEC responsiveness to lymphangiogenic mediators and lymphangiogenesis.


Subject(s)
Lymphangiogenesis , Lymphatic Vessels , Animals , Autophagy/genetics , Endothelial Cells/metabolism , Fatty Acids/metabolism , Lipid Droplets/metabolism , Lymphangiogenesis/genetics , Lymphatic Vessels/metabolism , Mice , Mitochondria , Transcription Factors/metabolism
7.
Front Oncol ; 9: 171, 2019.
Article in English | MEDLINE | ID: mdl-30949450

ABSTRACT

Cancer cell-stromal cell crosstalk is orchestrated by a plethora of ligand-receptor interactions generating a tumor microenvironment (TME) which favors tumor growth. The high pro-angiogenic nature of the TME perpetuates the chaotic network of structurally immature, low pericyte-covered vessels characteristic of the tumor vasculature. We previously demonstrated that chloroquine (CQ) -a lysosomotropic agent used as first-generation autophagy blocker in clinical trials- induced tumor vessel normalization and reduced tumor hypoxia. CQ improved both vessel structure and maturation, whereas the conditional knockout of the crucial autophagy gene Atg5 in endothelial cells (ECs) did not, thus highlighting a potential differential role for EC-associated autophagy and the lysosomes in pathological tumor angiogenesis. However, how CQ or ATG5-deficiency in ECs affect angiogenic signals regulating EC-pericyte interface and therefore vessel maturation, remains unknown. Here, we show that in ECs CQ constrained VEGF-A-mediated VEGF receptor (VEGFR)2 phosphorylation, a driver of angiogenic signaling. In the presence of CQ we observed increased expression of the decoy receptor VEGFR1 and of a lower molecular weight form of VEGFR2, suggesting receptor cleavage. Consequently, VEGF-A-driven EC spheroid sprouting was reduced by CQ treatment. Furthermore, CQ significantly affected the transcription and secretion of platelet-derived growth factor (PDGF)-AB/BB (upregulated) and Endothelin-1 (EDN1, downregulated), both modulators of perivascular cell (PC) behavior. In contrast, silencing of ATG5 in ECs had no effect on VEGFR2 to VEGFR1 ratio nor on PDGFB and EDN1 expression. Accordingly, mice harboring B16F10 melanoma tumors treated with CQ, displayed both an increased number of αSMA+ PCs covering tumor vessels and co-expressed PDGF receptor-ß, enabling PDGF ligand dependent recruitment. Moreover, upon CQ treatment the tumoral expression of angiopoietin-1 (Angpt1), which retains mural cells, and induces vessel stabilization by binding to the EC-localized cognate receptor (TIE2), was increased thus supporting the vessel normalization function of CQ. These features associated with improved tumor vasculature were not phenocopied by the specific deletion of Atg5 in ECs. In conclusion, this study further unravels endothelial cell autonomous and non-autonomous mechanisms by which CQ "normalizes" the intercellular communication in the tumor vasculature independent of autophagy.

8.
Ecancermedicalscience ; 11: 781, 2017.
Article in English | MEDLINE | ID: mdl-29225688

ABSTRACT

Chloroquine (CQ) and hydroxychloroquine (HCQ) are well-known 4-aminoquinoline antimalarial agents. Scientific evidence also supports the use of CQ and HCQ in the treatment of cancer. Overall, preclinical studies support CQ and HCQ use in anti-cancer therapy, especially in combination with conventional anti-cancer treatments since they are able to sensitise tumour cells to a variety of drugs, potentiating the therapeutic activity. Thus far, clinical results are mostly in favour of the repurposing of CQ. However, over 30 clinical studies are still evaluating the activity of both CQ and HCQ in different cancer types and in combination with various standard treatments. Interestingly, CQ and HCQ exert effects both on cancer cells and on the tumour microenvironment. In addition to inhibition of the autophagic flux, which is the most studied anti-cancer effect of CQ and HCQ, these drugs affect the Toll-like receptor 9, p53 and CXCR4-CXCL12 pathway in cancer cells. In the tumour stroma, CQ was shown to affect the tumour vasculature, cancer-associated fibroblasts and the immune system. The evidence reviewed in this paper indicates that both CQ and HCQ deserve further clinical investigations in several cancer types. Special attention about the drug (CQ versus HCQ), the dose and the schedule of administration should be taken in the design of new trials.

9.
Mol Cell Oncol ; 3(1): e970097, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27308577

ABSTRACT

Chloroquine is used clinically as an autophagy blocker to potentiate anticancer treatments. However, whether chloroquine acts solely through autophagy-dependent and cancer cell autonomous mechanisms has remained elusive. In a recent study we found that chloroquine reduced intratumoral hypoxia and metastasis, while improving chemotherapy response, largely through an autophagy-independent, NOTCH1-reliant mechanism of tumor vessel normalization.

10.
FEBS J ; 283(1): 25-38, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26443003

ABSTRACT

A common feature of solid tumors is their ability to incite the formation of new blood and lymph vessels trough the processes of angiogenesis and lymphangiogenesis, respectively, to support tumor growth and favor metastatic dissemination. As a result of the lack of feedback regulatory control mechanisms or due to the exacerbated presence of pro-angiogenic signals within the tumor microenvironment, the tumor endothelium receives continuous signals to sprout and develop, generating vessels that are structurally and functionally abnormal. An emerging mechanism playing a central role in shaping the tumor vasculature is the endothelial-vesicular network that regulates trafficking/export and degradation of key signaling proteins and membrane receptors, including the vascular endothelial growth-factor receptor-2/3 and members of the Notch pathway. Here we will discuss recent evidence highlighting how vesicular trafficking mechanisms in endothelial cells contribute to pathological angiogenesis/lymphangiogenesis and can provide novel and exploitable targets in antiangiogenic therapies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Endothelial Cells/pathology , Humans , Lymphangiogenesis/drug effects , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Protein Transport/drug effects
11.
Front Oncol ; 6: 240, 2016.
Article in English | MEDLINE | ID: mdl-27896217

ABSTRACT

Melanoma has emerged as a paradigm of a highly aggressive and plastic cancer, capable to co-opt the tumor stroma in order to adapt to the hostile microenvironment, suppress immunosurveillance mechanisms, and disseminate. In particular, oncogene- and aneuploidy-driven dysregulations of proteostasis in melanoma cells impose a rewiring of central proteostatic processes, such as the heat shock and unfolded protein responses, autophagy, and the endo-lysosomal system, to avoid proteotoxicity. Research over the past decade has indicated that alterations in key nodes of these proteostasis pathways act in conjunction with crucial oncogenic drivers to increase intrinsic adaptations of melanoma cells against proteotoxic stress, modulate the high metabolic demand of these cancer cells and the interface with other stromal cells, through the heightened release of soluble factors or exosomes. Here, we overview and discuss how key proteostasis pathways and vesicular trafficking mechanisms are turned into vital conduits of melanoma progression, by supporting cancer cell's adaptation to the microenvironment, limiting or modulating the ability to respond to therapy and fueling melanoma dissemination.

12.
Mol Cell Oncol ; 2(1): e975089, 2015.
Article in English | MEDLINE | ID: mdl-27308392

ABSTRACT

The endoplasmic reticulum (ER) is at the center of a number of vital cellular processes such as cell growth, death, and differentiation, crosstalk with immune or stromal cells, and maintenance of proteostasis or homeostasis, and ER functions have implications for various pathologies including cancer. Recently, a number of major hallmarks of cancer have been delineated that are expected to facilitate the development of anticancer therapies. However, therapeutic induction of ER stress as a strategy to broadly target multiple hallmarks of cancer has been seldom discussed despite the fact that several primary or secondary ER stress-inducing therapies have been found to exhibit positive clinical activity in cancer patients. In the present review we provide a brief historical overview of the major discoveries and milestones in the field of ER stress biology with important implications for anticancer therapy. Furthermore, we comprehensively discuss possible strategies enabling the targeting of multiple hallmarks of cancer with therapy-induced ER stress.

13.
Mitochondrion ; 19 Pt A: 58-68, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25042464

ABSTRACT

Autophagy, or self-eating, is the most extensively studied lysosomal degradation pathway for the recycling of obsolete or damaged cytoplasmic materials, including proteins and organelles. Although this pathway was initially thought to function as trafficking system for 'in bulk' degradation by the lysosomes of cytoplasmic material, it is now widely appreciated that cargo selection by the autophagic machinery is a major process underlying the cytoprotective or--possibly--pro-death functions ascribed to this catabolic process. Indeed increasing evidence suggests that in mammalian cells the removal of dysfunctional or aged mitochondria occurs through a selective degradation pathway known as 'mitophagy'. Due to the crucial role of mitochondria in energy metabolism, redox control and cell survival/death decision, deregulated mitophagy can potentially impact a variety of crucial cell autonomous and non-autonomous processes. Accumulating evidence indicates that during malignant transformation aggressive cancers hijack autophagy to preserve energy fitness and to acquire the plasticity required to adapt to the hostile microenvironment. However, whether and how mitophagy contributes to carcinogenesis, which pathways regulate this process in the cancer cells and how cancer cell-mitophagy impacts and modifies the tumor microenvironment and therapeutic responses, remain largely unanswered issues. In this review, we discuss novel paradigms and pathways regulating mitophagy in mammalian cells and the impact this process might have on one of the most dreadful human malignancies, melanoma.


Subject(s)
Autophagy/physiology , Melanoma/pathology , Mitochondria/metabolism , Mitophagy/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Melanoma/metabolism , Signal Transduction/physiology
14.
Autophagy ; 10(11): 2082-4, 2014.
Article in English | MEDLINE | ID: mdl-25484095

ABSTRACT

Chloroquine (CQ) is exploited in clinical trials as an autophagy blocker to potentiate anticancer therapy, but it is unknown if it solely acts by inhibiting cancer cell-autonomous autophagy. Our recent study shows that besides blocking cancer cell growth, CQ also affects endothelial cells (ECs) and promotes tumor vessel normalization. This vessel normalizing effect of CQ reduces tumor hypoxia, cancer cell intravasation, and metastasis, while improving the delivery and response to chemotherapy. By compromising autophagy in melanoma cells or using mice with a conditional knockout of ATG5 in ECs, we found that the favorable effects of CQ on the tumor vasculature do not rely on autophagy. CQ-induced vessel normalization relies mainly on altered endolysosomal trafficking and sustained NOTCH1 signaling in ECs. Remarkably these CQ-mediated effects are abrogated when tumors are grown in mice harboring EC-specific deletion of NOTCH1. The autophagy-independent vessel normalization by CQ leading to improved delivery and tumor response to chemotherapy further advocates its clinical use in combination with anticancer treatments.


Subject(s)
Antimalarials/chemistry , Autophagy , Chloroquine/chemistry , Neoplasms/drug therapy , Animals , Endosomes/metabolism , Gene Deletion , Humans , Hypoxia , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/pathology , Receptor, Notch1/metabolism
15.
Cancer Cell ; 26(2): 190-206, 2014 Aug 11.
Article in English | MEDLINE | ID: mdl-25117709

ABSTRACT

Chloroquine (CQ) has been evaluated as an autophagy blocker for cancer treatment, but it is unknown if it acts solely by inhibiting cancer cell autophagy. We report that CQ reduced tumor growth but improved the tumor milieu. By normalizing tumor vessel structure and function and increasing perfusion, CQ reduced hypoxia, cancer cell invasion, and metastasis, while improving chemotherapy delivery and response. Inhibiting autophagy in cancer cells or endothelial cells (ECs) failed to induce such effects. CQ's vessel normalization activity relied mainly on alterations of endosomal Notch1 trafficking and signaling in ECs and was abrogated by Notch1 deletion in ECs in vivo. Thus, autophagy-independent vessel normalization by CQ restrains tumor invasion and metastasis while improving chemotherapy, supporting the use of CQ for anticancer treatment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Autophagy , Chloroquine/pharmacology , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/prevention & control , Skin Neoplasms/drug therapy , Angiogenesis Inhibitors/therapeutic use , Animals , Autophagy-Related Protein 5 , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/therapeutic use , Drug Synergism , Endothelial Cells/drug effects , Endothelial Cells/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Microtubule-Associated Proteins/metabolism , Neoplasm Invasiveness , Neovascularization, Pathologic/metabolism , Receptor, Notch1/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
16.
Trends Mol Med ; 19(7): 428-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23714574

ABSTRACT

Autophagy, the major lysosomal pathway for recycling intracellular components including whole organelles, is emerging as a key process modulating tumorigenesis, tumor-stroma interactions, and cancer therapy. Research over the past decade has highlighted a context-dependent and dynamic role for autophagy in cancer: it is tumor suppressive in the early stages of cancer development, but fuels the growth of established tumors. Likewise, the stimulation of autophagy in response to therapeutics can contextually favor or weaken chemoresistance and antitumor immunity. From a therapeutic perspective, understanding whether, when, and how autophagy can be harnessed to kill cancer cells remains challenging. In this review, we discuss new connections that reveal the role of autophagy in shaping tumor-stroma interaction during carcinogenesis and in the context of anticancer treatments.


Subject(s)
Autophagy/physiology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Cell Communication , Disease Progression , Humans , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Stromal Cells/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
17.
PLoS One ; 8(10): e76550, 2013.
Article in English | MEDLINE | ID: mdl-24098529

ABSTRACT

Melanoma remains the most lethal skin cancer, mainly because of high resistance to therapy. Side population (SP) cells are found in many types of cancer and are usually enriched in therapy-resistant as well as tumorigenic cells. Here, we identified a Hoechst dye-effluxing SP in a large series of human melanoma samples representing different progression phases. The SP size did not change with disease stage but was correlated with the prognostic "Breslow's depth" in the primary (cutaneous) tumors. When injected into immunodeficient mice, the SP generated larger tumors than the bulk "main population" (MP) melanoma cells in two consecutive generations, and showed tumorigenic capacity at lower cell numbers than the MP. In addition, the SP reconstituted the heterogeneous composition of the human A375 melanoma cell line, and its clonogenic activity was 2.5-fold higher than that of the MP. Gene-expression analysis revealed upregulated expression in the melanoma SP (versus the MP) of genes associated with chemoresistance and anti-apoptosis. Consistent with these molecular characteristics, the SP increased in proportion when A375 cells were exposed to the melanoma standard chemotherapeutic agent dacarbazine, and to the aggravating condition of hypoxia. In addition, the SP showed enhanced expression of genes related to cell invasion and migration, as well as to putative (melanoma) cancer stem cells (CSC) including ABCB1 and JARID1B. ABCB1 immunoreactivity was detected in a number of tumor cells in human melanomas, and in particular in clusters at the invasive front of the primary tumors. Together, our findings support that the human melanoma SP is enriched in tumorigenic and chemoresistant capacity, considered key characteristics of CSC. The melanoma SP may therefore represent an interesting therapeutic target.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Side-Population Cells/metabolism , Skin Neoplasms/genetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Dacarbazine/pharmacology , Disease Progression , Drug Resistance, Neoplasm/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, SCID , Neoplasm Proteins/metabolism , Neoplasm Staging , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Side-Population Cells/drug effects , Side-Population Cells/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Burden
18.
Free Radic Biol Med ; 52(6): 1111-20, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22245094

ABSTRACT

The naturally occurring skin hypoxia has emerged as a crucial host factor of the epidermal microenvironment. We wanted to systematically investigate how reduced oxygen availability of the epidermis modulates the response of keratinocytes and melanocytes to noxious ultraviolet B radiation (UVB). We report that the exposure of normal human keratinocytes (NHKs) or melanocytes (NHEMs) to mild hypoxia drastically impacts cell death responses following UVB irradiation. The hypoxic microenvironment favors survival and reduces apoptosis of UVB-irradiated NHEMs and their malignant counterparts (melanoma cells). In contrast, NHKs, but not the transformed keratinocytes, under hypoxic conditions display increased levels of reactive oxygen species (ROS) and are significantly sensitized to UVB-mediated apoptosis as compared to NHKs treated under normoxic conditions. Prolonged exposure of UVB-treated NHKs to hypoxia triggers a sustained and reactive oxygen species-dependent activation of the stress kinases p38(MAPK) and JNKs, which in turn, engage the activation of Noxa and Bim proapoptotic proteins. Combined silencing of Noxa and Bim significantly inhibits UVB-mediated apoptosis under hypoxic conditions, demonstrating that hypoxia results in an amplification of the intrinsic apoptotic pathway. Physiologically occurring skin hypoxia, by facilitating the specific removal of UVB-damaged keratinocytes, may represent a decisive host factor impeding important steps of the photocarcinogenesis process.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Hypoxia/metabolism , Keratinocytes/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Skin Neoplasms/metabolism , Apoptosis/genetics , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Transformation, Neoplastic , Cytoprotection/genetics , Homeostasis , Humans , Hypoxia/pathology , Keratinocytes/pathology , Keratinocytes/radiation effects , Melanocytes/pathology , Melanocytes/radiation effects , Melanoma/genetics , Melanoma/pathology , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Radiation Tolerance/genetics , Reactive Oxygen Species/metabolism , Skin/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects
19.
Autophagy ; 6(7): 838-54, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20505317

ABSTRACT

Mounting evidence suggests that reactive oxygen species (ROS) are multifaceted signaling molecules implicated in a variety of cellular programs during physiological as well as pathological conditions. Recently, ROS produced endogenously, by deranged metabolism of cancer cells, or exogenously, by ROS-generating drugs, have been shown to promote macroautophagy, a lysosomal pathway of self-degradation with essential prosurvival functions. Several molecular aspects of the modulation of autophagy pathways by ROS have been revealed in the past years and it is now clear that these processes are mutually linked and play a crucial role in cancer progression and in response to cancer therapeutics. In this review we address the molecular mechanisms underlying the activation of autophagy pathways by ROS and focus on the role of autophagy in cancer cells responding to ROS-producing agents, which are utilized as a therapeutic modality to kill cancer cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/physiology , Neoplasms/therapy , Reactive Oxygen Species/metabolism , Apoptosis/physiology , Clinical Trials as Topic , Humans , Oxidation-Reduction , Oxidative Stress/physiology , Photochemotherapy , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL