Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 6108, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257646

ABSTRACT

Duchenne muscular dystrophy (DMD) affects 1 in 3500 live male births. To date, there is no effective cure for DMD, and the identification of novel molecular targets involved in disease progression is important to design more effective treatments and therapies to alleviate DMD symptoms. Here, we show that protein levels of the Bromodomain and extra-terminal domain (BET) protein BRD4 are significantly increased in the muscle of the mouse model of DMD, the mdx mouse, and that pharmacological inhibition of the BET proteins has a beneficial outcome, tempering oxidative stress and muscle damage. Alterations in reactive oxygen species (ROS) metabolism are an early event in DMD onset and they are tightly linked to inflammation, fibrosis, and necrosis in skeletal muscle. By restoring ROS metabolism, BET inhibition ameliorates these hallmarks of the dystrophic muscle, translating to a beneficial effect on muscle function. BRD4 direct association to chromatin regulatory regions of the NADPH oxidase subunits increases in the mdx muscle and JQ1 administration reduces BRD4 and BRD2 recruitment at these regions. JQ1 treatment reduces NADPH subunit transcript levels in mdx muscles, isolated myofibers and DMD immortalized myoblasts. Our data highlight novel functions of the BET proteins in dystrophic skeletal muscle and suggest that BET inhibitors may ameliorate the pathophysiology of DMD.


Subject(s)
Muscular Dystrophy, Duchenne/metabolism , Nuclear Proteins/metabolism , Oxidative Stress/drug effects , Transcription Factors/metabolism , Animals , Azepines/pharmacology , Disease Models, Animal , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , NADP , NADPH Oxidases/metabolism , Neuromuscular Diseases/metabolism , Nuclear Proteins/drug effects , Reactive Oxygen Species/metabolism , Transcription Factors/drug effects , Triazoles/pharmacology
2.
iScience ; 23(10): 101604, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33205017

ABSTRACT

SMYD3 is frequently overexpressed in a wide variety of cancers. Indeed, its inactivation reduces tumor growth in preclinical in vivo animal models. However, extensive characterization in vitro failed to clarify SMYD3 function in cancer cells, although confirming its importance in carcinogenesis. Taking advantage of a SMYD3 mutant variant identified in a high-risk breast cancer family, here we show that SMYD3 phosphorylation by ATM enables the formation of a multiprotein complex including ATM, SMYD3, CHK2, and BRCA2, which is required for the final loading of RAD51 at DNA double-strand break sites and completion of homologous recombination (HR). Remarkably, SMYD3 pharmacological inhibition sensitizes HR-proficient cancer cells to PARP inhibitors, thereby extending the potential of the synthetic lethality approach in human tumors.

3.
Cancers (Basel) ; 12(1)2020 Jan 06.
Article in English | MEDLINE | ID: mdl-31935919

ABSTRACT

SMYD3 is a member of the SMYD lysine methylase family and plays an important role in the methylation of various histone and non-histone targets. Aberrant SMYD3 expression contributes to carcinogenesis and SMYD3 upregulation was proposed as a prognostic marker in various solid cancers. Here we summarize SMYD3-mediated regulatory mechanisms, which are implicated in the pathophysiology of cancer, as drivers of distinct oncogenic pathways. We describe SMYD3-dependent mechanisms affecting cancer progression, highlighting SMYD3 interplay with proteins and RNAs involved in the regulation of cancer cell proliferation, migration and invasion. We also address the effectiveness and mechanisms of action for the currently available SMYD3 inhibitors. The findings analyzed herein demonstrate that a complex network of SMYD3-mediated cytoplasmic and nuclear interactions promote oncogenesis across different cancer types. These evidences depict SMYD3 as a modulator of the transcriptional response and of key signaling pathways, orchestrating multiple oncogenic inputs and ultimately, promoting transcriptional reprogramming and tumor transformation. Further insights into the oncogenic role of SMYD3 and its targeting of different synergistic oncogenic signals may be beneficial for effective cancer treatment.

4.
Nucleic Acids Res ; 47(3): 1278-1293, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30544196

ABSTRACT

SMYD3 is a methylase previously linked to cancer cell invasion and migration. Here we show that SMYD3 favors TGFß-induced epithelial-mesenchymal transition (EMT) in mammary epithelial cells, promoting mesenchymal and EMT transcription factors expression. SMYD3 directly interacts with SMAD3 but it is unnecessary for SMAD2/3 phosphorylation and nuclear translocation. Conversely, SMYD3 is indispensable for SMAD3 direct association to EMT genes regulatory regions. Accordingly, SMYD3 knockdown or its pharmacological blockade with the BCI121 inhibitor dramatically reduce TGFß-induced SMAD3 association to the chromatin. Remarkably, BCI121 treatment attenuates mesenchymal genes transcription in the mesenchymal-like MDA-MB-231 cell line and reduces their invasive ability in vivo, in a zebrafish xenograft model. In addition, clinical datasets analysis revealed that higher SMYD3 levels are linked to a less favorable prognosis in claudin-low breast cancers and to a reduced metastasis free survival in breast cancer patients. Overall, our data point at SMYD3 as a pivotal SMAD3 cofactor that promotes TGFß-dependent mesenchymal gene expression and cell migration in breast cancer, and support SMYD3 as a promising pharmacological target for anti-cancer therapy.


Subject(s)
Breast Neoplasms/genetics , Histone-Lysine N-Methyltransferase/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Chromatin/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Zebrafish
5.
Antioxid Redox Signal ; 24(18): 1031-44, 2016 06 20.
Article in English | MEDLINE | ID: mdl-26972385

ABSTRACT

UNLABELLED: Aquaporin-8 (AQP8) allows the bidirectional transport of water and hydrogen peroxide across biological membranes. Depending on its concentration, H2O2 exerts opposite roles, amplifying growth factor signaling in physiological conditions, but causing severe cell damage when in excess. Thus, H2O2 permeability is likely to be tightly controlled in living cells. AIMS: In this study, we investigated whether and how the transport of H2O2 through plasma membrane AQP8 is regulated, particularly during cell stress. RESULTS: We show that diverse cellular stress conditions, including heat, hypoxia, and ER stress, reversibly inhibit the permeability of AQP8 to H2O2 and water. Preventing the accumulation of intracellular reactive oxygen species (ROS) during stress counteracts AQP8 blockade. Once inhibition is established, AQP8-dependent transport can be rescued by reducing agents. Neither H2O2 nor water transport is impaired in stressed cells expressing a mutant AQP8, in which cysteine 53 had been replaced by serine. Cells expressing this mutant are more resistant to stress-, drug-, and radiation-induced growth arrest and death. INNOVATION AND CONCLUSION: The control of AQP8-mediated H2O2 transport provides a novel mechanism to regulate cell signaling and survival during stress. Antioxid. Redox Signal. 24, 1031-1044.


Subject(s)
Aquaporins/metabolism , Oxidative Stress , Aquaporins/genetics , Biological Transport , Cell Membrane Permeability , Cell Proliferation , Cell Survival , Gene Expression , HeLa Cells , Humans , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Saccharomyces cerevisiae , Water/metabolism
6.
Biochim Biophys Acta ; 1858(1): 1-11, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26456554

ABSTRACT

Aquaglyceroporins are integral membrane proteins that are permeable to glycerol as well as water. The movement of glycerol from a tissue/organ to the plasma and vice versa requires the presence of different aquaglyceroporins that can regulate the entrance or the exit of glycerol across the plasma membrane. Actually, different aquaglyceroporins have been discovered in the adipose tissue, small intestine, liver, kidney, heart, skeletal muscle, endocrine pancreas and capillary endothelium, and their differential expression could be related to obesity and the type 2 diabetes. Here we describe the expression and function of different aquaglyceroporins in physiological condition and in obesity and type 2 diabetes, suggesting they are potential therapeutic targets for metabolic disorders.


Subject(s)
Aquaglyceroporins/metabolism , Diabetes Mellitus, Type 2/metabolism , Glycerol/metabolism , Obesity/metabolism , Water/metabolism , Adipose Tissue/metabolism , Animals , Biological Transport, Active , Diabetes Mellitus, Type 2/pathology , Humans , Intestine, Small/metabolism , Kidney/metabolism , Liver/metabolism , Microvessels/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Obesity/pathology , Organ Specificity , Pancreas/metabolism
7.
Stem Cells Dev ; 25(4): 301-19, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26654173

ABSTRACT

Clonal endothelial progenitor cells (EPCs) have been implicated in the aberrant vascular growth that features infantile hemangioma (IH), the most common benign vascular tumor in childhood that may cause ulceration, bleeding, and/or permanent disfigurement. Endothelial colony-forming cells (ECFCs), truly endothelial EPCs endowed with clonal ability and capable of forming patent vessels in vivo, remodel their Ca(2+) toolkit in tumor-derived patients to acquire an adaptive advantage. Particularly, they upregulate the proangiogenic store-operated Ca(2+) entry (SOCE) pathway due to the overexpression of its underlying components, that is, stromal interaction molecule 1 (Stim1), Orai1, and transient receptor potential canonical 1 (TRPC1). The present work was undertaken to assess whether and how the Ca(2+) signalosome is altered in IH-ECFCs by employing Ca(2+) and nitric oxide (NO) imaging, real-time polymerase chain reaction, western blotting, and functional assays. IH-ECFCs display a lower intracellular Ca(2+) release in response to either pharmacological (i.e., cyclopiazonic acid) or physiological (i.e., ATP and vascular endothelial growth factor) stimulation. Conversely, Stim1, Orai1, and TRPC1 transcripts and proteins are normally expressed in these cells and mediate a constitutive SOCE, which is sensitive to BTP-2, La(3+), and Pyr6 and recharges the intracellular Ca(2+) pool. The resting SOCE in IH-ECFCs is also associated to an increase in their proliferation rate and the basal production of NO compared to normal cells. Likewise, the pharmacological blockade of SOCE and NO synthesis block IH-ECFC growth. Collectively, these data indicate that the constitutive SOCE activation enhances IH-ECFC proliferation by augmenting basal NO production and sheds novel light on the molecular mechanisms of IH.


Subject(s)
Calcium/metabolism , Colony-Forming Units Assay , Endothelial Cells/pathology , Endothelial Progenitor Cells/pathology , Hemangioma/pathology , Nitric Oxide/biosynthesis , Anilides/pharmacology , Cell Proliferation/drug effects , Child , Child, Preschool , Demography , Endothelial Cells/drug effects , Endothelial Progenitor Cells/drug effects , Female , Gene Expression Regulation/drug effects , Gentamicins/pharmacology , Humans , Indoles/pharmacology , Intracellular Space/metabolism , Lanthanum/pharmacology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thiadiazoles/pharmacology
8.
J Appl Toxicol ; 36(1): 113-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25854323

ABSTRACT

The main route of exposure to mercury in humans is through the diet. Consequently, the gastrointestinal mucosa is exposed to the mercurial forms, where they cause intestinal fluid accumulation, mucosal injuries and diarrhea. The relationship between inorganic mercury (HgCl2 ) and methylmercury (CH3 HgCl) exposure and water movement in the gastrointestinal tract is still unexplored. The leading role of aquaporins (AQPs) in the rapid bidirectional movement of fluid in the gastrointestinal tract of mammals is well established. The present study evaluates the effect of HgCl2 and CH3 HgCl exposure on AQP expression in different portions of the gastrointestinal tract of rats treated by gavage (5 mg kg(-1) of mercury species, single dose, 4 days). The results show that mercury species reduce mRNA and protein levels of AQPs in different parts of the gastrointestinal tract. In the stomach, treated rats show a significant reduction of expression of AQP3 (80-90% for mRNA and 50% for protein) and AQP4 (95-99% for mRNA and 20-40% for protein). In the small and large intestine, treated rats experience a significant reduction of AQP3 and AQP7 expression. Protein contents of both AQPs are reduced in similar proportions in jejunum (AQP3: 40-50%; AQP7: 45-50%) and colon (AQP3: 35-40%; AQP7: 45-60%), regardless of the treatment. Our results indicate that some AQPs are downregulated in the rat gastrointestinal tract by mercury exposure, suggesting a possible role of AQPs in the development of mercury gastrointestinal symptoms.


Subject(s)
Aquaporins/genetics , Gastrointestinal Tract/drug effects , Mercuric Chloride/toxicity , Methylmercury Compounds/toxicity , Animals , Aquaporins/analysis , Aquaporins/physiology , Gastrointestinal Tract/metabolism , Male , Rats , Rats, Wistar
9.
J Cell Physiol ; 230(1): 95-104, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24911002

ABSTRACT

Endothelial progenitor cells (EPCs) are mobilized into circulation to replace damaged endothelial cells and recapitulate the vascular network of injured tissues. Intracellular Ca(2+) signals are key to EPC activation, but it is yet to be elucidated whether they are endowed with the same blend of Ca(2+) -permeable channels expressed by mature endothelial cells. For instance, endothelial colony forming cells (ECFCs), the only EPC subset truly committed to acquire a mature endothelial phenotype, lack canonical transient receptor potential channels 3, 5 and 6 (TRPC3, 5 and 6), which are widely distributed in vascular endothelium; on the other hand, they express a functional store-operated Ca(2+) entry (SOCE). The present study was undertaken to assess whether human circulating EPCs possess TRP vanilloid channel 4 (TRPV4), which plays a master signalling role in mature endothelium, by controlling both vascular remodelling and arterial pressure. We found that EPCs express both TRPV4 mRNA and protein. Moreover, both GSK1016790A (GSK) and phorbol myristate acetate and, two widely employed TRPV4 agonists, induced intracellular Ca(2+) signals uniquely in presence of extracellular Ca(2+). GSK- and PMA-induced Ca(2+) elevations were inhibited by RN-1734 and ruthenium red, which selectively target TRPV4 in mature endothelium. However, TRPV4 stimulation with GSK did not cause EPC proliferation, while the pharmacological blockade of TRPV4 only modestly affected EPC growth in the presence of a growth factor-enriched culture medium. Conversely, SOCE inhibition with BTP-2, La(3+) and Gd(3+) dramatically decreased cell proliferation. These data indicate that human circulating EPCs possess a functional TRPV4 protein before their engraftment into nascent vessels.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Neovascularization, Physiologic/physiology , Stem Cells/metabolism , TRPV Cation Channels/biosynthesis , Adult , Anilides/pharmacology , Calcium/metabolism , Cation Transport Proteins/biosynthesis , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells/cytology , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , RNA, Messenger/biosynthesis , Ruthenium Red/pharmacology , Stem Cells/cytology , Sulfonamides/pharmacology , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Tetradecanoylphorbol Acetate/pharmacology , Thiadiazoles/pharmacology , Young Adult
10.
Biomed Res Int ; 2014: 739494, 2014.
Article in English | MEDLINE | ID: mdl-25126575

ABSTRACT

Store-operated Ca(2+) entry (SOCE) is activated following depletion of the inositol-1,4,5-trisphosphate (InsP3)-sensitive Ca(2+) pool to regulate proliferation in immortalized cell lines established from either primary or metastatic lesions. The molecular nature of SOCE may involve both Stim1, which senses Ca(2+) levels within the endoplasmic reticulum (ER) Ca(2+) reservoir, and a number of a Ca(2+)-permeable channels on the plasma membrane, including Orai1, Orai3, and members of the canonical transient receptor (TRPC1-7) family of ion channels. The present study was undertaken to assess whether SOCE is expressed and controls proliferation in primary cultures isolated from secondary lesions of heavily pretreated metastatic renal cell carcinoma (mRCC) patients. SOCE was induced following pharmacological depletion of the ER Ca(2+) store, but not by InsP3-dependent Ca(2+) release. Metastatic RCC cells express Stim1-2, Orai1-3, and TRPC1-7 transcripts and proteins. In these cells, SOCE was insensitive to BTP-2, 10 µM Gd(3+) and Pyr6, while it was inhibited by 100 µM Gd(3+), 2-APB, and carboxyamidotriazole (CAI). Neither Gd(3+) nor 2-APB or CAI impaired mRCC cell proliferation. Consistently, no detectable Ca(2+) signal was elicited by growth factor stimulation. Therefore, a functional SOCE is expressed but does not control proliferation of mRCC cells isolated from patients resistant to multikinase inhibitors.


Subject(s)
Calcium Signaling/genetics , Carcinoma, Renal Cell/metabolism , Cell Proliferation/genetics , Neoplasm Metastasis/genetics , Aged , Calcium Channels/biosynthesis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Female , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Male , Membrane Proteins/biosynthesis , Middle Aged , Neoplasm Metastasis/pathology , Neoplasm Proteins/biosynthesis , ORAI1 Protein , Primary Cell Culture , Protein Kinase Inhibitors/therapeutic use , Stromal Interaction Molecule 1 , TRPC Cation Channels
11.
PLoS One ; 9(3): e91099, 2014.
Article in English | MEDLINE | ID: mdl-24603752

ABSTRACT

BACKGROUND: An increase in the frequency of circulating endothelial colony forming cells (ECFCs), the only subset of endothelial progenitor cells (EPCs) truly belonging to the endothelial phenotype, occurs in patients affected by primary myelofibrosis (PMF). Herein, they might contribute to the enhanced neovascularisation of fibrotic bone marrow and spleen. Store-operated Ca2+ entry (SOCE) activated by the depletion of the inositol-1,4,5-trisphosphate (InsP3)-sensitive Ca2+ store drives proliferation in ECFCs isolated from both healthy donors (N-ECFCs) and subjects suffering from renal cellular carcinoma (RCC-ECFCs). SOCE is up-regulated in RCC-ECFCs due to the over-expression of its underlying molecular components, namely Stim1, Orai1, and TRPC1. METHODOLOGY/PRINCIPAL FINDINGS: We utilized Ca2+ imaging, real-time polymerase chain reaction, western blot analysis and functional assays to evaluate molecular structure and the functional role of SOCE in ECFCs derived from PMF patients (PMF-ECFCs). SOCE, induced by either pharmacological (i.e. cyclopiazonic acid or CPA) or physiological (i.e. ATP) stimulation, was significantly higher in PMF-ECFCs. ATP-induced SOCE was inhibited upon blockade of the phospholipase C/InsP3 signalling pathway with U73111 and 2-APB. The higher amplitude of SOCE was associated to the over-expression of the transcripts encoding for Stim2, Orai2-3, and TRPC1. Conversely, immunoblotting revealed that Stim2 levels remained constant as compared to N-ECFCs, while Stim1, Orai1, Orai3, TRPC1 and TRPC4 proteins were over-expressed in PMF-ECFCs. ATP-induced SOCE was inhibited by BTP-2 and low micromolar La3+ and Gd3+, while CPA-elicited SOCE was insensitive to Gd3+. Finally, BTP-2 and La3+ weakly blocked PMF-ECFC proliferation, while Gd3+ was ineffective. CONCLUSIONS: Two distinct signalling pathways mediate SOCE in PMF-ECFCs; one is activated by passive store depletion and is Gd3+-resistant, while the other one is regulated by the InsP3-sensitive Ca2+ pool and is inhibited by Gd3+. Unlike N- and RCC-ECFCs, the InsP3-dependent SOCE does not drive PMF-ECFC proliferation.


Subject(s)
Calcium Channels/metabolism , Endothelial Progenitor Cells/metabolism , Membrane Proteins/metabolism , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , TRPC Cation Channels/metabolism , Adenosine Triphosphate/pharmacology , Adult , Aged , Anilides/pharmacology , Calcium/metabolism , Calcium Channels/genetics , Cell Proliferation/drug effects , Cell Separation , Colony-Forming Units Assay , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endothelial Progenitor Cells/drug effects , Female , Gadolinium/pharmacology , Humans , Indoles/pharmacology , Inositol 1,4,5-Trisphosphate/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Lanthanum/pharmacology , Male , Membrane Potentials/drug effects , Membrane Proteins/genetics , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , TRPC Cation Channels/genetics , Thiadiazoles/pharmacology , Young Adult
12.
Curr Vasc Pharmacol ; 12(1): 87-105, 2014 Jan.
Article in English | MEDLINE | ID: mdl-22724469

ABSTRACT

Endothelial progenitor cells (EPCs) have recently been employed in cell-based therapy (CBT) to promote regeneration of ischemic organs, such as heart and limbs. Furthermore, EPCs may sustain tumour vascularisation and provide an additional target for anticancer therapies. CBT is limited by the paucity of cells harvested from peripheral blood and suffers from several pitfalls, including the low rate of engrafted EPCs, whereas classic antiangiogenic treatments manifest a number of side effects and may induce resistance into the patients. CBT will benefit of a better understanding of the signal transduction pathway(s) which drive(s) EPC proliferation, trafficking, and incorporation into injured tissues. At the same time, this information might outline alternative molecular targets to impair tumor neovascularisation and improve the therapeutic outcome of antiangiogenic strategies. An increase in intracellular Ca(2+) concentration is the key signal in the regulation of cellular replication, migration, and differentiation. In particular, Ca(2+) signalling may regulate cellcycle progression, due to the Ca(2+)-sensitivity of a number of cycline-dependent kinases, and gene expression, owing to the Ca(2+)-dependence of several transcription factors. Recent work has outlined the role of the so-called store-operated Ca(2+) entry in driving EPC proliferation and migration. Unravelling the mechanisms guiding EPC engraftment into neovessels might supply the biological bases required to improve CBT and anticancer treatments. For example, genetic manipulation of the Ca(2+) signalling machinery could provide a novel approach to increase the extent of limb regeneration or preventing tumour vascularisation by EPCs.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Endothelial Cells/metabolism , Neovascularization, Pathologic/prevention & control , Stem Cell Transplantation , Stem Cells/metabolism , Animals , Calcium Channels/metabolism , Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation , Humans , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , ORAI1 Protein , Stromal Interaction Molecule 1
13.
Stem Cells Dev ; 22(19): 2561-80, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23682725

ABSTRACT

Endothelial colony-forming cells (ECFCs) are the only endothelial progenitor cells (EPCs) that are capable of acquiring a mature endothelial phenotype. ECFCs are mainly mobilized from bone marrow to promote vascularization and represent a promising tool for cell-based therapy of severe ischemic diseases. Vascular endothelial growth factor (VEGF) stimulates the proliferation of peripheral blood-derived ECFCs (PB-ECFCs) through oscillations in intracellular Ca(2+) concentration ([Ca(2+)]i). VEGF-induced Ca(2+) spikes are driven by the interplay between inositol-1,4,5-trisphosphate (InsP3)-dependent Ca(2+) release and store-operated Ca(2+) entry (SOCE). The therapeutic potential of umbilical cord blood-derived ECFCs (UCB-ECFCs) has also been shown in recent studies. However, VEGF-induced proliferation of UCB-ECFCs is faster compared with their peripheral counterpart. Unlike PB-ECFCs, UCB-ECFCs express canonical transient receptor potential channel 3 (TRPC3) that mediates diacylglycerol-dependent Ca(2+) entry. The present study aimed at investigating whether the higher proliferative potential of UCB-ECFCs was associated to any difference in the molecular underpinnings of their Ca(2+) response to VEGF. We found that VEGF induces oscillations in [Ca(2+)]i that are patterned by the interaction between InsP3-dependent Ca(2+) release and SOCE. Unlike PB-ECFCs, VEGF-evoked Ca(2+) oscillations do not arise in the absence of extracellular Ca(2+) entry and after pharmacological (with Pyr3 and flufenamic acid) and genetic (by employing selective small interference RNA) suppression of TRPC3. VEGF-induced UCB-ECFC proliferation is abrogated on inhibition of the intracellular Ca(2+) spikes. Therefore, the Ca(2+) response to VEGF in UCB-ECFCs is shaped by a different Ca(2+) machinery as compared with PB-ECFCs, and TRPC3 stands out as a promising target in EPC-based treatment of ischemic pathologies.


Subject(s)
Calcium/metabolism , Ion Transport/physiology , Stem Cells/metabolism , TRPC Cation Channels/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adult , Anti-Inflammatory Agents/pharmacology , Cell Proliferation , Cells, Cultured , Endothelial Cells/metabolism , Female , Fetal Blood/cytology , Fetal Blood/metabolism , Flufenamic Acid/pharmacology , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Ion Transport/drug effects , Ion Transport/genetics , Middle Aged , Neovascularization, Physiologic , Pyrazoles/pharmacology , RNA Interference , RNA, Small Interfering , Signal Transduction/drug effects , TRPC Cation Channels/biosynthesis , TRPC Cation Channels/genetics , Young Adult
14.
PLoS One ; 7(9): e42541, 2012.
Article in English | MEDLINE | ID: mdl-23049731

ABSTRACT

BACKGROUND: Endothelial progenitor cells (EPCs) may be recruited from bone marrow to sustain tumor vascularisation and promote the metastatic switch. Understanding the molecular mechanisms driving EPC proliferation and tubulogenesis could outline novel targets for alternative anti-angiogenic treatments. Store-operated Ca(2+) entry (SOCE), which is activated by a depletion of the intracellular Ca(2+) pool, regulates the growth of human EPCs, where is mediated by the interaction between the endoplasmic reticulum Ca(2+)-sensor, Stim1, and the plasmalemmal Ca(2+) channel, Orai1. As oncogenesis may be associated to the capability of tumor cells to grow independently on Ca(2+) influx, it is important to assess whether SOCE regulates EPC-dependent angiogenesis also in tumor patients. METHODOLOGY/PRINCIPAL FINDINGS: The present study employed Ca(2+) imaging, recombinant sub-membranal and mitochondrial aequorin, real-time polymerase chain reaction, gene silencing techniques and western blot analysis to investigate the expression and the role of SOCE in EPCs isolated from peripheral blood of patients affected by renal cellular carcinoma (RCC; RCC-EPCs) as compared to control EPCs (N-EPCs). SOCE, activated by either pharmacological (i.e. cyclopiazonic acid) or physiological (i.e. ATP) stimulation, was significantly higher in RCC-EPCs and was selectively sensitive to BTP-2, and to the trivalent cations, La(3+) and Gd(3+). Furthermore, 2-APB enhanced thapsigargin-evoked SOCE at low concentrations, whereas higher doses caused SOCE inhibition. Conversely, the anti-angiogenic drug, carboxyamidotriazole (CAI), blocked both SOCE and the intracellular Ca(2+) release. SOCE was associated to the over-expression of Orai1, Stim1, and transient receptor potential channel 1 (TRPC1) at both mRNA and protein level The intracellular Ca(2+) buffer, BAPTA, BTP-2, and CAI inhibited RCC-EPC proliferation and tubulogenesis. The genetic suppression of Stim1, Orai1, and TRPC1 blocked CPA-evoked SOCE in RCC-EPCs. CONCLUSIONS: SOCE is remodelled in EPCs from RCC patients and stands out as a novel molecular target to interfere with RCC vascularisation due to its ability to control proliferation and tubulogenesis.


Subject(s)
Carcinoma, Renal Cell/blood supply , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/blood supply , Membrane Proteins/genetics , Neoplastic Stem Cells/metabolism , Adenosine Triphosphate/pharmacology , Adult , Aged , Aged, 80 and over , Boron Compounds/pharmacology , Cadmium/pharmacology , Calcium Channels/genetics , Calcium Channels/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Intracellular Calcium-Sensing Proteins , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Lanthanum/pharmacology , Male , Membrane Proteins/metabolism , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic , ORAI1 Protein , Primary Cell Culture , Signal Transduction/drug effects , Stromal Interaction Molecule 1 , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
15.
Stem Cells ; 29(11): 1898-907, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21905169

ABSTRACT

Endothelial progenitor cells (EPCs) home from the bone marrow to the site of tissue regeneration and sustain neovascularization after acute vascular injury and upon the angiogenic switch in solid tumors. Therefore, they represent a suitable tool for cell-based therapy (CBT) in regenerative medicine and provide a novel promising target in the fight against cancer. Intracellular Ca(2+) signals regulate numerous endothelial functions, such as proliferation and tubulogenesis. The growth of endothelial colony forming cells (ECFCs), which are EPCs capable of acquiring a mature endothelial phenotype, is governed by store-dependent Ca(2+) entry (SOCE). This study aimed at investigating the nature and the role of VEGF-elicited Ca(2+) signals in ECFCs. VEGF induced asynchronous Ca(2+) oscillations, whose latency, amplitude, and frequency were correlated to the growth factor dose. Removal of external Ca(2+) (0Ca(2+)) and SOCE inhibition with N-(4-[3,5-bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl)-4-methyl-1,2,3-thiadiazole-5-carboxamide (BTP-2) reduced the duration of the oscillatory signal. Blockade of phospholipase C-γ with U73122, emptying the inositol-1,4,5-trisphosphate (InsP(3))-sensitive Ca(2+) pools with cyclopiazonic acid (CPA), and inhibition of InsP(3) receptors with 2-APB prevented the Ca(2+) response to VEGF. VEGF-induced ECFC proliferation and tubulogenesis were inhibited by the Ca(2+)-chelant, BAPTA, and BTP-2. NF-κB activation by VEGF was impaired by BAPTA, BTP-2, and its selective blocker, thymoquinone. Thymoquinone, in turn, suppressed VEGF-dependent ECFC proliferation and tubulogenesis. These data indicate that VEGF-induced Ca(2+) oscillations require the interplay between InsP(3)-dependent Ca(2+) release and SOCE, and promote ECFC growth and tubulogenesis by engaging NF-κB. This novel signaling pathway might be exploited to enhance the outcome of CBT and chemotherapy.


Subject(s)
Calcium/metabolism , Endothelial Cells/cytology , Stem Cells/cytology , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Adult , Anilides/pharmacology , Benzoquinones/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors , Humans , Immunoblotting , Indoles/pharmacology , NF-kappa B/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Thiadiazoles/pharmacology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...