Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
2.
Exp Mol Med ; 2024 May 01.
Article in English | MEDLINE | ID: mdl-38689092

ABSTRACT

Hepatocellular carcinoma (HCC) is associated with a poor prognosis. Our previous study demonstrated that Pleomorphic adenoma gene like-2 (PLAGL2) was a potential therapeutic target in HCC. However, the mechanisms that lead to the upregulation of PLAGL2 in HCC remain unclear. The present study revealed that stress-induced epinephrine increased the expression of PLAGL2, thereby promoting the progression of HCC. Furthermore, PLAGL2 knockdown inhibited epinephrine-induced HCC development. Mechanistically, epinephrine upregulated ubiquitin-specific protease 10 (USP10) to stabilize PLAGL2 via the adrenergic ß-receptor-2-c-Myc (ADRB2-c-Myc) axis. Furthermore, PLAGL2 acted as a transcriptional regulator of USP10, forming a signaling loop. Taken together, these results reveal that stress-induced epinephrine activates the PLAGL2-USP10 signaling loop to enhance HCC progression. Furthermore, PLAGL2 plays a crucial role in psychological stress-mediated promotion of HCC progression.

3.
Adv Healthc Mater ; 11(3): e2101761, 2022 02.
Article in English | MEDLINE | ID: mdl-34811972

ABSTRACT

Circulating tumor cells (CTCs) are reported as the precursor of tumor metastases, implying that stifling CTCs would be beneficial for metastasis prevention. However, challenges remain for the application of therapies that aim at CTCs due to lack of effective CTC-targeting strategy and sensitive therapeutic agents. Herein, a general CTC-intervention strategy based on neutrophil cyto-pharmaceuticals is proposed for suppressing CTC colonization and metastasis formation. Breast cancer 4T1 cells are infused as the mimic CTCs, and 4T1 cells trapped are first elucidated in neutrophil extracellular traps (NETs) expressing high levels of hypoxia-inducible factor-1α (HIF-1α) due to NET formation and thus promoting tumor cell colonization through enhanced migration, invasion and stemness. After verifying HIF-1α as a potential target for metastasis prevention, living neutrophil cyto-pharmaceuticals (CytPNEs) loaded with HIF-1α inhibitor are fabricated to therapeutically inhibit HIF-1α. It is demonstrated that CytPNEs can specially convey the HIF-1α inhibitor to 4T1 cells according to the inflammatory chemotaxis of neutrophils and down-regulate HIF-1α, thereby inhibiting metastasis and prolonging the median survival of mice bearing breast cancer lung metastasis. The research offers a new perspective for understanding the mechanism of CTC colonization, and puts forward the strategy of targeted intervention of CTCs as a meaningful treatment for tumor metastasis.


Subject(s)
Breast Neoplasms , Neoplastic Cells, Circulating , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Neoplasm Metastasis/prevention & control , Neutrophils , Pharmaceutical Preparations
4.
Med Res Rev ; 41(1): 156-201, 2021 01.
Article in English | MEDLINE | ID: mdl-32844499

ABSTRACT

Immunotherapy has revolutionized the treatment of cancer in recent years and achieved overall success and long-term clinical benefit in patients with a wide variety of cancer types. However, there is still a large proportion of patients exhibiting limited or no responses to immunotherapeutic strategy, some of which were even observed with hyperprogressive disease. One major obstacle restricting the efficacy is that tumor-reactive CD8+ T cells, which are central for tumor control, undergo exhaustion, and lose their ability to eliminate cancer cells after infiltrating into the strongly immunosuppressive tumor microenvironment. Thus, as a potential therapeutic rationale in the development of cancer immunotherapy, targeting or reinvigorating exhausted CD8+ T cells has been attracting much interest. Hitherto, both intrinsic and extrinsic mechanisms that govern CD8+ T-cell exhaustion have been explored. Specifically, the transcriptional and epigenetic landscapes have been depicted utilizing single-cell RNA sequencing or mass cytometry (CyTOF). In addition, cellular metabolism dictating the tumor-infiltrating CD8+ T-cell fate is currently under investigation. A series of clinical trials are being carried out to further establish the current strategies targeting CD8+ T-cell exhaustion. Taken together, despite the proven benefit of immunotherapy in cancer patients, additional efforts are still needed to fully circumvent limitations of exhausted T cells in the treatment. In this review, we will focus on the current cellular and molecular understanding of metabolic changes, epigenetic remodeling, and transcriptional regulation in CD8+ T-cell exhaustion and describe hypothetical treatment approaches based on immunotherapy aiming at reinvigorating exhausted CD8+ T cells.


Subject(s)
Neoplasms , Tumor Microenvironment , CD8-Positive T-Lymphocytes , Humans , Immunotherapy , Neoplasms/therapy , T-Lymphocytes, Cytotoxic
5.
Hepatology ; 73(2): 674-691, 2021 02.
Article in English | MEDLINE | ID: mdl-32335942

ABSTRACT

BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide, hence a major public health threat. Pleomorphic adenoma gene like-2 (PLAGL2) has been reported to play a role in tumorigenesis. However, its precise function in HCC remains poorly understood. APPROACH AND RESULTS: In this study, we demonstrated that PLAGL2 was up-regulated in HCC compared with that of adjacent nontumorous tissues and also correlated with overall survival times. We further showed that PLAGL2 promoted HCC cell proliferation, migration, and invasion both in vitro and in vivo. PLAGL2 expression was positively correlated with epidermal growth factor receptor (EGFR) expression. Mechanistically, this study demonstrated that PLAGL2 functions as a transcriptional regulator of EGFR and promotes HCC cell proliferation, migration, and invasion through the EGFR-AKT pathway. Moreover, hypoxia was found to significantly induce high expression of PLAGL2, which promoted hypoxia inducible factor 1/2 alpha subunit (HIF1/2A) expression through EGFR. Therefore, this study demonstrated that a PLAGL2-EGFR-HIF1/2A signaling loop promotes HCC progression. More importantly, PLAGL2 expression reduced hepatoma cells' response to the anti-EGFR drug erlotinib. PLAGL2 knockdown enhanced the response to erlotinib. CONCLUSIONS: This study reveals the pivotal role of PLAGL2 in HCC cell proliferation, metastasis, and erlotinib insensitivity. This suggests that PLAGL2 can be a potential therapeutic target of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA-Binding Proteins/metabolism , Erlotinib Hydrochloride/pharmacology , Liver Neoplasms/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Adult , Aged , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Disease Progression , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride/therapeutic use , Feedback, Physiological , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kaplan-Meier Estimate , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Mice , Middle Aged , Neoplasm Invasiveness/genetics , RNA-Binding Proteins/genetics , RNA-Seq , Signal Transduction/genetics , Transcription Factors/genetics , Tumor Hypoxia , Up-Regulation , Xenograft Model Antitumor Assays
6.
Mol Cancer ; 19(1): 13, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31973707

ABSTRACT

BACKGROUND: As a novel class of noncoding RNAs, circRNAs have been recently identified to regulate tumorigenesis and aggressiveness. However, the function of circRNAs in colorectal cancer (CRC) metastasis remains unclear. We aimed to identify circRNAs that are upregulated in CRC tissues from patients and study their function in CRC metastasis. METHODS: We compared six pairs of CRC tissues and their matched adjacent non-tumor tissues by using circRNA microarray. We first evaluated the expression of circPTK2 (hsa_circ_0005273) in fresh tissues from CRC tumors and corresponding adjacent tissues by qPCR analysis. CircPTK2 expression levels in the tissue microarray with 5 years of survival information were determined by RNA-ISH analysis. Meanwhile, the expression levels of circulating circPTK2 were further analyzed according to the patients' clinical features. We analyzed cell apoptosis, colony formation, migration, and invasion in CRC cells. To further elucidate the effect of circPTK2 in CRC metastasis, we also conducted a colon cancer hepatic and pulmonary metastasis experiment. We used RNA biotin-labeled pull down and mass spectrometry to identify the target of circPTK2. We established a PDTX model to evaluate the effect of shRNA specifically targeting circPTK2 on tumor metastasis. RESULTS: We identified a novel circRNA, circPTK2, which is back-spliced of three exons (exons 27, 28 and 29) of PTK2 by using circRNA microarray, bioinformatics and functional studies. CircPTK2 was elevated in CRC tissues and positively associated with tumor growth and metastasis. CRC patients with increased circPTK2 expression were positively correlated with poorer survival rates. Furthermore, our studies showed that circPTK2 could promote EMT of CRC cells in vitro and in vivo by binding to vimentin protein on sites Ser38, Ser55 and Ser82. We further demonstrated the interaction of circPTK2 and vimentin mediated the regulation of CRC by knockdown or overexpression of vimentin. In addition, we revealed that tail vein injection of shRNA specifically targeting circPTK2 blunt tumor metastasis in a patient-derived CRC xenograft model. CONCLUSIONS: Collectively, these results demonstrate that circPTK2 exerts critical roles in CRC growth and metastasis and may serve as a potential therapeutic target for CRC metastasis, and also a promising biomarker for early diagnosis of metastasis.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , Focal Adhesion Kinase 1/genetics , Lung Neoplasms/secondary , Animals , Apoptosis , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , RNA, Circular/genetics , RNA, Circular/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Sci Rep ; 6: 30925, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27499046

ABSTRACT

Putative functions of the heterotrimeric G-protein subunit Gαi2-dependent signaling include ion channel regulation, cell differentiation, proliferation and apoptosis. Erythrocytes may, similar to apoptosis of nucleated cells, undergo eryptosis, characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine (PS) exposure. Eryptosis may be triggered by increased cytosolic Ca(2+) activity and ceramide. In the present study, we show that Gαi2 is expressed in both murine and human erythrocytes and further examined the survival of erythrocytes drawn from Gαi2-deficient mice (Gαi2(-/-)) and corresponding wild-type mice (Gαi2(+/+)). Our data show that plasma erythropoietin levels, erythrocyte maturation markers, erythrocyte counts, hematocrit and hemoglobin concentration were similar in Gαi2(-/-) and Gαi2(+/+) mice but the mean corpuscular volume was significantly larger in Gαi2(-/-) mice. Spontaneous PS exposure of circulating Gαi2(-/-) erythrocytes was significantly lower than that of circulating Gαi2(+/+) erythrocytes. PS exposure was significantly lower in Gαi2(-/-) than in Gαi2(+/+) erythrocytes following ex vivo exposure to hyperosmotic shock, bacterial sphingomyelinase or C6 ceramide. Erythrocyte Gαi2 deficiency further attenuated hyperosmotic shock-induced increase of cytosolic Ca(2+) activity and cell shrinkage. Moreover, Gαi2(-/-) erythrocytes were more resistant to osmosensitive hemolysis as compared to Gαi2(+/+) erythrocytes. In conclusion, Gαi2 deficiency in erythrocytes confers partial protection against suicidal cell death.


Subject(s)
Eryptosis , Erythrocytes/cytology , Erythrocytes/physiology , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Animals , Cell Survival , Erythrocyte Indices , Erythrocytes/chemistry , GTP-Binding Protein alpha Subunit, Gi2/deficiency , Humans , Mice , Mice, Knockout , Phosphatidylserines/analysis
8.
Cell Physiol Biochem ; 38(5): 1695-702, 2016.
Article in English | MEDLINE | ID: mdl-27160671

ABSTRACT

BACKGROUND/AIMS: The polyamine oxidase inhibitor MDL-72527 (N1,N4-bis(2,3-butadienyl)-1,4-butanediamine) were expected to increase the abundance of spermine, a powerful inhibitor of platelet activation. Nothing is known, however, on the sensitivity of platelet function and survival to MDL-72527 exposure. The present study thus explored whether MDL-72527 modifies function and survival of platelets without and with platelet activation by collagen related peptide (CRP). METHODS: Platelets isolated from wild-type mice were exposed for 30 minutes to MDL-72527 (100 µM) with or without subsequent activation with CRP (2-5 µg/ml). Flow cytometry was employed to estimate cytosolic Ca2+-activity ([Ca2+]i) from Fluo-3 fluorescence, platelet degranulation from P-selectin abundance, integrin activation from αIIbß3 integrin abundance, generation of reactive oxygen species (ROS) from DCFDA fluorescence, phospholipid scrambling of the cell membrane from annexin-V-binding, platelet volume from forward scatter and aggregation utilizing staining with CD9-APC and CD9-PE. RESULTS: In the absence of CRP, exposure of platelets to MDL-72527 did not significantly modify [Ca2+]i, P-selectin abundance, αIIbß3 integrin abundance, ROS, annexin-V-binding, and forward scatter. The addition of 2-5 µg/ml CRP was followed by significant increase of [Ca2+]i, P-selectin abundance, αIIbß3 integrin activation, ROS abundance, annexin-V-binding, and aggregation as well as a significant decrease of forward scatter, all effects significantly blunted or virtually abolished in the presence of MDL-72527. CONCLUSIONS: MDL-72527 is a powerful inhibitor of platelet activation, apoptosis and aggregation.


Subject(s)
Platelet Activation/drug effects , Putrescine/analogs & derivatives , Aniline Compounds/chemistry , Animals , Apoptosis/drug effects , Blood Platelets/cytology , Blood Platelets/metabolism , Calcium/chemistry , Calcium/metabolism , Carrier Proteins/pharmacology , Cell Membrane/metabolism , Female , Flow Cytometry , Male , Mice , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-NH Group Donors/metabolism , P-Selectin/metabolism , Peptides/pharmacology , Phosphatidylserines , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Putrescine/pharmacology , Reactive Oxygen Species/metabolism , Xanthenes/chemistry , Polyamine Oxidase
9.
Cell Physiol Biochem ; 38(6): 2272-84, 2016.
Article in English | MEDLINE | ID: mdl-27197532

ABSTRACT

BACKGROUND/AIMS: The novel antifungal drug Anidulafungin is used for the treatment of diverse fungal infections including candidiasis and aspergillosis. The traditional antifungal drug amphotericin B has previously been shown to trigger eryptosis, the suicidal death of erythrocytes characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Triggers of eryptosis include increase of cytosolic Ca2+ activity ([Ca2+]i), oxidative stress, ceramide, activated protein kinase C (PKC), casein kinase 1α or p38 kinase and activated caspases. Inhibitors of eryptosis include nitric oxide (NO). The present study explored, whether Anidulafungin induces eryptosis. METHODS: Flow cytometry was employed to estimate phosphatidylserine abundance at the erythrocyte surface from annexin-V-binding, cell volume from forward scatter, [Ca2+]i from Fluo3-fluorescence, abundance of reactive oxygen species (ROS) from DCFDA dependent fluorescence, and ceramide abundance at the erythrocyte surface utilizing specific antibodies. Hemolysis was quantified by measuring haemoglobin concentration in the supernatant. RESULTS: A 48 hours exposure of human erythrocytes to Anidulafungin (1.5 - 6 µg/ml) significantly increased hemolysis and the percentage of annexin-V-binding cells, and significantly decreased forward scatter. Anidulafungin (6 µg/ml) slightly, but significantly inceased Fluo3-fluorescence and the effect of Anidulafungin on annexin-V-binding was slightly, but significantly blunted by removal of extracellular Ca2+. The effect of Anidulafungin on annexin-V-binding was further significantly blunted by the p38 kinase inhibitor SB203580 (2 µM) and NO donor nitroprusside (1 µM). An increase of extracellular K+ concentration significantly blunted the effect of Anidulafungin on cell volume but not on annexin-V-binding. Anidulafungin rather decreased DCFDA fluorescence and the effect of Anidulafungin on annexin-V-binding was not significantly blunted by the antioxidant N-acetylcysteine (1 mM). Moreover, the effect of Anidulafungin on annexin-V-binding was not paralleled by significant increase of ceramide abundance and was not significantly blunted by PKC inhibitor staurosporine (1 µM), casein kinase 1α inhibitor D4476 (10 µM) or pancaspase inhibitor zVAD (10 µM). CONCLUSIONS: Anidulafungin triggers hemolysis and eryptosis with cell shrinkage and phospholipid scrambling of the erythrocyte cell membrane, an effect in part due to Ca2+ entry and activation of p38 kinase.


Subject(s)
Antifungal Agents/adverse effects , Echinocandins/adverse effects , Eryptosis/drug effects , Erythrocytes/drug effects , Hemolysis/drug effects , Anidulafungin , Calcium/metabolism , Cell Size/drug effects , Erythrocytes/cytology , Erythrocytes/metabolism , Erythrocytes/pathology , Humans , Reactive Oxygen Species/metabolism
10.
Cell Physiol Biochem ; 38(2): 726-36, 2016.
Article in English | MEDLINE | ID: mdl-26871421

ABSTRACT

BACKGROUND/AIMS: DAPT (24-diamino-5-phenylthiazole) inhibits γ-secretase, which cleaves the signaling molecule CD44, a negative regulator of platelet activation and apoptosis. CD44 is a co-receptor for macrophage migration inhibitory factor (MIF) an anti-apoptotic pro-inflammatory cytokine expressed and released from blood platelets. Whether DAPT influences platelet function, remained, however, elusive. Activators of platelets include collagen related peptide (CRP). The present study thus explored whether DAPT modifies the stimulating effect of CRP on platelet function. METHODS: Platelets isolated from wild-type mice were exposed for 30 minutes to DAPT (10 µM). Flow cytometry was employed to estimate Orai1 abundance with specific antibodies, cytosolic Ca(2+)-activity ([Ca2+]i) from Fluo-3 fluorescence, platelet degranulation from P-selectin abundance, integrin activation from α(IIb)ß3 integrin abundance, generation of reactive oxygen species (ROS) from DCFDA fluorescence, mitochondrial transmembrane potential from TMRE fluorescence, phospholipid scrambling of the cell membrane from annexin-V-binding, relative platelet volume from forward scatter and aggregation utilizing staining with CD9-APC and CD9-PE. RESULTS: Exposure of platelets to 2-5 µg/ml CRP was followed by significant increase of Orai1 abundance, [Ca2+]i, and P-selectin abundance, as well as by α(IIb)ß3 integrin activation, ROS generation, mitochondrial depolarization, enhanced annexin-V-binding, decreased cell volume, and aggregation. All CRP induced effects were significantly blunted in the presence of DAPT. CONCLUSIONS: The γ-secretase inhibitor DAPT counteracts agonist induced platelet activation, apoptosis and aggregation.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Blood Platelets/drug effects , Diamines/pharmacology , Enzyme Inhibitors/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Thiazoles/pharmacology , Amyloid Precursor Protein Secretases/immunology , Animals , Blood Platelets/immunology , Carrier Proteins/immunology , Female , Male , Mice , Peptides/immunology , Platelet Activation/immunology
11.
Thromb Haemost ; 115(1): 99-108, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26355696

ABSTRACT

CD44 is required for signalling of macrophage migration inhibitory factor (MIF), an anti-apoptotic pro-inflammatory cytokine. MIF is expressed and released from blood platelets, key players in the orchestration of occlusive vascular disease. Nothing is known about a role of CD44 in the regulation of platelet function. The present study thus explored whether CD44 modifies degranulation (P-selectin exposure), integrin activation, caspase activity, phosphatidylserine exposure on the platelet surface, platelet volume, Orai1 protein abundance and cytosolic Ca(2+)-activity ([Ca2+]i). Platelets from mice lacking CD44 (cd44(-/-)) were compared to platelets from corresponding wild-type mice (cd44(+/+)). In resting platelets, P-selectin abundance, α(IIb)ß3 integrin activation, caspase-3 activity and phosphatidylserine exposure were negligible in both genotypes and Orai1 protein abundance, [Ca2+]i, and volume were similar in cd44(-/-) and cd44(+/+) platelets. Platelet degranulation and α(IIb)ß3 integrin activation were significantly increased by thrombin (0.02 U/ml), collagen related peptide (CRP, 2 µg/ml and Ca(2+)-store depletion with thapsigargin (1 µM), effects more pronounced in cd44(-/-) than in cd44(+/+) platelets. Thrombin (0.02 U/ml) increased platelet [Ca2+]i, caspase-3 activity, phosphatidylserine exposure and Orai1 surface abundance, effects again significantly stronger in cd44(-/-) than in cd44(+/+) platelets. Thrombin further decreased forward scatter in cd44(-/-) and cd44(+/+) platelets, an effect which tended to be again more pronounced in cd44(-/-) than in cd44(+/+) platelets. Platelet adhesion and in vitro thrombus formation under high arterial shear rates (1,700 s(-1)) were significantly augmented in cd44(-/-) mice. In conclusion, genetic deficiency of CD44 augments activation, apoptosis and pro-thrombotic potential of platelets.


Subject(s)
Blood Platelets/metabolism , Cell Membrane/metabolism , Hyaluronan Receptors/metabolism , Mechanotransduction, Cellular , Phospholipids/metabolism , Platelet Adhesiveness , Thrombosis/metabolism , Animals , Apoptosis , Blood Coagulation , Calcium Channels/metabolism , Calcium Signaling , Caspase 3/metabolism , Cell Degranulation , Chlorides , Disease Models, Animal , Female , Ferric Compounds , Genotype , Hyaluronan Receptors/blood , Hyaluronan Receptors/genetics , Male , Mice, Knockout , ORAI1 Protein , Phenotype , Phospholipids/blood , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Regional Blood Flow , Selenoprotein P/metabolism , Stress, Mechanical , Thrombin/metabolism , Thrombosis/blood , Thrombosis/genetics
12.
BMC Cancer ; 15: 995, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26690689

ABSTRACT

BACKGROUND: Membrane androgen receptors (mAR) are functionally expressed in a variety of tumor-cells including the breast tumor-cell line MCF-7. They are specifically activated by testosterone albumin conjugates (TAC). The mAR sensitive signaling includes activation of Ras-related C3 botulinum toxin substrate 1 (Rac1) and reorganization of the actin filament network. Signaling of tumor-cells may further involve up-regulation of pore forming Ca(2+) channel protein Orai1, which accomplishes store operated Ca(2+) entry (SOCE). This study explored the regulation of Orai1 abundance and SOCE by mAR. METHODS: Actin filaments were visualized utilizing confocal microscopy, Rac1 activity using GST-GBD assay, Orai1 transcript levels by RT-PCR and total protein abundance by western blotting, Orai1 abundance at the cell surface by confocal microscopy and FACS-analysis, cytosolic Ca(2+) activity ([Ca(2+)]i) utilizing Fura-2-fluorescence, and SOCE from increase of [Ca(2+)]i following readdition of Ca(2+) after store depletion with thapsigargin (1 µM). RESULTS: TAC treatment of MCF-7 cells was followed by Rac1 activation, actin polymerization, transient increase of Orai1transcript levels and protein abundance, and transient increase of SOCE. The transient increase of Orai1 protein abundance was abrogated by Rac1 inhibitor NSC23766 (50 µM) and by prevention of actin reorganization with cytochalasin B (1 µM). CONCLUSIONS: mAR sensitive Rac1 activation and actin reorganization contribute to the regulation of Orai1 protein abundance and SOCE.


Subject(s)
Breast Neoplasms/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Receptors, Androgen/physiology , Actin Cytoskeleton/physiology , Blotting, Western , Breast Neoplasms/physiopathology , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Female , Humans , MCF-7 Cells , ORAI1 Protein , Real-Time Polymerase Chain Reaction , Thapsigargin/pharmacology , rac1 GTP-Binding Protein/metabolism
13.
Cell Physiol Biochem ; 37(5): 1759-66, 2015.
Article in English | MEDLINE | ID: mdl-26584277

ABSTRACT

BACKGROUND: Blood platelets accomplish primary hemostasis following vascular injury and contribute to the orchestration of occlusive vascular disease. Platelets are activated by an increase of cytosolic Ca2+-activity ([Ca2+]i), which is accomplished by Ca2+-release from intracellular stores and subsequent store operated Ca2+ entry (SOCE) through Ca2+ release activated Ca2+ channel moiety Orai1. Powerful activators of platelets include thrombin and collagen related peptide (CRP), which are in part effective by activation of small G- protein Rac1. The present study explored the influence of thrombin and CRP on Orai1 protein abundance and cytosolic Ca2+-activity ([Ca2+]i) in platelets drawn from wild type mice. METHODS: Orai1 protein surface abundance was quantified utilizing CF™488A conjugated antibodies, and [Ca2+]i was determined with Fluo3-fluorescence. RESULTS: In resting platelets, Orai1 protein abundance and [Ca2+]i were low. Thrombin (0.02 U/ml) and CRP (5ug/ml) within 2 min increased [Ca2+]i and Orai1 protein abundance at the platelet surface. [Ca2+]i was further increased by Ca2+ ionophore ionomycin (1 µM) and by store depletion with the sarcoendoplasmatic Ca2+ ATPase inhibitor thapsigargin (1 µM). However, Orai1 protein abundance at the platelet surface was not significantly affected by ionomycin and only slightly increased by thapsigargin. The effect of thrombin and CRP on Orai1 abundance and [Ca2+]i was significantly blunted by Rac1 inhibitor NSC23766 (50 µM). CONCLUSION: The increase of [Ca2+]i following stimulation of platelets with thrombin and collagen related peptide is potentiated by ultrarapid Rac1 sensitive translocation of Orai1 into the cell membrane.


Subject(s)
Blood Platelets/drug effects , Calcium Channels/metabolism , Collagen/chemistry , Peptides/pharmacology , Thrombin/pharmacology , Up-Regulation/drug effects , Aminoquinolines/pharmacology , Animals , Blood Platelets/cytology , Blood Platelets/metabolism , Calcium/metabolism , Calcium Channels/genetics , Cell Membrane/metabolism , Ionomycin/pharmacology , Ions/chemistry , Ions/metabolism , Mice , Mice, Inbred C57BL , Neuropeptides/antagonists & inhibitors , Neuropeptides/genetics , Neuropeptides/metabolism , ORAI1 Protein , Peptides/chemistry , Pyrimidines/pharmacology , Thapsigargin/pharmacology , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
14.
Cell Physiol Biochem ; 37(5): 1934-44, 2015.
Article in English | MEDLINE | ID: mdl-26584292

ABSTRACT

BACKGROUND/AIMS: The ubiquitously expressed Ca2+ Activated Cl- Channel Ano6 participates in the stimulation of cell membrane scrambling. Defective Ano6 underlies the Scott syndrome, an inherited bleeding disorder with impaired scrambling of plasma membrane phospholipids. At least in theory, the bleeding disorder of Scott syndrome may result from impaired platelet function. Activators of platelets include thrombin and collagen related peptide (CRP), which trigger increase of cytosolic Ca2+-activity ([Ca2+]i), production of reactive oxygen species (ROS), degranulation, integrin activation, as well as cell shrinkage and phospholipid scrambling of the cell membrane. The present study thus explored whether Ano6 modifies activation-induced alterations of cytosolic Ca2+-activity ([Ca2+]i), degranulation (P-selectin exposure), integrin activation, phosphatidylserine exposure on the platelet surface and platelet volume. METHODS: Platelets from mice lacking Ano6 (ano6-/-) were compared to platelets from corresponding wild-type mice (ano6+/+). [Ca2+]i was estimated from Fluo-3 fluorescence, ROS from DCFDA fluorescence, degranulation from P-selectin abundance, integrin activation from αIIbß3-integrin abundance, phosphatidylserine abundance from annexin-V-binding, and cell volume from forward scatter. RESULTS: Platelet number in blood was slightly higher in ano6-/- mice than in ano6+/+ mice. Without activation [Ca2+]i and volume were similar in ano6-/- and ano6+/+ platelets as well as ROS abundance, P-selectin abundance, αIIbß3 integrin activation, and phosphatidylserine exposure were negligible in both genotypes. Thrombin (0.01 U/ml) and CRP (2 or 5 µg/ml) increased [Ca2+]i, ROS abundance, platelet degranulation, αIIbß3 integrin activation, and triggered annexin-V-binding as well as cell shrinkage, all effects less pronounced in ano6-/- than in ano6+/+ platelets. CONCLUSIONS: Genetic knockout of Ano6 blunts thrombin- and CRP-induced activation and apoptosis of blood platelets.


Subject(s)
Apoptosis , Blood Platelets/metabolism , Platelet Activation , Aniline Compounds/chemistry , Animals , Anoctamins , Apoptosis/drug effects , Blood Platelets/cytology , C-Reactive Protein/pharmacology , Calcium/analysis , Mice , Mice, Knockout , P-Selectin/metabolism , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/deficiency , Phospholipid Transfer Proteins/genetics , Platelet Activation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Thrombin/pharmacology , Xanthenes/chemistry
15.
Cell Physiol Biochem ; 36(4): 1395-405, 2015.
Article in English | MEDLINE | ID: mdl-26159705

ABSTRACT

BACKGROUND: The antimalarial drug mefloquine has previously been shown to stimulate apoptosis of nucleated cells. Similar to apoptosis, erythrocytes may enter suicidal death or eryptosis, which is characterized by cell shrinkage and phospholipid scrambling of the erythrocyte cell membrane with phosphatidylserine translocation to the erythrocyte surface. Stimulators of eryptosis include oxidative stress, increase of cytosolic Ca2+-activity ([Ca2+]i), and ceramide. METHODS: Phosphatidylserine abundance at the cell surface was estimated from annexin V binding, cell volume from forward scatter, reactive oxidant species (ROS) from 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence, [Ca2+]i from Fluo3- fluorescence, and ceramide abundance from specific antibody binding. RESULTS: A 48 h treatment of human erythrocytes with mefloquine significantly increased the percentage of annexin-V-binding cells (≥5 µg/ml), significantly decreased forward scatter (≥5 µg/ml), significantly increased ROS abundance (5 µg/ml), significantly increased [Ca2+]i (7.5 µg/ml) and significantly increased ceramide abundance (10 µg/ml). The up-regulation of annexin- V-binding following mefloquine treatment was significantly blunted but not abolished by removal of extracellular Ca2+. Even in the absence of extracellular Ca2+, mefloquine significantly increased annexin-V-binding. CONCLUSIONS: Mefloquine treatment leads to erythrocyte shrinkage and erythrocyte membrane scrambling, effects at least partially due to induction of oxidative stress, increase of [Ca2+]i and up-regulation of ceramide abundance.


Subject(s)
Antimalarials/pharmacology , Apoptosis/drug effects , Erythrocytes/drug effects , Mefloquine/pharmacology , Calcium/metabolism , Cell Size/drug effects , Ceramides/metabolism , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Humans , Malaria/drug therapy , Malaria/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
16.
Cell Physiol Biochem ; 36(2): 773-83, 2015.
Article in English | MEDLINE | ID: mdl-26021265

ABSTRACT

BACKGROUND/AIMS: Anemia, a common condition in the elderly, could result from impaired formation and/or from accelerated loss of circulating erythrocytes. The latter could result from premature suicidal erythrocyte death or eryptosis characterized by phosphatidylserine (PS) exposure at the erythrocyte surface. Triggers of eryptosis include increased cytosolic Ca(2+)-concentration ([Ca(2+)]i), oxidative stress and ceramide. The present study explored whether eryptosis is altered in elderly individuals and, if so, to identify underlying mechanisms. METHODS: Blood was drawn from healthy young (n=11, age 31.3 ± 1.7 years) and elderly (n=16, age 88.6 ± 0.9 years) individuals. PS exposure was estimated from annexin V-binding, cell volume from forward scatter, [Ca(2+)]i from Fluo3-fluorescence, reactive oxygen species (ROS) from 2',7'dichlorodihydrofluorescein fluorescence, reduced glutathione (GSH) from mercury orange fluorescence and ceramide from FITC-conjugated antibody binding in flow cytometry. Measurements were made in erythrocytes from freshly drawn blood and in erythrocytes exposed in vitro for 24 h to plasma from young or elderly individuals. RESULTS: Elderly individuals suffered from severe anemia (hemoglobin 10.5 ± 0.3 g/100 ml) despite enhanced number of reticulocytes (2.3 ± 0.2%). The percentage of PS-exposing erythrocytes was significantly higher in the elderly (2.5 ± 0.2%) than in the young volunteers (1.3 ± 0.1%). The increase in PS exposure was paralleled by significant increase of ROS and significantly decreased levels of reduced GSH. Erythrocyte [Ca(2+)]i, and ceramide abundance tended to be higher in the elderly, differences, however, not reaching statistical significance. CONCLUSIONS: The anemia of elderly individuals is mainly if not exclusively due to enhanced eryptosis, resulting at least in part from GSH deficiency and increased oxidative stress.


Subject(s)
Aging , Anemia/blood , Anemia/etiology , Erythrocytes/pathology , Adult , Aged, 80 and over , Anemia/metabolism , Anemia/pathology , Cell Death , Cell Size , Erythrocytes/cytology , Erythrocytes/metabolism , Female , Hemolysis , Humans , Male , Oxidative Stress , Phosphatidylserines/metabolism , Reactive Oxygen Species/metabolism
17.
Toxins (Basel) ; 7(5): 1396-410, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25915718

ABSTRACT

The peptide antibiotic and ionophore gramicidin has previously been shown to trigger apoptosis of nucleated cells. In analogy to apoptosis, the suicidal death of erythrocytes or eryptosis involves cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Triggers of eryptosis include oxidative stress, increase of cytosolic Ca2+ activity ([Ca2+]i), and ceramide. The present study explored, whether gramicidin triggers eryptosis. To this end phosphatidylserine exposure at the cell surface was estimated from annexin V binding, cell volume from forward scatter, red blood cell distribution width (RDW) from electronic particle counting, reactive oxidant species (ROS) from 2',7'-dichlorodihydrofluorescein diacetate (DCFDA) fluorescence, [Ca2+]i from Fluo3- and Fluo4 fluorescence, and ceramide abundance from binding of specific antibodies. As a result, a 24 h exposure of human erythrocytes to gramicidin significantly increased the percentage of annexin-V-binding cells (≥1 µg/mL), forward scatter (≥0.5 µg/mL) and hemolysis. Gramicidin enhanced ROS activity, [Ca2+]i and ceramide abundance at the erythrocyte surface. The stimulation of annexin-V-binding by gramicidin was significantly blunted but not abolished by removal of extracellular Ca2+. In conclusion, gramicidin stimulates phospholipid scrambling of the erythrocyte cell membrane, an effect at least partially due to induction of oxidative stress, increase of [Ca2+]i and up-regulation of ceramide abundance. Despite increase of [Ca2+]i, gramicidin increases cell volume and slightly reduces RWD.


Subject(s)
Anti-Bacterial Agents/pharmacology , Erythrocytes/drug effects , Gramicidin/pharmacology , Calcium/metabolism , Cell Death/drug effects , Cell Size/drug effects , Ceramides/metabolism , Erythrocytes/metabolism , Humans , Oxidative Stress/drug effects , Phosphatidylserines/metabolism , Reactive Oxygen Species/metabolism
18.
Oncotarget ; 6(12): 10309-19, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25871399

ABSTRACT

Chorein encoded by VPS13A (vacuolar protein sorting-associated protein 13A) is defective in chorea-acanthocytosis. Chorein fosters neuronal cell survival, cortical actin polymerization and cell stiffness. In view of its anti-apoptotic effect in neurons, we explored whether chorein is expressed in cancer cells and influences cancer cell survival. RT-PCR was employed to determine transcript levels, specific siRNA to silence chorein, FACS analysis to follow apoptosis and Western blotting to quantify protein abundance. Chorein transcripts were detected in various cancer cell types. The mRNA coding for chorein and chorein protein were most abundant in drug resistant, poorly differentiated human rhabdomyosarcoma cells. Chorein silencing significantly reduced the ratio of phosphorylated (and thus activated) to total phosphoinositide 3 kinase (PI-3K), pointing to inactivation of this crucial pro-survival signaling molecule. Moreover, chorein silencing diminished transcript levels and protein expression of anti-apoptotic BCL-2 and enhanced transcript levels of pro-apoptotic Bax. Silencing of chorein in rhabdomyosarcoma cells was followed by mitochondrial depolarization, caspase 3 activation and stimulation of early and late apoptosis. In conclusion, chorein is expressed in various cancer cells. In cells with high chorein expression levels chorein silencing promotes apoptotic cell death, an effect paralleled by down-regulation of PI-3K activity and BCL-2/Bax expression ratio.


Subject(s)
Neuroacanthocytosis/metabolism , Rhabdomyosarcoma/metabolism , Vesicular Transport Proteins/biosynthesis , Caco-2 Cells , Child , Female , Humans , Neuroacanthocytosis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rhabdomyosarcoma/genetics , Transfection , Tumor Cells, Cultured , Vesicular Transport Proteins/genetics
19.
Hepatology ; 61(1): 275-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25065608

ABSTRACT

UNLABELLED: Hepatic failure is commonly associated with anemia, which may result from gastrointestinal bleeding, vitamin deficiency, or liver-damaging diseases, such as infection and alcohol intoxication. At least in theory, anemia during hepatic failure may result from accelerated clearance of circulating erythrocytes. Here we show that bile duct ligation (BDL) in mice leads to severe anemia despite increased reticulocyte numbers. Bilirubin stimulated suicidal death of human erythrocytes. Mechanistically, bilirubin triggered rapid Ca(2+) influx, sphingomyelinase activation, formation of ceramide, and subsequent translocation of phosphatidylserine to the erythrocyte surface. Consistent with our in vitro and in vivo findings, incubation of erythrocytes in serum from patients with liver disease induced suicidal death of erythrocytes in relation to their plasma bilirubin concentration. Consistently, patients with hyperbilirubinemia had significantly lower erythrocyte and significantly higher reticulocyte counts compared to patients with low bilirubin levels. CONCLUSION: Bilirubin triggers suicidal erythrocyte death, thus contributing to anemia during liver disease.


Subject(s)
Anemia/etiology , Bilirubin/blood , Erythrocytes/physiology , Liver Failure/complications , Aged , Animals , Calcium/metabolism , Case-Control Studies , Cell Death , Female , Healthy Volunteers , Humans , Liver Failure/blood , Male , Mice , Middle Aged , Sphingomyelin Phosphodiesterase/metabolism
20.
Cancer Biol Ther ; 16(1): 52-9, 2015.
Article in English | MEDLINE | ID: mdl-25427201

ABSTRACT

Functional membrane androgen receptors (mAR) have previously been described in MCF-7 breast cancer cells. Their stimulation by specific testosterone albumin conjugates (TAC) activate rapidly non-genomic FAK/PI3K/Rac1/Cdc42 signaling, trigger actin reorganization and inhibit cell motility. PI3K stimulates serum and glucocorticoid inducible kinase SGK1, which in turn regulates the function of mAR. In the present study we addressed the role of SGK1 in mAR-induced apoptosis. TAC-stimulated mAR activation elicited apoptosis of MCF-7 cells, an effect significantly potentiated by concomitant incubation of the cells with TAC and the specific SGK1 inhibitors EMD638683 and GSK650394. In line with this, TAC and EMD638683 activated caspase-3. These effects were insensitive to the classical androgen receptor (iAR) antagonist flutamide, pointing to iAR-independent, mAR-induced responses. mAR activation and SGK1 inhibition further considerably augmented the radiation-induced apoptosis of MCF-7 cells. Moreover, TAC- and EMD638683 triggered early actin polymerization in MCF-7 cells. Blocking actin restructuring with cytochalasin B abrogated the TAC- and EMD638683-induced pro-apoptotic responses. Further analysis of the molecular signaling revealed late de-phosphorylation of FAK and Akt. Our results demonstrate that mAR activation triggers pro-apoptotic responses in breast tumor cells, an effect significantly enhanced by SGK1 inhibition, involving actin reorganization and paralleled by down-regulation of FAK/Akt signaling.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/metabolism , Cell Membrane/metabolism , Immediate-Early Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Androgen/metabolism , Actins/metabolism , Caspase 3/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , MCF-7 Cells , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Testosterone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...