Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(9): e0291023, 2023.
Article in English | MEDLINE | ID: mdl-37682902

ABSTRACT

Helicase-like transcription factor (HLTF) also known as SMARCA3, protects genome integrity. A tumor suppressor, HLTF is expressed in tumor cells but not in the tumor microenvironment (TME) in early-stage colorectal cancer (CRC). With disease progression, there is high concordance between epigenetic silencing of HLTF in CRC cells and negligible HLTF expression in the TME. We developed a cell line-derived xenograft (CDX) model and show for the first time that HLTF-deletion in cancer cells and the TME results in metabolic reprogramming that mitigates oxidative stress in lymphatic intravascular metastatic niches. The two metabolic pathways that derive energy from glucose-glycolysis and oxidative phosphorylation (OXPHOS)-are variously utilized by cancer cells depending upon the TME. HIF-1α, a master regulator of glycolysis, was eliminated from a role in reprogramming metabolism to satisfy CDX energetic requirements by RNAseq and spatial transcriptomics. Variability in the gut microbiome, with a putative role in altered metabolism, was also eliminated. HLTF-deleted cancer cells recovered from DNA damage at a transcriptomic level induction of DNA repair and OXPHOS genes linked to an amoeboid-associated phenotype at the tumor border (confocal microscopy). HLTF-deleted cancer and endothelial cells of lymphatic (PDPN) intravascular niches in the TME shared a site-specific protein S-glutathionylation signature (2D DIGE, MALDI-TOF/TOF mass spectrometry) for three glycolytic enzymes (PGK1 Cys379/380, PGAM1 Cys55, ENOA1 Cys119) that diverted glycolysis in support of continued glutathione biosynthesis. The collective absence of HLTF/Hltf from tumor and TME achieved redox homeostasis throughout the CDX and promoted metastasis.


Subject(s)
Colorectal Neoplasms , Oxidative Phosphorylation , Humans , Animals , Endothelial Cells , Tumor Microenvironment/genetics , Transcription Factors/genetics , Glycolysis/genetics , Cell Line , Disease Models, Animal , Colorectal Neoplasms/genetics , DNA-Binding Proteins
2.
PLoS One ; 18(8): e0286109, 2023.
Article in English | MEDLINE | ID: mdl-37624843

ABSTRACT

Epigenetic mechanisms are integral to pancreatic ß cell function. Promoter hypermethylation of the helicase like-transcription factor (HLTF) gene-a component of the cellular DNA damage response that contributes to genome stability-has been implicated in age-associated changes in ß cells. To study HLTF, we generated global and ß cell-specific (ß) Hltf knockout (KO) immune competent (IC) and immune deficient (ID) Rag2-/IL2- mice. IC global and ß Hltf KO mice were neonatal lethal whereas ID global and ß Hltf KO newborn mice had normal survival. This focused our investigation on the effects of Rag2 interruption with common gamma chain interruption on ß cell function/survival. Three-way transcriptomic (RNAseq) analyses of whole pancreata from IC and ID newborn ß Hltf KO and wild type (Hltf +/+) controls combined with spatially resolved transcriptomic analysis of formalin fixed paraffin embedded tissue, immunohistochemistry and laser scanning confocal microscopy showed DNA damage caused by ß Hltf KO in IC mice upregulated the Hmgb1-Rage axis and a gene signature for innate immune cells. Perforin-delivered granzyme A (GzmA) activation of DNase, Nme1, showed damaged nuclear single-stranded DNA (γH2AX immunostaining). This caspase-independent method of cell death was supported by transcriptional downregulation of Serpinc1 gene that encodes a serine protease inhibitor of GzmA. Increased transcriptional availability of complement receptors C3ar1 and C5ar1 likely invited crosstalk with Hmgb1 to amplify inflammation. This study explores the complex dialog between ß cells and immune cells during development. It has implications for the initiation of type I diabetes in utero when altered gene expression that compromises genome stability invokes a localized inflammatory response.


Subject(s)
Insulin-Secreting Cells , Animals , Mice , Caspases , Causality , Granzymes , Transcription Factors
3.
PLoS One ; 16(5): e0251132, 2021.
Article in English | MEDLINE | ID: mdl-34010296

ABSTRACT

Methylation of the HLTF gene in colorectal cancer (CRC) cells occurs more frequently in men than women. Progressive epigenetic silencing of HLTF in tumor cells is accompanied by negligible expression in the tumor microenvironment (TME). Cell line-derived xenografts (CDX) were established in control (Hltf+/+) and Hltf-deleted male Rag2-/-IL2rg-/- mice by direct orthotopic cell microinjection (OCMI) of HLTF+/+HCT116 Red-FLuc cells into the submucosa of the cecum. Combinatorial induction of IL6 and S100A8/A9 in the Hltf-deleted TME with ICAM-1 and IL8 in the primary tumor activated a positive feedback loop. The proinflammatory niche produced a major shift in CDX metastasis to peritoneal dissemination compared to controls. Inducible nitric oxide (iNOS) gene expression and transactivation of the iNOS-S100A8/A9 signaling complex in Hltf-deleted TME reprogrammed the human S-nitroso-proteome. POTEE, TRIM52 and UN45B were S-nitrosylated on the conserved I/L-X-C-X2-D/E motif indicative of iNOS-S100A8/A9-mediated S-nitrosylation. 2D-DIGE and protein identification by MALDI-TOF/TOF mass spectrometry authenticated S-nitrosylation of 53 individual cysteines in half-site motifs (I/L-X-C or C-X-X-D/E) in CDX tumors. POTEE in CDX tumors is both a general S-nitrosylation target and an iNOS-S100A8/A9 site-specific (Cys638) target in the Hltf-deleted TME. REL is an example of convergence of transcriptomic-S-nitroso-proteomic signaling. The gene is transcriptionally activated in CDX tumors with an Hltf-deleted TME, and REL-SNO (Cys143) was found in primary CDX tumors and all metastatic sites. Primary CDX tumors from Hltf-deleted TME shared 60% of their S-nitroso-proteome with all metastatic sites. Forty percent of SNO-proteins from primary CDX tumors were variably expressed at metastatic sites. Global S-nitrosylation of proteins in pathways related to cytoskeleton and motility was strongly implicated in the metastatic dissemination of CDX tumors. Hltf-deletion from the TME played a major role in the pathogenesis of inflammation and linked protein S-nitrosylation in primary CDX tumors with spatiotemporal continuity in metastatic progression when the tumor cells expressed HLTF.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins/deficiency , Transcription Factors/deficiency , Animals , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Progression , HCT116 Cells , Heterografts , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Protein Interaction Maps , Proteome/genetics , Proteome/metabolism , S100 Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology
4.
Methods Mol Biol ; 2175: 47-63, 2020.
Article in English | MEDLINE | ID: mdl-32681483

ABSTRACT

The existence of nuclear pore complexes in the nuclear envelope has led to the assumption that ions move freely from the cytosol into the nucleus, and that the molecular mechanisms at the plasma membrane that regulate cytosolic pH also regulate nuclear pH. Furthermore, studies to measure pH in the nucleus have produced contradictory results, since it has been found that the nuclear pH is either similar to the cytosol or more alkaline than the cytosol. However, most studies of nuclear pH have lacked the rigor needed to understand pH regulation in the nucleus. A major problem has been the lack of in situ titrations in the nucleus and cytosol, since the intracellular environment is different in the cytosol and nucleus and the behavior of fluorescent pH probes is different in these environments. Here we present a method that uses the fluorescence of SNARF-1 that labels both cytosol and nucleus. Using ratio imaging microscopy, regions of interest corresponding to the nucleus and cytosol to perform steady-state pH measurements followed by in situ titrations, to correctly assign pH in those cellular domains.


Subject(s)
Cell Nucleus/physiology , Cytosol/physiology , Hydrogen-Ion Concentration , Microscopy, Fluorescence/methods , Benzopyrans/chemistry , Cell Line , Cell Nucleus/chemistry , Cell Physiological Phenomena , Cytosol/chemistry , Fluorescent Dyes/chemistry , Humans , Nuclear Envelope/physiology , Protons
5.
Mater Sci Eng C Mater Biol Appl ; 107: 110313, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31761227

ABSTRACT

Blood brain barrier (BBB), a barrier formed by endothelial cells, separates the brain from the circulatory system and protects the stability of central neural system normally, however, it also results in low permeability of vast majority of drugs for brain disease therapy. In this work, the cytotoxicity, uptake and transportation of 2D graphene nanosheet through BBB were investigated in in vitro models of BBB constructed by human brain microvascular endothelia cells (hBMECs). Permeability of two types of graphene nanosheet, including graphene oxide (GO) and porphyrin conjugated graphene oxide (PGO) through BBB were studied. With hydrophobic chemicals conjugation on its surface, permeability of PGO was greatly improved compared to GO. Furthermore, transportation behavior of assorted sizes of PGO obtained by differential velocity centrifugation through BBB was also explored, revealing that PGO with larger size has higher permeability than smaller-size PGO. The significant improved permeability of 2D graphene nanosheet through BBB compared to traditional drugs provides promising applications in drug delivery and disease therapy for brain disease in the near future.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems/methods , Graphite , Porphyrins , Cell Line , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Graphite/chemistry , Graphite/pharmacokinetics , Humans , Nanostructures/chemistry , Porphyrins/chemistry , Porphyrins/pharmacokinetics
6.
Biochim Biophys Acta Gen Subj ; 1863(1): 1-12, 2019 01.
Article in English | MEDLINE | ID: mdl-30279148

ABSTRACT

BACKGROUND: Metastatic tumor cells have acidic extracellular pH and differential electrochemical H+ gradients generated across their cell membranes by V-type H+-ATPases. This study shows that inhibition of the V-ATPases by the plant-derived monoterpene Myrtenal results in tumor cell death and decreased metastatic dissemination in mice. METHODS: The Myrtenal anticancer toxicity was evaluated in vitro using murine (B16F0 and B16F10) and human (SkMel-5) melanoma cell lines, and in in vivo mouse metastatic dissemination model. Proton flux and extracellular acidification were directly evaluated at the surface of living cells using a non-invasive selective ion electrode approach. RESULTS: The inhibition of V-ATPases by 100 µM Myrtenal disrupted the electrochemical H+ gradient across the cell membranes, strongly induced cell death (4-5 fold), and decreased tumor cells migration and invasion in vitro. Myrtenal (15 mg/kg) also significantly reduced metastasis induced by B16F10 in vivo, further reinforcing that V-ATPase is a molecular target to halt the progression of cancers. CONCLUSIONS: These data revealed the therapeutic potential of Myrtenal as inhibitor of melanoma progression proposing a mechanism of action by which once inhibited by this monoterpene the proton pumps fail to activate cancer-related differential electrochemical gradients and H+ fluxes across the tumor cell membranes, disrupting pH signatures inherent in tumor progression, resulting in reprogrammed cell death and metastasis inhibition. GENERAL SIGNIFICANCE: The work represents a new mechanistic strategy for contention of melanoma, the most aggressive and deadly form of cutaneous neoplasm, and highlights Myrtenal, other related monoterpenes and derivatives as promising proton pump inhibitors with high chemotherapeutic potential.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma/pathology , Skin Neoplasms/pathology , Terpenes/pharmacology , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Animals , Bicyclic Monoterpenes , Cell Death , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Disease Models, Animal , Drug Resistance, Neoplasm , Electrodes , Humans , Hydrogen-Ion Concentration , Male , Melanoma/drug therapy , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/prevention & control , Protons , Skin Neoplasms/drug therapy , Vacuolar Proton-Translocating ATPases/metabolism
7.
PLoS One ; 13(7): e0200211, 2018.
Article in English | MEDLINE | ID: mdl-29975766

ABSTRACT

Hltf is regulated by intron retention, and global Hltf-deletion causes perinatal lethality from hypoglycemia. In heart, full-length Hltf is a transcriptional regulator of Hif-1α that controls transport systems. Thus, we tested the hypothesis that Hltf deletion from placenta caused or exacerbated neonatal hypoglycemia via Hif-1α regulation of nutrient transporters. RNA-seq data analyses identified significant changes in transcript expression and alternative splicing (AS) in E18.5 placentome. iPathwayGuide was used for gene ontology (GO) analysis of biological processes, molecular functions and cellular components. Elim pruning algorithm identified hierarchical relationships. The methylome was interrogated by Methyl-MiniSeq Epiquest analysis. GO analysis identified gene enrichment within biological processes. Protein expression was visualized with immunohistochemistry. Although two Hltf mRNA isoforms are quantifiable in most murine tissues, only the truncated Hltf isoform is expressed in placenta. The responsible intron retention event occurs in the absence of DNA methylation. iPathwayGuide analysis identified 157 target genes of 11,538 total genes with measured expression. These were obtained using a threshold of 0.05 for statistical significance (p-value) and a long fold change of expression with absolute value of at least 0.6. Hltf deletion altered transcription of trophoblast lineage-specific genes, and increased transcription of the Cxcr7 (p = 0.004) gene whose protein product is a co-receptor for human and simian immunodeficiency viruses. Concomitant increased Cxcr7 protein was identified with immunolabeling. Hltf deletion had no effect on transcription or site-specific methylation patterns of Hif-1α, the major glucose transporters, or System A amino acid transporters. There was no measureable evidence of uteroplacental dysfunction or fetal compromise. iPathGuide analysis revealed Hltf suppresses cytolysis (10/21 genes; p-value 1.900e-12; p-value correction: Elim pruning; GO:019835) including the perforin-granzyme pathway in uterine natural killer cells. Our findings 1) prove the truncated Hltf protein isoform is a transcription factor, 2) establish a functional link between AS of Hltf and immunosuppression at the feto-maternal interface, 3) correlate intron retention with the absence of DNA methylation, and 4) underscore the importance of differential splicing analysis to identify Hltf's functional diversity.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Immune Tolerance/genetics , Maternal-Fetal Exchange/immunology , Placenta/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Alternative Splicing , Animals , Carrier Proteins , DNA Methylation , Exons , Female , Fetomaternal Transfusion/genetics , Fetomaternal Transfusion/pathology , Gene Expression Profiling , Introns , Mice , Mice, Inbred C57BL , Pregnancy , Protein Isoforms , Receptors, CXCR/genetics , Receptors, CXCR/immunology
8.
Cancer Biother Radiopharm ; 32(2): 49-56, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28301259

ABSTRACT

Recent successes in the development of new therapies for metastatic melanoma, such as mitogen-activated protein kinase pathway inhibitors, anticytotoxic T lymphocyte-associated antigen-4, and programmed cell death protein 1/programmed cell death ligand 1 (PD-L1) pathway-blocking antibodies, as well as combination strategies, all yielded promising results, changing the continually evolving landscape of therapeutic options for patients with melanoma. One promising new treatment modality is based on the use of immunomodulatory monoclonal antibodies that enhance the function of components of the antitumor immune response such as T cells or block immunologic checkpoints that restrain effective antitumor immunity. Program death-1 receptor and its ligand, PD-L1, is a major mechanism by which a tumor suppresses T cell-mediated antitumor immune responses. Studies in mice have shown that GK-1, an 18 amino acid peptide from Taenia crassiceps cisticerci, has the potential to be used as a primary or adjuvant component for the treatment of cancers by stimulating proinflammatory cytokines. The authors hypothesized that treatment with GK-1 in combination with anti-PD-L1 will increase survival in mice bearing melanoma tumors. C57BL/6 mice were injected with B16-F10-luc2 cells and separated into four groups: control, GK-1, anti-PD-L1, and GK-1/anti-PD-L1. The tumor sizes were measured and monitored using calipers and bioluminescence. The GK-1 peptide in combination with anti-PD-L1 showed significantly longer survival (34 days) compared with the other groups (23-27 days). This means an increase; survival increased 47.82% in the mice treated with GK-1+anti-PD-L1, 21.7% in mice treated with GK-1 alone, and 6.08% in those mice treated with anti-PD-L1 only. Blood samples were collected at days 0, 14, and at euthanization or end of the experiment and monitored for cytokines using mouse-specific V-PLEX Proinflammatory Panel. A decrease in TNF-α, IL-4, IL-5, IL-6, and IL-10 serum levels was observed in the GK-1/anti-PD-L1 combination group that may explain the beneficial effects of the combination treatment in prolonging the life of mice bearing melanoma. The data indicate that GK-1/anti-PD-L1 combined therapy affectively increases survival and warrants further clinical investigations.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Melanoma/drug therapy , Peptides, Cyclic/therapeutic use , Animals , Cell Line, Tumor , Cytokines/metabolism , Gene Expression Regulation, Neoplastic , Immunotherapy/methods , Inflammation , Luminescence , Male , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Peptides/therapeutic use , Proportional Hazards Models , Taenia
9.
Am J Physiol Cell Physiol ; 311(4): C547-C558, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27510904

ABSTRACT

The regulation of the luminal pH of each organelle is crucial for its function and must be controlled tightly. Nevertheless, it has been assumed that the nuclear pH is regulated by the cytoplasmic proton transporters via the diffusion of H+ across the nuclear pores because of their large diameter. However, it has been demonstrated that ion gradients exist between cytosol and nucleus, suggesting that the permeability of ions across the nuclear pores is restricted. Vacuolar H+-ATPase (V-H+-ATPase) is responsible for the creation and maintenance of trans-membrane electrochemical gradient. We hypothesize that V-H+-ATPase located in the nuclear membranes functions as the primary mechanism to regulate nuclear pH and generate H+ gradients across the nuclear envelope. We studied the subcellular heterogeneity of H+ concentration in the nucleus and cytosol using ratio imaging microscopy and SNARF-1, a pH indicator, in prostate cells. Our results indicate that there are proton gradients across the nuclear membranes that are generated by V-H+-ATPase located in the outer and inner nuclear membranes. We demonstrated that these gradients are mostly dissipated by inhibiting V-H+-ATPase. Immunoblots and V-H+-ATPase activity corroborated the existence of V-H+-ATPase in the nuclear membranes. This study demonstrates that V-H+-ATPase is functionally expressed in nuclear membranes and is responsible for nuclear H+ gradients that may promote not only the coupled transport of substrates, but also most electrochemically driven events across the nuclear membranes. This study represents a paradigm shift that the nucleus can regulate its own pH microenvironment, providing new insights into nuclear ion homeostasis and signaling.


Subject(s)
Cell Nucleus/metabolism , Cytosol/metabolism , Nuclear Envelope/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Vacuoles/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Hydrogen-Ion Concentration , Protons
10.
J Nutr Biochem ; 30: 14-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27012617

ABSTRACT

Intimal macrophages are determinant cells for atherosclerotic lesion formation by releasing inflammatory factors and taking up oxidized low-density lipoprotein (oxLDL) via scavenger receptors, primarily the CD36 receptor. (-)-Epigallocatechin-3-gallate (EGCG) has a potential to decrease cholesterol accumulation and inflammatory responses in macrophages. We made EGCG-loaded nanoparticles (Enano) using phosphatidylcholine, kolliphor HS15, alpha-tocopherol acetate and EGCG. 1-(Palmitoyl)-2-(5-keto-6-octene-dioyl) phosphatidylcholine (KOdiA-PC), a CD36-targeted ligand found on oxLDL, was incorporated on the surface of Enano to make ligand-Enano (L-Enano). The objectives of this study are to deliver EGCG to macrophages via CD36-targeted L-Enano and to determine its antiatherogenic bioactivities. The optimized nanoparticles obtained in our study were spherical and around 108 nm in diameter, and had about 10% of EGCG loading capacity and 96% of EGCG encapsulation efficiency. Compared to Enano, CD36-targeted L-Enano had significantly higher binding affinity to and uptake by macrophages at the same pattern as oxLDL. CD36-targeted L-Enano dramatically improved EGCG stability, increased macrophage EGCG content, delivered EGCG to macrophage cytosol and avoided lysosomes. L-Enano significantly decreased macrophage mRNA levels and protein secretion of monocyte chemoattractant protein 1, but did not significantly change macrophage cholesterol content. The innovative CD36-targeted nanoparticles may facilitate targeted delivery of diagnostic, preventive and therapeutic compounds to intimal macrophages for the diagnosis, prevention and treatment of atherosclerosis with enhanced efficacy and decreased side effects.


Subject(s)
Atherosclerosis/prevention & control , CD36 Antigens/chemistry , Catechin/analogs & derivatives , Nanoparticles , Catechin/administration & dosage , Catechin/chemistry , Humans
11.
ACS Biomater Sci Eng ; 2(8): 1357-1366, 2016 Aug 08.
Article in English | MEDLINE | ID: mdl-33434989

ABSTRACT

Brain cancer is a fatal disease that is difficult to treat because of poor targeting and low permeability of chemotherapeutic drugs through the blood brain barrier. In a comparison to current treatments, such as surgery followed by chemotherapy and/or radiotherapy, photothermal therapy is a remarkable noninvasive therapy developed in recent years. In this work, porphyrin immobilized nanographene oxide (PNG) was synthesized and bioconjugated with a peptide to achieve enhanced and targeted photothermal therapy for brain cancer. PNG was dispersed into the agar based artificial tissue model and demonstrated a photo-to-thermal conversion efficiency of 19.93% at a PNG concentration of only 0.5 wt %, with a heating rate of 0.6 °C/s at the beginning of irradiation. In comparison, 0.5 wt % graphene oxide (GO) indicated a photo-to-thermal conversion efficiency of 12.20% and a heating rate of 0.3 °C/s. To actively target brain tumor cells without harming healthy cells and tissues surrounding the laser path, a tripeptide l-arginyl-glycyl-l-aspartic (RGD) was further grafted to PNG. The photothermal therapy effects of PNG-RGD completely eliminated the tumor in vivo, indicating its excellent therapeutic effect for the treatment of brain cancer.

12.
J Control Release ; 220(Pt A): 61-70, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26450668

ABSTRACT

Current approaches to the diagnosis and therapy of atherosclerosis cannot target lesion-determinant cells in the artery wall. Intimal macrophage infiltration promotes atherosclerotic lesion development by facilitating the accumulation of oxidized low-density lipoproteins (oxLDL) and increasing inflammatory responses. The presence of these cells is positively associated with lesion progression, severity and destabilization. Hence, they are an important diagnostic and therapeutic target. The objective of this study was to noninvasively assess the distribution and accumulation of intimal macrophages using CD36-targeted nanovesicles. Soy phosphatidylcholine was used to synthesize liposome-like nanovesicles. 1-(Palmitoyl)-2-(5-keto-6-octene-dioyl) phosphatidylcholine was incorporated on their surface to target the CD36 receptor. All in vitro data demonstrate that these targeted nanovesicles had a high binding affinity for the oxLDL binding site of the CD36 receptor and participated in CD36-mediated recognition and uptake of nanovesicles by macrophages. Intravenous administration into LDL receptor null mice of targeted compared to non-targeted nanovesicles resulted in higher uptake in aortic lesions. The nanovesicles co-localized with macrophages and their CD36 receptors in aortic lesions. This molecular target approach may facilitate the in vivo noninvasive imaging of atherosclerotic lesions in terms of intimal macrophage accumulation and distribution and disclose lesion features related to inflammation and possibly vulnerability thereby facilitate early lesion detection and targeted delivery of therapeutic compounds to intimal macrophages.


Subject(s)
Atherosclerosis/diagnosis , CD36 Antigens/metabolism , Macrophages/metabolism , Nanoparticles/chemistry , Animals , Cells, Cultured , Humans , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, LDL/physiology
13.
Biochem Biophys Res Commun ; 464(1): 312-7, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26119689

ABSTRACT

Removal of cholesterol from macrophage-derived foam cells is a critical step to the prevention of atherosclerotic lesions. We have recently demonstrated the functional importance of retinoids in the regulation of the steroidogenic acute regulatory (StAR) protein that predominantly mediates the intramitochondrial transport of cholesterol in target tissues. In the present study, treatment of mouse macrophages with retinoids, particularly all-trans retinoic acid (atRA) and 9-cis RA, resulted in increases in cholesterol efflux to apolipoprotein AI (Apo-A1). Activation of the PKA pathway by a cAMP analog, (Bu)2cAMP, markedly augmented retinoid mediated cholesterol efflux. Macrophages overexpressing hormone-sensitive lipase increased the hydrolysis of cholesteryl esters and concomitantly enhanced the efficacy of retinoic acid receptor and liver X receptor (LXR) ligands on StAR and ATP-binding cassette transporter A1 (ABCA1) protein levels. RAs elevated StAR promoter activity in macrophages, and an increase in StAR levels augmented cholesterol efflux to Apo-A1, suggesting retinoid-mediated efflux of cholesterol involves enhanced oxysterol production. Further studies revealed that retinoids activate the LXR regulated genes, sterol receptor-element binding protein-1c and ABCA1. These findings provide insights into the regulatory events in which retinoid signaling effectively enhances macrophage cholesterol efflux and indicate that retinoid therapy may have important implications in limiting and/or regressing atherosclerotic cardiovascular disease.


Subject(s)
Cholesterol/metabolism , Macrophages/drug effects , Orphan Nuclear Receptors/agonists , Tretinoin/analogs & derivatives , Tretinoin/pharmacology , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Apolipoprotein A-I/metabolism , Biological Transport/drug effects , Bucladesine/pharmacology , Cell Line , Cholesterol Esters/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Hydrolysis/drug effects , Liver X Receptors , Macrophages/cytology , Macrophages/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Signal Transduction , Sterol Esterase/genetics , Sterol Esterase/metabolism
14.
J Biol Chem ; 290(6): 3680-92, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25505184

ABSTRACT

The vacuolar (H(+))-ATPases (V-ATPases) are a family of ATP-driven proton pumps that couple ATP hydrolysis with translocation of protons across membranes. Previous studies have implicated V-ATPases in cancer cell invasion. It has been proposed that V-ATPases participate in invasion by localizing to the plasma membrane and causing acidification of the extracellular space. To test this hypothesis, we utilized two separate approaches to specifically inhibit plasma membrane V-ATPases. First, we stably transfected highly invasive MDA-MB231 cells with a V5-tagged construct of the membrane-embedded c subunit of the V-ATPase, allowing for extracellular expression of the V5 epitope. We evaluated the effect of addition of a monoclonal antibody directed against the V5 epitope on both V-ATPase-mediated proton translocation across the plasma membrane and invasion using an in vitro Matrigel assay. The addition of anti-V5 antibody resulted in acidification of the cytosol and a decrease in V-ATPase-dependent proton flux across the plasma membrane in transfected but not control (untransfected) cells. These results demonstrate that the anti-V5 antibody inhibits activity of plasma membrane V-ATPases in transfected cells. Addition of the anti-V5 antibody also inhibited in vitro invasion of transfected (but not untransfected) cells. Second, we utilized a biotin-conjugated form of the specific V-ATPase inhibitor bafilomycin. When bound to streptavidin, this compound cannot cross the plasma membrane. Addition of this compound to MDA-MB231 cells also inhibited in vitro invasion. These studies suggest that plasma membrane V-ATPases play an important role in invasion of breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Protons , Vacuolar Proton-Translocating ATPases/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Ion Transport , Macrolides/pharmacology , Neoplasm Invasiveness , Protein Transport , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors
15.
PLoS One ; 8(11): e80461, 2013.
Article in English | MEDLINE | ID: mdl-24278285

ABSTRACT

HLTF/Hltf regulates transcription, remodels chromatin, and coordinates DNA damage repair. Hltf is expressed in mouse brain and heart during embryonic and postnatal development. Silencing Hltf is semilethal. Seventy-four percent of congenic C57BL/6J Hltf knockout mice died, 75% within 12-24 hours of birth. Previous studies in neonatal (6-8 hour postpartum) brain revealed silencing Hltf disrupted cell cycle progression, and attenuated DNA damage repair. An RNA-Seq snapshot of neonatal heart transcriptome showed 1,536 of 20,000 total transcripts were altered (p < 0.05) - 10 up- and 1,526 downregulated. Pathway enrichment analysis with MetaCore™ showed Hltf's regulation of the G2/M transition (p=9.726E(-15)) of the cell cycle in heart is nearly identical to its role in brain. In addition, Brca1 and 12 members of the Brca1 associated genome surveillance complex are also downregulated. Activation of caspase 3 coincides with transcriptional repression of Bcl-2. Hltf loss caused downregulation of Wt1/Gata4/Hif-1a signaling cascades as well as Myh7b/miR499 transcription. Hltf-specific binding to promoters and/or regulatory regions of these genes was authenticated by ChIP-PCR. Hif-1a targets for prolyl (P4ha1, P4ha2) and lysyl (Plod2) collagen hydroxylation, PPIase enzymes (Ppid, Ppif, Ppil3) for collagen trimerization, and lysyl oxidase (Loxl2) for collagen-elastin crosslinking were downregulated. However, transcription of genes for collagens, fibronectin, Mmps and their inhibitors (Timps) was unaffected. The collective downregulation of genes whose protein products control collagen biogenesis caused disorganization of the interstitial and perivascular myocardial collagen fibrillar network as viewed with picrosirius red-staining, and authenticated with spectral imaging. Wavy collagen bundles in control hearts contrasted with collagen fibers that were thin, short and disorganized in Hltf null hearts. Collagen bundles in Hltf null hearts were tangled and fragmented. Thus, silencing Hltf during heart organogenesis compromised DNA double-strand break repair, and caused aberrant collagen biogenesis altering the structural network that transmits cardiomyocyte force into muscle contraction.


Subject(s)
Cell Division , DNA-Binding Proteins/physiology , G2 Phase , GATA4 Transcription Factor/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myocardium/metabolism , Transcription Factors/physiology , Transcription, Genetic , WT1 Proteins/metabolism , Animals , Base Sequence , Chromatin Immunoprecipitation , DNA Primers , Echocardiography , Female , Mice , Mice, Knockout , Polymerase Chain Reaction , Pregnancy
16.
Methods Mol Biol ; 937: 253-71, 2013.
Article in English | MEDLINE | ID: mdl-23007592

ABSTRACT

Although changes in both pH(in) and [Ca(2+)](i) have been observed in response to a variety of agonists, it is not clear whether these ionic events work independently or are coordinated to lead to a specific physiological response. One of the fundamental problems in studying these ionic events is that changes in pH(in) modify Ca(2+) regulatory mechanisms and changes in Ca(2+) may modify pH regulation. It is desirable to use a technique that allows concomitant monitoring of these two ions in cell populations with high time resolution. Furthermore, like many Ca(2+) binding proteins, all Ca(2+)-sensitive fluoroprobes are inherently sensitive to pH owing to competition of H(+) for the Ca(2+)-binding sites. This chapter describes experimental paradigms that provide optimum conditions for simultaneous measurement of pH from the fluorescence emission of snarf-1, and Ca(2+) using fura-2. The fluorescence spectra of these compounds are sufficiently different to allow simultaneous measurement of pH and Ca(2+) both in vitro and in vivo. Moreover, the ratio of the H(+)-sensitive wavelengths of snarf-1 is unaffected by Ca(2+), or the concomitant presence of fura-2 in cells. Although the fluorescence ratio of fura-2 is insensitive to the presence of snarf-1, it is affected by pH, as indicated above. We describe procedures to correct for this effect and to obtain calibration parameters for fura-2 and snarf-1 required to facilitate analysis of pH and Ca(2+) concentrations within cell populations.


Subject(s)
Calcium/metabolism , Benzopyrans/metabolism , Cell Line , Fura-2/metabolism , Humans , Hydrogen-Ion Concentration , Naphthols/metabolism , Rhodamines/metabolism
17.
Biochim Biophys Acta ; 1820(7): 1111-20, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22366469

ABSTRACT

BACKGROUND: Despite recent advances, it is not clear to correlate the mechanical compliances and the metastatic potential of cancer cells. In this study, we investigated combined signatures of mechanical compliances, adhesions, and calcium dynamics correlated with the metastatic potential of cancer cells. SCOPE OF REVIEW: We used the lowly (LNCaP) and highly (CL-1, CL-2) metastatic human prostate cancer cells. The AFM-based nanomechanics was performed to determine the elastic moduli and the cell-to-substrate adhesion. The intracellular calcium dynamics was evaluated by fluorescence spectroscopy. Cell migration and the distribution of cytoskeleton were evaluated using the wounded monolayer model and immunofluorescence, respectively. The elastic moduli, the calcium dynamics, and the migratory ability are greater in CL-1 and CL-2 than LNCaP. CL-1 and CL-2 also display a significantly larger area of cell-to-substrate adhesions while the LNCaP displays a limited adhesion. These properties were slightly reduced in CL-2 compared with CL-1 cells. The enhanced elastic moduli and calcium dynamics found in CL-1 and CL-2 can be consistently explained by the intensified tensile stress generated by actin cytoskeletons anchored at more focal adhesion sites. MAJOR CONCLUSIONS: Although the suppressed mechanical compliance of highly metastatic cells may not support the enhanced cancer metastasis, the enhanced adhesion and calcium dynamics are favorable for invasion and extra-vasation required for malignant progression. GENERAL SIGNIFICANCE: Our results suggest that the mechanical compliance alone may fail to indicate the metastatic progression, but the combined biomechanical signatures of mechanical compliance, adhesion, and calcium dynamics can provide critical clues to determine the metastatic potential of cells.


Subject(s)
Calcium/metabolism , Extracellular Matrix/chemistry , Microscopy, Atomic Force , Nanotechnology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Cell Adhesion , Cell Movement , Humans , Male , Neoplasm Metastasis , Tensile Strength , Tumor Cells, Cultured
18.
Curr Protein Pept Sci ; 13(2): 152-63, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22044157

ABSTRACT

Up-regulated aerobic glycolysis is a hallmark of malignant cancers. Little is understood about the reasons why malignant tumors up-regulate glycolysis and acidify their microenvironment. Signaling pathways involved in glucose changes are numerous. However, the identity of the internal glucose signal remains obscure. In this review we address the question of the significance of vacuolar proton ATPase (V-ATPase) and its relationship to up-regulated glycolysis in tumors. We know that glycolysis is extremely sensitive to changes in pH. Importantly, the V-ATPase activity is sensitive to glucose availability. Therefore, we propose that pH acts as the glucose signal via the V-ATPase that responds to changes in intracellular pH and acts as a sensor. We hypothesize that the increase in glycolysis leads to intracellular acidification and activates the V-ATPase to maintain a more alkaline intracellular pH in tumors by up-regulating glycolysis. This review attempts to provide a comprehensive description of the current knowledge about the role of V-ATPase in cancer, highlighting its role as a key player in the pH signaling pathway.


Subject(s)
Neoplasms/enzymology , Signal Transduction , Vacuolar Proton-Translocating ATPases/analysis , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Glycolysis , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1/metabolism , Models, Molecular , Neoplasms/metabolism , Protein Subunits/analysis , Protein Subunits/chemistry , Protein Subunits/metabolism , Vacuolar Proton-Translocating ATPases/chemistry , Wnt Proteins/metabolism , beta Catenin/metabolism
19.
J Biol Chem ; 284(24): 16400-16408, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19366680

ABSTRACT

It has previously been shown that highly invasive MDA-MB231 human breast cancer cells express vacuolar proton-translocating ATPase (V-ATPases) at the cell surface, whereas the poorly invasive MCF7 cell line does not. Bafilomycin, a specific V-ATPase inhibitor, reduces the in vitro invasion of MB231 cells but not MCF7 cells. Targeting of V-ATPases to different cellular membranes is controlled by isoforms of subunit a. mRNA levels for a subunit isoforms were measured in MB231 and MCF7 cells using quantitative reverse transcription-PCR. The results show that although all four isoforms are detectable in both cell types, levels of a3 and a4 are much higher in MB231 than in MCF7 cells. Isoform-specific small interfering RNAs (siRNA) were employed to selectively reduce mRNA levels for each isoform in MB231 cells. V-ATPase function was assessed using the fluorescent indicators SNARF-1 and pyranine to monitor the pH of the cytosol and endosomal/lysosomal compartments, respectively. Cytosolic pH was decreased only on knockdown of a3, whereas endosome/lysosome pH was increased on knockdown of a1, a2, and a3. Treatment of cells with siRNA to a4 did not affect either cytosolic or endosome/lysosome pH. Measurement of invasion using an in vitro transwell assay revealed that siRNAs to both a3 and a4 significantly inhibited invasion of MB231 cells. Immunofluorescence staining of MB231 cells for V-ATPase distribution revealed extensive intracellular staining, with plasma membrane staining observed in approximately 18% of cells. Knockdown of a4 had the greatest effect on plasma membrane staining, leading to a 32% reduction. These results suggest that the a4 isoform may be responsible for targeting V-ATPases to the plasma membrane of MB231 cells and that cell surface V-ATPases play a significant role in invasion. However, other V-ATPases affecting the pH of the cytosol and intracellular compartments, particularly those containing a3, are also involved in invasion.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Cathepsin L , Cathepsins/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cytosol/metabolism , Endosomes/metabolism , Enzyme Precursors/metabolism , Fluorescent Antibody Technique , Homeostasis/physiology , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Lysosomes/metabolism , Protein Subunits , Proton-Translocating ATPases/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Vacuolar Proton-Translocating ATPases/genetics
20.
J Bioenerg Biomembr ; 39(5-6): 427-33, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18058006

ABSTRACT

Angiogenesis, i.e., new blood vessel formation, is required in normal and pathological states. A dysfunction in the microvascular endothelium occurs in diabetes, leading to decreased blood flow and limb amputation. In cancer, angiogenesis is increased to allow for growth, invasion, and metastasis of tumor cells. Better understanding of the molecular events that cause or are associated with either of these diseases is needed to develop therapies. The tumor and angiogenic cells micro-environment is acidic and not permissive for growth. We have shown that to survive this environment, highly metastatic and angiogenic cells employ vacuolar H+-ATPase at their plasma membranes (pmV-ATPases) to maintain an alkaline pHcyt. However, in lowly metastatic and in microvascular endothelial cells from diabetic model, the density of pmV-ATPase and the cell invasiveness are decreased. Therefore, the overexpression of the pmV-ATPase is important for cell invasion, and essential for tumor progression, angiogenesis and metastasis. Both, cancer and diabetes are heterogenous diseases that involve many different proteins and signaling pathways. Changes in pHcyt have been associated with the regulation of a myriad of proteins, signaling molecules and pathways affecting many if not all cellular functions. Since changes in pHcyt are pleiotropic, we hypothesize that alteration in a single protein, pmV-ATPase, that can regulate pHcyt may explain the dysfunction of many proteins and cellular pathways in diabetes and cancer. Our long term goal is to determine the molecular mechanisms by which pmV-ATPase expression regulates tumor angiogenesis and metastasis. Such knowledge would be useful to identify targets for cancer therapy.


Subject(s)
Diabetes Mellitus/enzymology , Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Vacuolar Proton-Translocating ATPases/physiology , Animals , Diabetes Complications , Gene Expression Regulation, Enzymologic , Humans , Hydrogen-Ion Concentration , Neoplasms/etiology , Neoplasms/pathology , Vacuolar Proton-Translocating ATPases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...