Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 78(17-18): 6283-6304, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34279699

ABSTRACT

Proper functioning of each secretory and endocytic compartment relies on its unique pH micro-environment that is known to be dictated by the rates of V-ATPase-mediated H+ pumping and its leakage back to the cytoplasm via an elusive "H+ leak" pathway. Here, we show that this proton leak across Golgi membranes is mediated by the AE2a (SLC4A2a)-mediated bicarbonate-chloride exchange, as it is strictly dependent on bicarbonate import (in exchange for chloride export) and the expression level of the Golgi-localized AE2a anion exchanger. In the acidic Golgi lumen, imported bicarbonate anions and protons then facilitate a common buffering reaction that yields carbon dioxide and water before their egress back to the cytoplasm via diffusion or water channels. The flattened morphology of the Golgi cisternae helps this process, as their high surface-volume ratio is optimal for water and gas exchange. Interestingly, this net acid efflux pathway is often upregulated in cancers and established cancer cell lines, and responsible for their markedly elevated Golgi resting pH and attenuated glycosylation potential. Accordingly, AE2 knockdown in SW-48 colorectal cancer cells was able to restore these two phenomena, and at the same time, reverse their invasive and anchorage-independent growth phenotype. These findings suggest a possibility to return malignant cells to a benign state by restoring Golgi resting pH.


Subject(s)
Golgi Apparatus/metabolism , Animals , COS Cells , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chloride-Bicarbonate Antiporters/antagonists & inhibitors , Chloride-Bicarbonate Antiporters/genetics , Chloride-Bicarbonate Antiporters/metabolism , Chlorocebus aethiops , Glycosylation , Humans , Hydrogen-Ion Concentration , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation
2.
Biomaterials ; 34(36): 9134-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24008034

ABSTRACT

Tumour targeting nanoparticles (NPs) have demonstrated great potential for enhancing anticancer drug delivery to tumour sites and for reducing the side effects of chemotherapy. However, many nanoparticulate delivery systems still lack efficient tumour accumulation. In this work, we present a porous silicon (PSi) nanovector functionalized with a tumour-homing peptide, which targets the mammary-derived growth inhibitor (MDGI) expressing cancer cells both in vitro and in vivo, thereby enhancing the accumulation of the NPs in the tumours. We demonstrated that the tumour homing peptide (herein designated as CooP) functionalized thermally hydrocarbonized PSi (THCPSi) NPs homed specifically to the subcutaneous MDGI-expressing xenograft tumours. The THCPSi-CooP NPs were stable in human plasma and their uptake by MDGI-expressing cancer cells measured by confocal microscopy and flow cytometry was significantly increased compared to the non-functionalized THCPSi NPs. After intravenous injections into nude mice bearing MDGI-expressing tumours, effective targeting was detected and THCPSi-CooP NPs showed ~9-fold higher accumulation in the tumour site compared to the control THCPSi NPs. Accumulation of both NPs in the vital organs was negligible.


Subject(s)
Drug Delivery Systems , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Peptides/therapeutic use , Silicon/chemistry , Adsorption , Animals , Blood Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Fatty Acid-Binding Proteins/metabolism , Female , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Peptides/pharmacology , Porosity , Spectroscopy, Fourier Transform Infrared , Tissue Distribution/drug effects
3.
Ann Med ; 44(6): 542-54, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21585247

ABSTRACT

Most organelles within the exocytic and endocytic pathways typically acidify their interiors, a phenomenon that is known to be crucial for their optimal functioning in eukaryotic cells. This review highlights recent advances in our understanding of how Golgi acidity is maintained and regulated, and how its misregulation contributes to organelle dysfunction and disease. Both its biosynthetic products (glycans) and protein-sorting events are highly sensitive to changes in Golgi luminal pH and are affected in certain human disease states such as cancers and cutis laxa. Other potential disease states that are caused by, or are associated with, Golgi pH misregulation will also be discussed.


Subject(s)
Disease/etiology , Eukaryotic Cells/metabolism , Golgi Apparatus/metabolism , Organelles/metabolism , Cutis Laxa/etiology , Cutis Laxa/physiopathology , Cystic Fibrosis/etiology , Cystic Fibrosis/physiopathology , Endocytosis/physiology , Eukaryotic Cells/physiology , Exocytosis/physiology , Glycosylation , Golgi Apparatus/physiology , Humans , Hydrogen-Ion Concentration , Neoplasms/etiology , Neoplasms/physiopathology , Organelles/physiology , Pemphigus, Benign Familial/etiology , Pemphigus, Benign Familial/physiopathology
4.
Am J Pathol ; 179(2): 1004-14, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21689627

ABSTRACT

Nucleophosmin (NPM) is a multifunctional protein involved in a complex network of interactions. The role of NPM in oncogenesis is controversial. The NPM gene (NPM1) is mutated or rearranged in a number of hematological disorders, but such changes have not been detected in solid cancers. However, experiments with cultured NPM-null cells and with mice carrying a single inactivated NPM allele indicate a tumor suppressor function for NPM. To resolve the role of NPM in solid cancers, we examined its expression and localization in histologically normal breast tissue and a large array of human breast carcinoma samples (n = 1160), and also evaluated its association with clinicopathological variables and patient survival. The intensity and localization (nucleolar, nuclear, cytoplasmic) of NPM varied across clinical samples. No mutations explaining the differences were found, but the present findings indicate that expression levels of NPM affected its localization. Our study also revealed a novel granular staining pattern for NPM, which was an independent prognostic factor of poor prognosis. In addition, reduced levels of NPM protein were associated with poor prognosis. Furthermore, luminal epithelial cells of histologically normal breast displayed high levels of NPM and overexpression of NPM in the invasive MDA-MB-231 cells abrogated their growth in soft agar. These results support a tumor suppressive role for NPM in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Nuclear Proteins/biosynthesis , Nuclear Proteins/physiology , Adult , Aged , Carcinoma/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Separation , Estradiol/metabolism , Estrogens/metabolism , Female , Flow Cytometry , HEK293 Cells , Humans , Immunohistochemistry/methods , Middle Aged , Nucleophosmin , Time Factors
5.
Methods Mol Biol ; 683: 401-15, 2011.
Article in English | MEDLINE | ID: mdl-21053146

ABSTRACT

Each normal organ and pathological condition expresses a distinct set of molecules on their vasculature. These molecular signatures have been efficiently profiled using in vivo phage display technology. Using this technology, several peptides homing specifically to tumour blood vessels, lymphatic vessels, and/or tumour cells as well as to various normal organs have been isolated. Peptides homing to specific vascular addresses have revealed novel tissue-specific biomarkers of the normal and diseased vasculature. Tumour homing peptides have been successfully used to target therapies and imaging agents to tumours. In this review, we describe experimental setup for a combined ex vivo and in vivo screening procedure to select peptides homing to tumours.


Subject(s)
Peptide Library , Peptides/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Dystrophin/genetics , Exons/genetics , Female , Mice , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Transport , Reproducibility of Results
6.
J Biol Chem ; 285(23): 17771-7, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20378551

ABSTRACT

Glycans (i.e. oligosaccharide chains attached to cellular proteins and lipids) are crucial for nearly all aspects of life, including the development of multicellular organisms. They come in multiple forms, and much of this diversity between molecules, cells, and tissues is generated by Golgi-resident glycosidases and glycosyltransferases. However, their exact mode of functioning in glycan processing is currently unclear. Here we investigate the supramolecular organization of the N-glycosylation pathway in live cells by utilizing the bimolecular fluorescence complementation approach. We show that all four N-glycosylation enzymes tested (beta-1,2-N-acetylglucosaminyltransferase I, beta-1,2-N-acetylglucosaminyltransferase II, 1,4-galactosyltransferase I, and alpha-2,6-sialyltransferase I) form Golgi-localized homodimers. Intriguingly, the same enzymes also formed two distinct and functionally relevant heterodimers between the medial Golgi enzymes beta-1,2-N-acetylglucosaminyltransferase I and beta-1,2-N-acetylglucosaminyltransferase II and the trans-Golgi enzymes 1,4-galactosyltransferase I and alpha-2,6-sialyltransferase I. Given their strict Golgi localization and sequential order of function, the two heterodimeric complexes are probably responsible for the processing and maturation of N-glycans in live cells.


Subject(s)
Bacterial Proteins/chemistry , Glucosyltransferases/chemistry , Golgi Apparatus/metabolism , Animals , Base Sequence , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Dimerization , Glycosylation , Glycosyltransferases/metabolism , HeLa Cells , Humans , Lipids/chemistry , Molecular Sequence Data , Polysaccharides/chemistry
7.
J Cell Physiol ; 220(1): 144-54, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19277980

ABSTRACT

Acidic pH of the Golgi lumen is known to be crucial for correct glycosylation, transport and sorting of proteins and lipids during their transit through the organelle. To better understand why Golgi acidity is important for these processes, we have examined here the most pH sensitive events in N-glycosylation by sequentially raising Golgi luminal pH with chloroquine (CQ), a weak base. We show that only a 0.2 pH unit increase (20 microM CQ) is sufficient to markedly impair terminal alpha(2,3)-sialylation of an N-glycosylated reporter protein (CEA), and to induce selective mislocalization of the corresponding alpha(2,3)-sialyltransferase (ST3) into the endosomal compartments. Much higher pH increase was required to impair alpha(2,6)-sialylation, or the proximal glycosylation steps such as beta(1,4)-galactosylation or acquisition of Endo H resistance, and the steady-state localization of the key enzymes responsible for these modifications (ST6, GalT I, MANII). The overall Golgi morphology also remained unaltered, except when Golgi pH was raised close to neutral. By using transmembrane domain chimeras between the ST6 and ST3, we also show that the luminal domain of the ST6 is mainly responsible for its less pH sensitive localization in the Golgi. Collectively, these results emphasize that moderate Golgi pH alterations such as those detected in cancer cells can impair N-glycosylation by inducing selective mislocalization of only certain Golgi glycosyltransferases.


Subject(s)
Glycosyltransferases/metabolism , Golgi Apparatus/enzymology , Protein Processing, Post-Translational , Animals , COS Cells , Carcinoembryonic Antigen/metabolism , Chlorocebus aethiops , Chloroquine/pharmacology , Dose-Response Relationship, Drug , Endosomes/enzymology , Galactosyltransferases/metabolism , Glycosylation , Glycosyltransferases/genetics , Golgi Apparatus/drug effects , Hydrogen-Ion Concentration , Lysosomes/enzymology , Mannosidases/metabolism , Mutation , Protein Processing, Post-Translational/drug effects , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/metabolism , Sialyltransferases/metabolism , Time Factors , Transfection
8.
Int J Cancer ; 121(11): 2443-50, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17657737

ABSTRACT

Carcinoembryonic antigen (CEA, ceacam5) is an important tumor-associated antigen with reported roles, e.g., in immunological defense, cell adhesion, cell survival and metastasis. Its overexpression in cancer cells is known to involve transcriptional activation of the CEA gene, but the underlying molecular details remain unclear. Here, we show that hypoxia and intracellular alkalinization, 2 factors commonly found in solid tumors, increase CEA protein expression in breast (MCF-7) and colorectal (CaCo-2 and HT-29) cancer cells. The increase was comparable (2-3-fold) to that observed in colorectal carcinomas in vivo. CEA promoter analyses further revealed that this upregulation involves a known binding site for HIF-1 transcription factor (5'-ACGTG-3') within one of the CEA promoter's positive regulatory elements (the FP1 site; the E-box). Accordingly, deletion or targeted mutagenesis of this motif rendered the CEA promoter unresponsive to hypoxia. Our chromatin immunoprecipitation data confirmed that endogenous HIF-1alpha binds to the CEA promoter in hypoxic cells but not in normoxic cells. Moreover, overexpression of the hypoxia-inducible factor (HIF-1alpha) was sufficient to increase CEA protein expression in the cells. In contrast, c-Myc, which is known to bind to the overlapping E-box, did not potentiate HIF-1alpha-induced CEA expression. CEA overexpression in vivo was also found to coincide with the expression of carbonic anhydrase IX, a well-known hypoxia marker. Collectively, these results define CEA as a hypoxia-inducible protein and suggest an important role for the tumor microenvironmental factors in CEA overexpression during tumorigenesis.


Subject(s)
Breast Neoplasms/metabolism , Carcinoembryonic Antigen/metabolism , Colorectal Neoplasms/metabolism , Blotting, Western , Caco-2 Cells , Carcinoembryonic Antigen/genetics , Cell Hypoxia , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Female , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Polymerase Chain Reaction , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Up-Regulation
9.
J Cell Physiol ; 208(1): 167-74, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16547942

ABSTRACT

Altered glycosylation has turned out to be a universal feature of cancer cells, and in many cases, to correlate with altered expression or localization of relevant glycosyltransferases. However, no such correlation exists between observed enzymatic changes and the expression of the oncofetal Thomsen-Friedenreich (T)-antigen, a core 1 (Gal-beta1 --> 3-GalNAc-ser/thr) carbohydrate structure. Here we report that T-antigen expression, instead, correlates with elevated Golgi pH in cancer cells. Firstly, using a Golgi-targeted green fluorescent protein (GT-EGFP) as a probe, we show that the medial/trans-Golgi pH (pHG) in a high proportion of breast (MCF-7) and colorectal (HT-29, SW-48) cancer cells is significantly more alkaline (pHG > or = 6.75) than that of control cells (pHG 5.9-6.5). The pH gradient between the cytoplasm and the Golgi lumen is also markedly reduced in MCF-7 cells, suggesting a Golgi acidification defect. Secondly, we show that T-antigen expression is highly sensitive to changes in Golgi pH, as only a 0.2 pH unit increase was sufficient to increase T-antigen expression in control cells. Thirdly, we found that T-antigen expressing MCF-7 cells have 0.3 pH units more alkaline Golgi pH than non-expressing MCF-7 cells. Fourthly, in all cell types examined, we observed significant correlation between the number of T-antigen expressing cells and cells with a markedly elevated Golgi pH (pHG > or = 6.75). Consistent with these observations in cultured cells, cells in solid tumors also heterogenously expressed the T-antigen. Thus, elevated Golgi pH appears to be directly linked to T-antigen expression in cancer cells, but it may also act as a more general factor for altered glycosylation in cancer by affecting the distribution of Golgi-localized glycosyltransferases.


Subject(s)
Antigens, Neoplasm/physiology , Antigens, Tumor-Associated, Carbohydrate/physiology , Breast Neoplasms/physiopathology , Colorectal Neoplasms/physiopathology , Golgi Apparatus/physiology , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , Antigens, Tumor-Associated, Carbohydrate/analysis , Antigens, Tumor-Associated, Carbohydrate/genetics , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Line, Tumor , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , Flow Cytometry , Gene Expression Regulation, Neoplastic , Glycosylation , Glycosyltransferases/analysis , Golgi Apparatus/enzymology , Humans , Hydrogen-Ion Concentration , Immunohistochemistry , Microscopy, Fluorescence , Polysaccharides/metabolism
10.
J Biol Chem ; 279(38): 39982-8, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15258139

ABSTRACT

Aberrant secretion of lysosomal hydrolases such as (pro)cathepsin D (proCD) is a common phenotypic change in many human cancers. Here we explore the underlying molecular defect(s) and find that MCF-7 breast and CaCo-2 colorectal cancer cells that are unable to acidify their endosomal compartments secreted higher amounts of proCD than did acidification-competent cancer cell types. The latter secreted equivalent amounts of proCD only after dissipation of their organellar pH gradients with NH(4)Cl. Assessing the critical steps that resulted in proCD secretion revealed that the Golgi-associated sorting receptor for CD, i.e. the cation-independent mannose-6-phosphate receptor (MPR300), was aberrantly distributed in acidification-defective MCF-7 cells. It accumulated mainly in late endosomes and/or lysosomes as a complex with its ligand (proCD or intermediate CD), as evidenced by its co-localization with both CD and LAMP-2, a late endosome/lysosome marker. Our immunoprecipitation analyses also showed that MCF-7 cells possessed 7-fold higher levels of receptor-enzyme complexes than did acidification-competent cells. NH(4)Cl induced similar receptor redistribution into LAMP-2-positive structures in acidification-competent cells but not in MCF-7 cells. The receptor also recovered its normal Golgi localization upon drug removal. Based on these observations, we conclude that defective acidification results in the aberrant secretion of proCD in certain cancer cells and interferes mainly with the normal disassembly of the receptor-enzyme complexes and efficient receptor reutilization in the Golgi.


Subject(s)
Acids/metabolism , Cathepsin D/metabolism , Golgi Apparatus/metabolism , Ammonium Chloride/pharmacology , Animals , Antigens, CD/metabolism , Breast Neoplasms , COS Cells , Caco-2 Cells , Diuretics/pharmacology , Endosomes/drug effects , Endosomes/metabolism , Golgi Apparatus/drug effects , HT29 Cells , Humans , Lysosomal Membrane Proteins , Lysosomes/drug effects , Lysosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...