Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Curr Mol Med ; 15(9): 836-41, 2015.
Article in English | MEDLINE | ID: mdl-26511709

ABSTRACT

Cancer cell cannibalism is currently defined as a phenomenon in which an ensemble of a larger cell containing a smaller one, often in a big cytoplasmic vacuole, is detected in either cultured tumor cells or a tumor sample. After almost one century of considering this phenomenon as a sort of neglected curiosity, some recent studies have first proposed tumor cell cannibalism as a sort of "aberrant phagocytosis", making malignant cells very similar to professional phagocytes. Later, further research has shown that, differently to macrophages, exclusively ingesting exogenous material, apoptotic bodies, or cell debris, tumor cells are able to engulf other cells, including lymphocytes and erythrocytes, either dead or alive, with the main purpose to feed on them. This phenomenon has been associated to the malignancy of tumors, mostly exclusive of metastatic cells, and often associated to poor prognosis. The cannibalistic behavior increased depending on the microenvironmental condition of tumor cells, such as low nutrient supply or low pH, suggesting its key survival option for malignant cancers. However, the evidence that malignant cells may cannibalize tumor-infiltrating lymphocytes that act as their killers, suggests that tumor cell cannibalism could be a very direct and efficient way to neutralize immune response, as well. Tumor cell cannibalism may represent a sign of regression to a simpler, ancestral or primeval life style, similar to that of unicellular microorganisms, such as amoebas, where the goal is to survive and propagate in an overcrowded and very hostile microenvironment. In fact, we discovered that metastatic melanoma cells share with amoebas a transmembrane protein TM9SF4, indeed related to the cannibal behavior of these cells. This review attempts to provide a comprehensive description of the current knowledge about the role of TM9SF4 in cancer, highlighting its role as a key player in the cannibal behavior of malignant cancer cells. Moreover, we discuss differences and similarities between tumor cannibalism, entosis, phagocytosis and emperipolesis.


Subject(s)
Cytophagocytosis , Neoplasms/pathology , Amoeba/metabolism , Animals , Cell Survival , Emperipolesis , Entosis , Humans , Membrane Proteins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Phagocytosis
2.
Oncogene ; 34(40): 5163-74, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25659576

ABSTRACT

An inverted pH gradient across the cell membranes is a typical feature of malignant cancer cells that are characterized by extracellular acidosis and cytosol alkalization. These dysregulations are able to create a unique milieu that favors tumor progression, metastasis and chemo/immune-resistance traits of solid tumors. A key event mediating tumor cell pH alterations is an aberrant activation of ion channels and proton pumps such as (H+)-vacuolar-ATPase (V-ATPase). TM9SF4 is a poorly characterized transmembrane protein that we have recently shown to be related to cannibal behavior of metastatic melanoma cells. Here, we demonstrate that TM9SF4 represents a novel V-ATPase-associated protein involved in V-ATPase activation. We have observed in HCT116 and SW480 colon cancer cell lines that TM9SF4 interacts with the ATP6V1H subunit of the V-ATPase V1 sector. Suppression of TM9SF4 with small interfering RNAs strongly reduces assembly of V-ATPase V0/V1 sectors, thus reversing tumor pH gradient with a decrease of cytosolic pH, alkalization of intracellular vesicles and a reduction of extracellular acidity. Such effects are associated with a significant inhibition of the invasive behavior of colon cancer cells and with an increased sensitivity to the cytotoxic effects of 5-fluorouracil. Our study shows for the first time the important role of TM9SF4 in the aberrant constitutive activation of the V-ATPase, and the development of a malignant phenotype, supporting the potential use of TM9SF4 as a target for future anticancer therapies.


Subject(s)
Colonic Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , Membrane Proteins/metabolism , Neoplasm Invasiveness/pathology , Vacuolar Proton-Translocating ATPases/metabolism , Blotting, Western , Cell Line, Tumor , Colonic Neoplasms/metabolism , Fluorescence Resonance Energy Transfer , Humans , Hydrogen-Ion Concentration , Immunoprecipitation , Microscopy, Confocal , Polymerase Chain Reaction , Transfection
3.
Cell Death Dis ; 1: e87, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-21368860

ABSTRACT

Proton pump inhibitors (PPI) target tumour acidic pH and have an antineoplastic effect in melanoma. The PPI esomeprazole (ESOM) kills melanoma cells through a caspase-dependent pathway involving cytosolic acidification and alkalinization of tumour pH. In this paper, we further investigated the mechanisms of ESOM-induced cell death in melanoma. ESOM rapidly induced accumulation of reactive oxygen species (ROS) through mitochondrial dysfunctions and involvement of NADPH oxidase. The ROS scavenger N-acetyl-L-cysteine (NAC) and inhibition of NADPH oxidase significantly reduced ESOM-induced cell death, consistent with inhibition of cytosolic acidification. Autophagy, a cellular catabolic pathway leading to lysosomal degradation and recycling of proteins and organelles, represents a defence mechanism in cancer cells under metabolic stress. ESOM induced the early accumulation of autophagosomes, at the same time reducing the autophagic flux, as observed by WB analysis of LC3-II accumulation and by fluorescence microscopy. Moreover, ESOM treatment decreased mammalian target of rapamycin signalling, as reduced phosphorylation of p70-S6K and 4-EBP1 was observed. Inhibition of autophagy by knockdown of Atg5 and Beclin-1 expression significantly increased ESOM cytotoxicity, suggesting a protective role for autophagy in ESOM-treated cells. The data presented suggest that autophagy represents an adaptive survival mechanism to overcome drug-induced cellular stress and cytotoxicity, including alteration of pH homeostasis mediated by proton pump inhibition.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy , Esomeprazole/therapeutic use , Melanoma/drug therapy , Oxidative Stress , Proton Pump Inhibitors/therapeutic use , Acetylcysteine/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Cell Cycle Proteins , Cell Line, Tumor , Humans , Hydrogen-Ion Concentration , Melanoma/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , NADPH Oxidases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
4.
Oncogene ; 25(24): 3357-64, 2006 Jun 08.
Article in English | MEDLINE | ID: mdl-16462768

ABSTRACT

Activating BRAF or NRAS mutations have been found in 80% of human sporadic melanomas, but only one of these genetic alterations could be detected in each tumour. This suggests that BRAF and NRAS 'double mutants' may not provide advantage for tumour growth, or may even be selected against during tumorigenesis. However, by applying mutant-allele-specific-amplification-PCR method to short-term melanoma lines, one out of 14 tumours was found to harbour both BRAFV600E and the activating NRASQ61R mutations. On the other hand, analysis of 21 melanoma clones isolated by growth in soft agar from this tumour indicated that 16/21 clones harboured a BRAFV600E, but were wild-type for NRAS, whereas the remaining had the opposite genotype (NRASQ61R/wild-type BRAF). When compared to BRAFV600E clones, NRASQ61R clones displayed reduced growth in soft agar, but higher proliferative ability in vitro in liquid medium and even in vivo after grafting into SCID/SCID mice. These data suggest that NRAS and BRAF activating mutations can coexist in the same melanoma, but are mutually exclusive at the single-cell level. Moreover, the presence of NRASQ61R or BRAFV600E is associated with distinct in vitro and in vivo growth properties of neoplastic cells.


Subject(s)
Genes, ras , Melanoma/genetics , Melanoma/metabolism , Mutation , Proto-Oncogene Proteins B-raf/genetics , Animals , Base Sequence , Cell Line, Tumor , Female , Genotype , Humans , Mice , Mice, SCID , Molecular Sequence Data , Neoplasm Transplantation
5.
Apoptosis ; 10(5): 941-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16151629

ABSTRACT

The acquisition of a cell polarity is a crucial requirement for a number of cellular functions, including apoptosis. Cell polarization is an actin cytoskeleton-driven process, through a connection between actin and an increasing number of membrane proteins. The major actors in this connection are ezrin, radixin and moesin, a family of proteins with a high level of homology. Their structure includes an epitope that links to membrane proteins and the other that binds to the actin molecule. In this review we discuss recent data showing that the Fas linkage to the actin cytoskeleton is ezrin mediated and it is an essential requirement for susceptibility to the Fas-mediated apoptosis. The ezrin region responsible of Fas binding consists of 18 aminoacids mapped on the median lobe of the ezrin FERM domain. This binding is specific and of key importance in the control of cell homeostasis. Moreover, Fas-ezrin co-localization, ezrin phosphorylation and early acquisition of susceptibility to Fas-mediated apoptosis, may have a role in some human diseases in which programmed cell death seems to be a central pathogenetic mechanism, such as AIDS.


Subject(s)
Apoptosis/physiology , Cell Polarity/physiology , Phosphoproteins/physiology , fas Receptor/physiology , Animals , Apoptosis/drug effects , Blood Proteins/physiology , Cytoskeletal Proteins/physiology , Cytoskeleton/physiology , Humans , Membrane Microdomains/physiology , Membrane Proteins/physiology , Microfilament Proteins/physiology , Neurofibromin 2/physiology , Phosphorylation , Signal Transduction
6.
Cell Death Differ ; 11(5): 574-82, 2004 May.
Article in English | MEDLINE | ID: mdl-14739941

ABSTRACT

CD95(APO-1/Fas)-mediated apoptosis of bystander uninfected T cells exerts a major role in the HIV-1-mediated CD4+ T-cell depletion. HIV-1 gp120 has a key role in the induction of sensitivity of human lymphocytes to CD95-mediated apoptosis through its interaction with the CD4 receptor. Recently, we have shown the importance of CD95/ezrin/actin association in CD95-mediated apoptosis. In this study, we explored the hypothesis that the gp120-mediated CD4 engagement could be involved in the induction of susceptibility of primary human T lymphocytes to CD95-mediated apoptosis through ezrin phosphorylation and ezrin-to-CD95 association. Here, we show that gp120/IL-2 combined stimuli, as well as the direct CD4 triggering, on human primary CD4(+)T lymphocytes induced an early and stable ezrin activation through phosphorylation, consistent with the induction of ezrin/CD95 association and susceptibility to CD95-mediated apoptosis. Our results provide a new mechanism through which HIV-1-gp120 may predispose resting CD4(+)T cell to bystander CD95-mediated apoptosis and support the key role of ezrin/CD95 linkage in regulating susceptibility to CD95-mediated apoptosis.


Subject(s)
Apoptosis/physiology , HIV Envelope Protein gp120/toxicity , Phosphoproteins/metabolism , T-Lymphocytes/metabolism , fas Receptor/metabolism , Apoptosis/drug effects , CD4 Antigens/metabolism , Cytoskeletal Proteins , Humans , Interleukin-2/metabolism , Lymphocyte Activation/physiology , Phosphorylation
7.
EMBO J ; 19(19): 5123-34, 2000 Oct 02.
Article in English | MEDLINE | ID: mdl-11013215

ABSTRACT

CD95 (APO-1/Fas) is a member of the tumor necrosis factor receptor family, which can trigger apoptosis in a variety of cell types. However, little is known of the mechanisms underlying cell susceptibility to CD95-mediated apoptosis. Here we show that human T cells that are susceptible to CD95-mediated apoptosis, exhibit a constitutive polarized morphology, and that CD95 colocalizes with ezrin at the site of cellular polarization. In fact, CD95 co-immunoprecipitates with ezrin exclusively in lymphoblastoid CD4(+) T cells and primary long-term activated T lymphocytes, which are prone to CD95-mediated apoptosis, but not in short-term activated T lymphocytes, which are refractory to the same stimuli, even expressing equal levels of CD95 on the cell membrane. Pre-treatment with ezrin antisense oligonucleotides specifically protected from the CD95-mediated apoptosis. Moreover, we show that the actin cytoskeleton integrity is essential for this function. These findings strongly suggest that the CD95 cell membrane polarization, through an ezrin-mediated association with the actin cytoskeleton, is a key intracellular mechanism in rendering human T lymphocytes susceptible to the CD95-mediated apoptosis.


Subject(s)
Actin Cytoskeleton/metabolism , Apoptosis/physiology , Phosphoproteins/metabolism , T-Lymphocytes/cytology , fas Receptor/metabolism , Blood Proteins/metabolism , Blotting, Western , Cell Polarity , Cells, Cultured , Cytoskeletal Proteins/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Microscopy, Electron, Scanning , Microscopy, Video , Oligonucleotides, Antisense , Protein Binding , T-Lymphocytes/metabolism , fas Receptor/physiology
8.
Int J Cancer ; 87(4): 569-73, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10918199

ABSTRACT

Severe combined immunodeficient (SCID) mice generally do not reject allogeneic or xenogeneic organ grafts and represent a unique model for investigating in vivo the behaviour of both normal and neoplastic human cells. However, cells from human primary tumors often do not grow in SCID mice. We have previously shown that the major reaction of SCID mice to the engraftment of human peripheral blood leukocytes is a massive granulocyte recruitment into the site of transplantation. In this study, we have investigated the role of murine granulocytes in the control of human tumor cell growth in SCID mice. We report here that murine granulocytes infiltrate and delimit the human tumor mass and that treatment of SCID mice with anti-murine granulocyte antibody markedly improves the growth of human tumor cell lines of different origin through suppression of the host granulocyte reaction. This finding provides a new tool for improving the human tumor take in SCID mice, thus opening new perspectives for a practical in vivo preclinical test of anti-tumor strategies. Moreover, this study, even with the limits of the known natural reaction against xenotransplants, further supports the importance of granulocytes in the control of tumor take and growth.


Subject(s)
Granulocytes/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Division/physiology , Female , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Neutrophil Infiltration/immunology , Tumor Cells, Cultured
9.
Histol Histopathol ; 15(2): 539-49, 2000 04.
Article in English | MEDLINE | ID: mdl-10809375

ABSTRACT

Asymmetric organization of the plasma membrane and cytosolic organelles is fundamental for a variety of cells, including bacteria, yeast and eukaryotic cells (Nelson, 1992). The degree into which cells polarize is characterized by their ability to create and maintain morphologically and biochemically distinct plasma membrane domains. The generation and maintenance of polarized distribution of membrane components (proteins and lipids) is thus critical to the ability of cells to perform complex activities such as cell-to-cell interactions, vectorial transport and secretion, cellular immunity, development and morphogenesis. Modification of cellular polarity may potentially lead to abnormal cellular activities and various pathological disorders (Molitoris, 1991; Carone et al., 1994; Chen et al., 1995). Our review shows the complex interplay between membrane proteins and the cytoskeletal network in determining the "polarized phenotype" in the cell. We provide evidence that membrane/cytoskeleton interaction is the key to regulation of the vast majority of cellular functions.


Subject(s)
Actins/metabolism , Cytoskeleton/physiology , Membrane Proteins/metabolism , Animals , Cell Membrane/metabolism , Cytoskeleton/metabolism , Humans
10.
J Natl Cancer Inst ; 88(7): 419-29, 1996 Apr 03.
Article in English | MEDLINE | ID: mdl-8618233

ABSTRACT

BACKGROUND: Phosphorothioate oligodeoxynucleotides ([S]ODNs) contain a modified internucleoside phosphate backbone. Antisense [S]ODNs targeted to specific oncogenes have been used with some therapeutic success in animal models human leukemia; however, the potential for antisense [S]ODN treatment of solid tumors has only recently been explored. PURPOSE: We evaluated the effects of antisense [S]ODNs targeted to the c-myc oncogene on the proliferation of human melanoma cells in vitro and on the growth of human melanoma xenografts in CD-1 nude (nu/nu) mice, METHODS: The effects of 15-mer [S]ODNs containing c-myc sense, c-myc antisense, and two different scrambled sequences on the proliferation and viability of cultures of three established human melanoma cell lines (M14, JR8, and PLF2) were determined by measuring cell numbers and use of the trypan blue exclusion test. The induction of apoptosis in these cells following treatment with [S]ODNs was evaluated by fluorescence-activated cell sorter (FACS) analysis. FACS analysis was also used to determine the effects of [S]ODN treatment on the proliferation of primary cultures of a human melanoma explant (NG cells). The expression of c-Myc protein in cultured NG cells after treatment with [S]ODNs was examined by western blot analysis. The antitumor activity and the toxic effects of several [S]ODN treatment regimens were monitored by measuring differences in tumor weight (percent tumor weight inhibition), tumor growth rate (tumor growth inhibition), animal lifespan (percent increase in lifespan), the number of toxic deaths and the median number of long metastases in treated and control mice bearing NG xenografts. c-Myc protein expression in NG tumor cells following [S]ODN treatment was evaluated by FACS analysis, and the extent of apoptosis in these cells was determined by FACS analysis and morphologic examination. RESULTS: Treatment with antisense [S]ODNs, but not the others, inhibited the growth of all tested melanoma cultures in vitro; FACS analysis revealed that growth inhibition was associated with the induction of apoptosis. Antisense [S]ODN treatment also led to reduced celluLar levels of c-Myc protein. In vivo, [S]ODN antitumor activity and toxicity were dose and schedule dependent; however, only antisense [S]ODNs exhibited antitumor activity. Mice bearing NG xenografts treated with antisense [S]ODNs showed a marked inhibition of tumor growth, a reduction in the number of long metastases, and an increase in life span. Reduced levels of c-Myc protein and increased levels of apoptosis were also observed in NG tumor cells following antisense [S]ODN treatment. CONCLUSIONS: treatment of human melanoma cells and solid tumors with antisense [S]ODNs targeted to c-Myc inhibits their growth and is associated with the induction of apoptosis.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Genes, myc , Melanoma/drug therapy , Oligonucleotides, Antisense/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Animals , Apoptosis/drug effects , Base Sequence , Blotting, Western , Cell Division/drug effects , Drug Administration Schedule , Flow Cytometry , Humans , Melanoma/pathology , Mice , Mice, Nude , Molecular Sequence Data , Proto-Oncogene Proteins c-myc/biosynthesis , RNA, Messenger/metabolism , Thionucleotides , Time Factors , Transplantation, Heterologous , Tumor Cells, Cultured
11.
Cancer Immunol Immunother ; 41(6): 375-83, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8635195

ABSTRACT

The antitumor and antimetastatic activity of dacarbazine (DTIC) alone or in combination with cyclophosphamide (CY) was tested in C57BL/6 mice bearing Lewis lung carcinoma (3LL). Treatment with both agents significantly reduced tumor growth and the number of metastases. These effects were associated with marked changes of the biochemical and immunological properties of drug-treated 3LL cells, i.e. (a) reduction of alpha 6 integrin expression, (b) increased susceptibility to natural immunity in vivo, as measured in terms of rapid clearance from mouse lungs of prelabeled 3LL cells injected i.v. and (c) increased immunogenicity, as assessed by T-cell-mediated immune responses (i.e. graft rejection by intact syngeneic mice, and frequency of specific CTL precursors recognizing DTIC/CY-treated cancer cells). The immunotherapeutic advantage afforded by increased immunosensitivity and immunogenicity of 3LL cells exposed to DTIC + CY appears to be markedly reduced in vivo by the profound immunodepressive effects of these drugs. Within this context, addition of interleukin-2 was found to increase the antitumor and antimetastatic activity of this chemotherapeutic regimen. The present study shows, for the first time in a solid tumor model, that a biological response modifier increases the antitumor efficacy of drugs that are able to affect the immunological properties of cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/therapy , Immunologic Factors/therapeutic use , Interleukin-2/therapeutic use , Lung Neoplasms/therapy , Neoplasm Metastasis/prevention & control , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/pharmacology , Carcinoma/drug therapy , Carcinoma/secondary , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Cyclophosphamide/pharmacology , Cytotoxicity, Immunologic/drug effects , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Drug Administration Schedule , Drug Screening Assays, Antitumor , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Immunologic Factors/pharmacology , Integrin alpha6 , Interleukin-2/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...