Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Cell Mol Life Sci ; 78(3): 853-865, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32940721

ABSTRACT

Adaptive antitumor immune responses, either cellular or humoral, aim at eliminating tumor cells expressing the cognate antigens. There are some instances, however, where these same immune responses have tumor-promoting effects. These effects can lead to the expansion of antigen-negative tumor cells, tumor cell proliferation and tumor growth, metastatic dissemination, resistance to antitumor therapy and apoptotic stimuli, acquisition of tumor-initiating potential and activation of various forms of survival mechanisms. We describe the basic mechanisms that underlie tumor-promoting adaptive immune responses and try to identify the variables that induce the switching of a tumor-inhibitory, cellular or humoral immune response, into a tumor-promoting one. We suggest that tumor-promoting adaptive immune responses may be at the origin of at least a fraction of hyperprogressive diseases (HPD) that are observed in cancer patients during therapy with immune checkpoint inhibitors (ICI) and, less frequently, with single-agent chemotherapy. We also propose the use of non-invasive biomarkers allowing to predict which patients may undergo HPD during ICI and other forms of antitumor therapy. Eventually, we suggest possibilities of therapeutic intervention allowing to inhibit tumor-promoting adaptive immune responses.


Subject(s)
Adaptive Immunity , Neoplasms/pathology , Antibodies/immunology , Antibodies/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Cytokines/metabolism , Humans , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
Biochim Biophys Acta Rev Cancer ; 1868(2): 571-583, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29056539

ABSTRACT

Inhibitory or stimulatory immune checkpoint molecules are expressed on a sizeable fraction of tumor cells in different tumor types. It was thought that the main function of tumor cell-associated immune checkpoint molecules would be the modulation (down- or upregulation) of antitumor immune responses. In recent years, however, it has become clear that the expression of immune checkpoint molecules on tumor cells has important consequences on the biology of the tumor cells themselves. In particular, a causal relationship between the expression of these molecules and the acquisition of malignant traits has been demonstrated. Thus, immune checkpoint molecules have been shown to promote the epithelial-mesenchymal transition of tumor cells, the acquisition of tumor-initiating potential and resistance to apoptosis and antitumor drugs, as well as the propensity to disseminate and metastasize. Herein, we review this evidence, with a main focus on PD-L1, the most intensively investigated tumor cell-associated immune checkpoint molecule and for which most information is available. Then, we discuss more concisely other tumor cell-associated immune checkpoint molecules that have also been shown to induce the acquisition of malignant traits, such as PD-1, B7-H3, B7-H4, Tim-3, CD70, CD28, CD137, CD40 and CD47. Open questions in this field as well as some therapeutic approaches that can be derived from this knowledge, are also addressed.


Subject(s)
B7-H1 Antigen/physiology , Neoplasms/etiology , Animals , B7 Antigens/physiology , CD47 Antigen/physiology , Epithelial-Mesenchymal Transition , Humans , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/physiology , Programmed Cell Death 1 Receptor/physiology , Proto-Oncogene Proteins c-akt/physiology , TOR Serine-Threonine Kinases/physiology , Tumor Microenvironment , V-Set Domain-Containing T-Cell Activation Inhibitor 1/physiology
3.
J Immunol ; 196(2): 902-14, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26673138

ABSTRACT

Human colorectal cancer (CRC) is a frequent neoplasia in Western countries, and its metastatic progression is a major cause of cancer-related death. In search of specific molecules upregulated in CRC, with possible clinical relevance, we performed a differential gene-profiling analysis in surgery-derived CRC samples and adjacent uninvolved intestinal mucosa. The chemokine CX3CL1 and its specific receptor CX3CR1 were significantly upregulated in tumors. Higher expression of CX3CL1 and CX3CR1 was confirmed by immunohistochemistry in 100 CRC tumor samples (stages I-III). Unexpectedly, high immune scores of CX3CL1 did not correlate with the density of tumor-infiltrating CD3(+) T cells or CD68(+) macrophages. Coexpression of ligand and receptor by tumor cells (axis-positive tumors) significantly associated with longer disease-free (p = 0.01) and disease-specific survival (p = 0.001). Conversely, axis-negative tumors (with low expression of both ligand and receptor) had increased risk of tumor relapse (p = 0.02), and increased likelihood of metachronous metastasis (p = 0.001), including after stage adjustment (p = 0.006). Transduction of CX3CL1 and CX3CR1 in CRC tumor cell lines induced cell aggregation that strongly inhibited in vitro migration in chemotaxis assays. In a mouse model of spleen-liver metastases, cancer dissemination to liver was dramatically reduced in CX3CL1-CX3CR1-expressing tumors, and ligand-receptor interaction was confirmed in cancer cells in vivo by fluorescence resonance energy transfer analysis. In conclusion, tumoral expression of the CX3CL1-CX3CR1 chemokine axis functions as a retention factor, increasing homotypic cell adhesion and limiting tumor spreading to metastatic sites. Lack or low levels of expression of CX3CL1-CX3CR1 by tumor cells identifies a group of CRC patients at increased risk of metastatic progression.


Subject(s)
Chemokine CX3CL1/biosynthesis , Colorectal Neoplasms/pathology , Neoplasm Invasiveness/pathology , Receptors, Chemokine/biosynthesis , Animals , CX3C Chemokine Receptor 1 , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , Disease-Free Survival , Female , Fluorescence Resonance Energy Transfer , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Nude , Neoplasm Metastasis , Polymerase Chain Reaction , Prognosis , Proportional Hazards Models , Transcriptome
4.
Mol Cancer ; 16(1): 3, 2017 01 30.
Article in English | MEDLINE | ID: mdl-28137290

ABSTRACT

Epithelial-mesenchymal transition (EMT) and cancer stem-like cells (CSC) are becoming highly relevant targets in anticancer drug discovery. A large body of evidence suggests that epithelial-mesenchymal transitioned tumor cells (EMT tumor cells) and CSCs have similar functions. There is also an overlap regarding the stimuli that can induce the generation of EMT tumor cells and CSCs. Moreover, direct evidence has been brought that EMT can give rise to CSCs. It is unclear however, whether EMT tumor cells should be considered CSCs or if they have to undergo further changes. In this article we summarize available evidence suggesting that, indeed, additional programs must be engaged and we propose that macroautophagy (hereafter, autophagy) represents a key trait distinguishing CSCs from EMT tumor cells. Thus, CSCs have often been reported to be in an autophagic state and blockade of autophagy inhibits CSCs. On the other hand, there is ample evidence showing that EMT and autophagy are distinct events. CSCs, however, represent, by themselves, a heterogeneous population. Thus, CSCs have been distinguished in predominantly non-cycling and cycling CSCs, the latter representing CSCs that self-renew and replenish the pool of differentiated tumor cells. We now suggest that the non-cycling CSC subpopulation is in an autophagic state. We propose also two models to explain the relationship between EMT tumor cells and these two major CSC subpopulations: a branching model in which EMT tumor cells can give rise to cycling or non-cycling CSCs, respectively, and a hierarchical model in which EMT tumor cells are first induced to become autophagic CSCs and, subsequently, cycling CSCs. Finally, we address the therapeutic consequences of these insights.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/pathology , Autophagy , Cell Communication , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells/metabolism
5.
Neural Plast ; 2017: 6468356, 2017.
Article in English | MEDLINE | ID: mdl-28367336

ABSTRACT

The c-Jun N-terminal kinase (JNK) is part of a stress signalling pathway strongly activated by NMDA-stimulation and involved in synaptic plasticity. Many studies have been focused on the post-synaptic mechanism of JNK action, and less is known about JNK presynaptic localization and its physiological role at this site. Here we examined whether JNK is present at the presynaptic site and its activity after presynaptic NMDA receptors stimulation. By using N-SIM Structured Super Resolution Microscopy as well as biochemical approaches, we demonstrated that presynaptic fractions contained significant amount of JNK protein and its activated form. By means of modelling design, we found that JNK, via the JBD domain, acts as a physiological effector on T-SNARE proteins; then using biochemical approaches we demonstrated the interaction between Syntaxin-1-JNK, Syntaxin-2-JNK, and Snap25-JNK. In addition, taking advance of the specific JNK inhibitor peptide, D-JNKI1, we defined JNK action on the SNARE complex formation. Finally, electrophysiological recordings confirmed the role of JNK in the presynaptic modulation of vesicle release. These data suggest that JNK-dependent phosphorylation of T-SNARE proteins may have an important functional role in synaptic plasticity.


Subject(s)
Cerebral Cortex/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Presynaptic Terminals/enzymology , Receptors, N-Methyl-D-Aspartate/metabolism , SNARE Proteins/metabolism , Animals , Cerebral Cortex/physiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials , Female , Glycine/pharmacology , Male , Mice , Mitogen-Activated Protein Kinase 10/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , N-Methylaspartate/pharmacology , Synaptosomes/metabolism
6.
Mol Cancer ; 13: 23, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24495286

ABSTRACT

BACKGROUND: Recent studies demonstrated that engagement of sodium glucose transporter 1 (SGLT-1) by orally administered D-glucose protects the intestinal mucosa from lipopolysaccharide (LPS)-induced injury. We tested whether SGLT-1 engagement might protect the intestinal mucosa from doxorubicin (DXR)- and 5-fluorouracil (5-FU)-induced injury in animal models mimicking acute or chronic mucositis. METHODS: Mice were treated intraperitoneally with DXR, alone or in combination with 5-FU, and orally with BLF501, a glucose-derived synthetic compound with high affinity for SGLT-1. Intestinal mucosal epithelium integrity was assessed by histological analysis, cellular proliferation assays, real-time PCR gene expression assays and Western blot assays. Student's t-test (paired two-tailed) and χ2 analyses were used for comparisons between groups. Differences were considered significant at p < 0.05. RESULTS: BLF501 administration in mice treated with DXR and/or 5-FU decreased the injuries to the mucosa in terms of epithelial integrity and cellular proliferative ability. Co-treatment with BLF501 led to a normal expression and distribution of both zonula occludens-1 (ZO-1) and beta-catenin, which were underexpressed after treatment with either chemotherapeutic agent alone. BLF501 administration also restored normal expression of caspase-3 and ezrin/radixin/moesin (ERM), which were overexpressed after treatment with DXR and 5-FU. In SGLT1-/- mice, BLF501 had no detectable effects. BLF501 administration in wild-type mice with growing A431 tumors did not modify antitumor activity of DXR. CONCLUSIONS: BLF501-induced protection of the intestinal mucosa is a promising novel therapeutic approach to reducing the severity of chemotherapy-induced mucositis.


Subject(s)
Antineoplastic Agents/toxicity , Gastrointestinal Diseases/chemically induced , Glucose/analogs & derivatives , Mucositis/chemically induced , Sodium-Glucose Transporter 1/agonists , Animals , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , Doxorubicin/toxicity , Female , Fluorescent Antibody Technique , Fluorouracil/toxicity , Gastrointestinal Diseases/pathology , Gastrointestinal Diseases/prevention & control , Glucose/pharmacology , Heterografts , Humans , Immunohistochemistry , Ligands , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Mucositis/pathology , Mucositis/prevention & control , Real-Time Polymerase Chain Reaction , Transcriptome/drug effects
7.
Discov Med ; 36(185): 1109-1126, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38926098

ABSTRACT

Aerobic glycolysis, i.e., non-oxidative glycolysis occurring under aerobic conditions (the so-called Warburg effect) is now recognized as a hallmark of cancer. However, evidence increasingly indicates that upregulated oxidative metabolism is also pivotal in tumorigenesis. In this article, we discuss factors that upregulate oxidative metabolism in tumor cells. These factors are associated with tumor cell-intrinsic and -extrinsic stimuli including antitumor drugs, requirements related to the different steps of tumorigenesis (initiation and acquisition of cancer stem-like cell functions, primary tumor growth, quiescence, metastatic dissemination), factors related to the phenotypic changes of tumor cells (e.g., autophagy and epithelial-mesenchymal transition), and particular metabolic requirements of proliferating tumor cells. In this context, we also discuss drug resistance associated with upregulated oxidative metabolism. We conclude by proposing a model whereby these factors, either individually or in combination, promote upregulation of oxidative metabolism. In the following, we address some mechanistic aspects that underlie the upregulation of oxidative metabolism and discuss the consequences on tumor prognosis. In the conclusion section of this article, we discuss possible therapeutic implications of the knowledge gathered in this field over the years.


Subject(s)
Carcinogenesis , Drug Resistance, Neoplasm , Neoplasms , Humans , Carcinogenesis/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/drug therapy , Oxidation-Reduction , Animals , Epithelial-Mesenchymal Transition , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Glycolysis , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
8.
Cancer Lett ; 585: 216661, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38309613

ABSTRACT

Inhibitory immune checkpoint (ICP) molecules are pivotal in inhibiting innate and acquired antitumor immune responses, a mechanism frequently exploited by cancer cells to evade host immunity. These evasion strategies contribute to the complexity of cancer progression and therapeutic resistance. For this reason, ICP molecules have become targets for antitumor drugs, particularly monoclonal antibodies, collectively referred to as immune checkpoint inhibitors (ICI), that counteract such cancer-associated immune suppression and restore antitumor immune responses. Over the last decade, however, it has become clear that tumor cell-associated ICPs can also induce tumor cell-intrinsic effects, in particular epithelial-mesenchymal transition (EMT) and macroautophagy (hereafter autophagy). Both of these processes have profound implications for cancer metastasis and drug responsiveness. This article reviews the positive or negative cross-talk that tumor cell-associated ICPs undergo with autophagy and EMT. We discuss that tumor cell-associated ICPs are upregulated in response to the same stimuli that induce EMT. Moreover, ICPs themselves, when overexpressed, become an EMT-inducing stimulus. As regards the cross-talk with autophagy, ICPs have been shown to either stimulate or inhibit autophagy, while autophagy itself can either up- or downregulate the expression of ICPs. This dynamic equilibrium also extends to the autophagy-apoptosis axis, further emphasizing the complexities of cellular responses. Eventually, we delve into the intricate balance between autophagy and apoptosis, elucidating its role in the broader interplay of cellular dynamics influenced by ICPs. In the final part of this article, we speculate about the driving forces underlying the contradictory outcomes of the reciprocal, inhibitory, or stimulatory effects between ICPs, EMT, and autophagy. A conclusive identification of these driving forces may allow to achieve improved antitumor effects when using combinations of ICIs and compounds acting on EMT and/or autophagy. Prospectively, this may translate into increased and/or broadened therapeutic efficacy compared to what is currently achieved with ICI-based clinical protocols.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Autophagy , Epithelial-Mesenchymal Transition , Antibodies, Monoclonal/pharmacology
9.
J Cell Physiol ; 228(1): 242-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22689438

ABSTRACT

Numerous investigations have found a relationship between higher risk of cancer and increased intake of fats, while results of clinical studies of fat reduction and breast cancer recurrence have been mixed. A diet completely free of fats cannot be easily administered to humans, but experimental studies in mice can be done to determine whether this extreme condition influences tumor development. Here, we examined the effects of a FA-free diet on mammary tumor development and growth rate in female FVB-neu proto-oncogene transgenic mice that develop spontaneous multifocal mammary tumors after a long latency period. Mice were fed a fatty acid-free diet beginning at 112, 35, and 30 days of age. In all these experiments, tumor appearance was delayed, tumor incidence was reduced and the mean number of palpable mammary tumors per mouse was lower, as compared to standard diet-fed mice. By contrast, tumor growth rate was unaffected in mice fed the fatty acid-free diet. Plasma of mice fed the fatty acid-free diet revealed significantly higher contents of oleic, palmitoleic and 20:3ω9 acids and lower contents of linoleic and palmitic acids. In conclusion, these findings indicate that a FA-free diet reduces tumor incidence and latency but not tumor growth rate, suggesting that a reduction in dietary FAs in humans may have a protective effect on tumorigenesis but not on tumors once they appear.


Subject(s)
Animal Feed/analysis , Fatty Acids/adverse effects , Mammary Neoplasms, Animal/prevention & control , Receptor, ErbB-2/metabolism , Aging , Animals , Diet , Fatty Acids/chemistry , Female , Mammary Neoplasms, Animal/diet therapy , Mice , Mice, Transgenic , Proto-Oncogene Mas , Receptor, ErbB-2/genetics
10.
Cells ; 12(8)2023 04 10.
Article in English | MEDLINE | ID: mdl-37190033

ABSTRACT

Reprogramming energy production from mitochondrial respiration to glycolysis is now considered a hallmark of cancer. When tumors grow beyond a certain size they give rise to changes in their microenvironment (e.g., hypoxia, mechanical stress) that are conducive to the upregulation of glycolysis. Over the years, however, it has become clear that glycolysis can also associate with the earliest steps of tumorigenesis. Thus, many of the oncoproteins most commonly involved in tumor initiation and progression upregulate glycolysis. Moreover, in recent years, considerable evidence has been reported suggesting that upregulated glycolysis itself, through its enzymes and/or metabolites, may play a causative role in tumorigenesis, either by acting itself as an oncogenic stimulus or by facilitating the appearance of oncogenic mutations. In fact, several changes induced by upregulated glycolysis have been shown to be involved in tumor initiation and early tumorigenesis: glycolysis-induced chromatin remodeling, inhibition of premature senescence and induction of proliferation, effects on DNA repair, O-linked N-acetylglucosamine modification of target proteins, antiapoptotic effects, induction of epithelial-mesenchymal transition or autophagy, and induction of angiogenesis. In this article we summarize the evidence that upregulated glycolysis is involved in tumor initiation and, in the following, we propose a mechanistic model aimed at explaining how upregulated glycolysis may play such a role.


Subject(s)
Glycolysis , Neoplasms , Humans , Cell Transformation, Neoplastic/metabolism , Neoplasms/metabolism , Autophagy , DNA Repair , Tumor Microenvironment
11.
J Cell Physiol ; 227(3): 1107-13, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21567398

ABSTRACT

The secretory activity of Paneth cells is related to the bacterial milieu in the small intestine; however, the molecules involved in inducing Paneth cell secretion of enzymes and antimicrobial peptides are not well-defined. Mice treated orally with CpG-oligodeoxynucleotide (ODN), an agonist of Toll-like receptor (TLR) 9, showed rapid and massive Paneth cell degranulation. CpG-ODN-induced degranulation was not observed in TLR9(-/-) mice or in chimeric TLR9(-/-) mice reconstituted with wild-type (WT) bone marrow, but was observed in WT mice reconstituted with TLR9(-/-) bone marrow, indicating a role for TLR9-expressing gastrointestinal cells in CpG recognition. The TLR3 agonist polyinosinic-polycytidylic acid also induced rapid degranulation, whereas the TLR4 and TLR5 agonists LPS and flagellin, respectively, induced late degranulation mediated by TNF-α. Our evidence that TLR9 and TLR3 agonists induce Paneth cell degranulation points to the need for further studies of the mechanisms underlying Paneth cell function as an avenue toward preventing infection and treating inflammatory bowel diseases.


Subject(s)
Cell Degranulation/physiology , Paneth Cells/metabolism , Toll-Like Receptor 3/physiology , Toll-Like Receptor 4/physiology , Toll-Like Receptor 5/physiology , Toll-Like Receptor 9/metabolism , Administration, Oral , Animals , Cell Degranulation/drug effects , Flagellin/pharmacology , Interleukin-17/metabolism , Jejunum/cytology , Ligands , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides/pharmacology , Paneth Cells/cytology , Toll-Like Receptor 3/agonists , Toll-Like Receptor 4/agonists , Toll-Like Receptor 5/agonists , Toll-Like Receptor 5/deficiency , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/physiology , Tumor Necrosis Factor-alpha/genetics
12.
Dig Dis Sci ; 57(5): 1227-37, 2012 May.
Article in English | MEDLINE | ID: mdl-22278339

ABSTRACT

BACKGROUND: Vasostatin-1 (VS-1), the N-terminal fragment of chromogranin A (CgA), decreases the permeability of endothelial cells in vitro and in vivo. AIMS: Here, we investigated whether a similar effect could be observed also on intestinal epithelial cells (IECs) in vitro and whether VS-1 could have favorable effects on animal models of acute or chronic colitis, which are characterized by increased permeability of the intestinal epithelium. METHODS: In vitro, VS-1 was tested on IEC monolayers showing increased permeability, on mechanically injured IEC monolayers, and on the production of the chemokine IL-8/KC by lipopolysaccharide (LPS)-stimulated IECs. In vivo, VS-1 was tested in animal models of dextran sodium salt (DSS)-induced acute or chronic colitis. RESULTS: In vitro, VS-1 inhibited increased permeability of IECs induced by interferon-γ and tumor necrosis factor-α. Moreover, VS-1 promoted healing of mechanically injured IEC monolayers, most likely through stimulation of cell migration, rather than cell proliferation. Eventually, VS-1 inhibited LPS-induced production of IL-8. In vivo, VS-1 exerted protective effects in animal models of acute or chronic colitis upon oral, but not systemic administration. CONCLUSIONS: VS-1 is therapeutically active in animal models of acute or chronic, DSS-induced colitis. The mechanisms underlying this effect are likely to be multiple, and may include inhibition of enhanced intestinal permeability, repair of injured intestinal mucosae, and inhibition of the production of IL-8/KC and possibly other inflammatory cytokines.


Subject(s)
Cell Membrane Permeability/drug effects , Chromogranin A , Colitis , Colon/metabolism , Epithelial Cells/metabolism , Peptide Fragments , Administration, Oral , Animals , Cell Line, Tumor , Cell Movement/drug effects , Chromogranin A/administration & dosage , Chromogranin A/pharmacokinetics , Chronic Disease , Colitis/metabolism , Colitis/pathology , Colon/drug effects , Colon/pathology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Humans , Interferon-gamma/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , Lipopolysaccharides/metabolism , Mice , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Protective Agents/administration & dosage , Protective Agents/pharmacokinetics , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism
13.
Cells ; 11(6)2022 03 18.
Article in English | MEDLINE | ID: mdl-35326492

ABSTRACT

Upregulation of glycolysis, induction of epithelial-mesenchymal transition (EMT) and macroautophagy (hereafter autophagy), are phenotypic changes that occur in tumor cells, in response to similar stimuli, either tumor cell-autonomous or from the tumor microenvironment. Available evidence, herein reviewed, suggests that glycolysis can play a causative role in the induction of EMT and autophagy in tumor cells. Thus, glycolysis has been shown to induce EMT and either induce or inhibit autophagy. Glycolysis-induced autophagy occurs both in the presence (glucose starvation) or absence (glucose sufficiency) of metabolic stress. In order to explain these, in part, contradictory experimental observations, we propose that in the presence of stimuli, tumor cells respond by upregulating glycolysis, which will then induce EMT and inhibit autophagy. In the presence of stimuli and glucose starvation, upregulated glycolysis leads to adenosine monophosphate-activated protein kinase (AMPK) activation and autophagy induction. In the presence of stimuli and glucose sufficiency, upregulated glycolytic enzymes (e.g., aldolase or glyceraldehyde 3-phosphate dehydrogenase) or decreased levels of glycolytic metabolites (e.g., dihydroxyacetone phosphate) may mimic a situation of metabolic stress (herein referred to as "pseudostarvation"), leading, directly or indirectly, to AMPK activation and autophagy induction. We also discuss possible mechanisms, whereby glycolysis can induce a mixed mesenchymal/autophagic phenotype in tumor cells. Subsequently, we address unresolved problems in this field and possible therapeutic consequences.


Subject(s)
AMP-Activated Protein Kinases , Epithelial-Mesenchymal Transition , AMP-Activated Protein Kinases/metabolism , Autophagy/genetics , Epithelial-Mesenchymal Transition/genetics , Glucose/metabolism , Glycolysis/genetics
14.
Immunol Cell Biol ; 89(5): 630-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21102537

ABSTRACT

The innate immune system is present throughout the female reproductive tract and functions in synchrony with the adaptive immune system to provide protection in a way that enhances the chances for fetal survival, while protecting against potential pathogens. Recent data show that activation of Toll-like receptor (TLR)2 and 4 by low-molecular weight hyaluronic acid (LMW-HA) in the epidermis induces secretion of the antimicrobial peptide ß-defensin 2. In the present work, we show that LMW-HA induces vaginal epithelial cells to release different antimicrobial peptides, via activation of TLR2 and TLR4. Further, we found that LMW-HA favors repair of vaginal epithelial injury, involving TLR2 and TLR4, and independently from its classical receptor CD44. This wound-healing activity of LMW-HA is dependent from an Akt/phosphatidylinositol 3 kinase pathway. Therefore, these findings suggest that the vaginal epithelium is more than a simple physical barrier to protect against invading pathogens: on the contrary, this surface acts as efficient player of innate host defense, which may modulate its antimicrobial properties and injury restitution activity, following LMW-HA stimulation; this activity may furnish an additional protective activity to this body compartment, highly and constantly exposed to microbiota, ameliorating the self-defense of the vaginal epithelium in both basal and pathological conditions.


Subject(s)
Adjuvants, Immunologic/pharmacology , Epithelium/drug effects , Hyaluronic Acid/pharmacology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Vagina/drug effects , Vagina/immunology , Cell Line, Transformed , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelium/immunology , Epithelium/metabolism , Female , Gene Expression Regulation, Archaeal/drug effects , Humans , Hyaluronic Acid/metabolism , Immunity, Innate , Immunologic Factors/immunology , Immunologic Factors/metabolism , Inflammation Mediators/metabolism , Ligands , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Wound Healing/drug effects , Wound Healing/genetics , beta-Defensins/genetics , beta-Defensins/metabolism
15.
Neoplasia ; 23(2): 234-245, 2021 02.
Article in English | MEDLINE | ID: mdl-33418276

ABSTRACT

Tumor cells often switch from mitochondrial oxidative metabolism to glycolytic metabolism even under aerobic conditions. Tumor cell glycolysis is accompanied by several nonenzymatic activities among which induction of drug resistance has important therapeutic implications. In this article, we review the main aspects of glycolysis-induced drug resistance. We discuss the classes of antitumor drugs that are affected and the components of the glycolytic pathway (transporters, enzymes, metabolites) that are involved in the induction of drug resistance. Glycolysis-associated drug resistance occurs in response to stimuli, either cell-autonomous (e.g., oncoproteins) or deriving from the tumor microenvironment (e.g., hypoxia or pseudohypoxia, mechanical cues, etc.). Several mechanisms mediate the induction of drug resistance in response to glycolytic metabolism: inhibition of apoptosis, induction of epithelial-mesenchymal transition, induction of autophagy, inhibition of drug influx and increase of drug efflux. We suggest that drug resistance in response to glycolysis comes into play in presence of qualitative (e.g., expression of embryonic enzyme isoforms, post-translational enzyme modifications) or quantitative (e.g., overexpression of enzymes or overproduction of metabolites) alterations of glycolytic metabolism. We also discern similarities between changes occurring in tumor cells in response to stimuli inducing glycolysis-associated drug resistance and those occurring in cells of the innate immune system in response to danger signals and that have been referred to as danger-associated metabolic modifications. Eventually, we briefly address that also mitochondrial oxidative metabolism may induce drug resistance and discuss the therapeutic implications deriving from the fact that the main energy-generating metabolic pathways may be both at the origin of antitumor drug resistance.


Subject(s)
Drug Resistance, Neoplasm , Glucose/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Energy Metabolism , Epithelial-Mesenchymal Transition/drug effects , Glycolysis , Humans , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/pathology , Oxidative Phosphorylation/drug effects
16.
Cells ; 10(4)2021 04 12.
Article in English | MEDLINE | ID: mdl-33921301

ABSTRACT

Antibodies against inhibitory immune checkpoint molecules (ICPMs), referred to as immune checkpoint inhibitors (ICIs), have gained a prominent place in cancer therapy. Several ICIs in clinical use have been engineered to be devoid of effector functions because of the fear that ICIs with preserved effector functions could deplete immune cells, thereby curtailing antitumor immune responses. ICPM ligands (ICPMLs), however, are often overexpressed on a sizeable fraction of tumor cells of many tumor types and these tumor cells display an aggressive phenotype with changes typical of tumor cells undergoing an epithelial-mesenchymal transition. Moreover, immune cells expressing ICPMLs are often endowed with immunosuppressive or immune-deviated functionalities. Taken together, these observations suggest that compounds with the potential of depleting cells expressing ICPMLs may become useful tools for tumor therapy. In this article, we summarize the current state of the art of these compounds, including avelumab, which is the only ICI targeting an ICPML with preserved effector functions that has gained approval so far. We also discuss approaches allowing to obtain compounds with enhanced tumor cell-depleting potential compared to native antibodies. Eventually, we propose treatment protocols that may be applied in order to optimize the therapeutic efficacy of compounds that deplete cells expressing ICPMLs.


Subject(s)
Immune Checkpoint Proteins/metabolism , Neoplasms/pathology , Antibodies, Neoplasm/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Ligands , Treatment Outcome
17.
Life Sci ; 264: 118618, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33141040

ABSTRACT

AIMS: Obesity represents a global health problem. Excessive caloric intake promotes the release of inflammatory mediators by hypertrophic adipocytes and obesity-induced inflammation is now recognized as a risk factor for the development of several diseases, such as cardiovascular diseases, insulin resistance, type-II diabetes, liver steatosis and cancer. Since obesity causes inflammation, we tested the ability of acetylsalicylic acid (ASA), a potent anti-inflammatory drug, in counteracting this inflammatory process and in mitigating obesity-associated health complications. MAIN METHODS: Mice were fed with standard (SD) or high fat diet (HFD) for 3 months and then treated with acetylsalicylic acid for the subsequent two months. We then analyzed the metabolic and inflammatory status of their adipose and liver tissue by histological, molecular and biochemical analysis. KEY FINDINGS: Although ASA did not exert any effect on body weight, quantification of adipocyte size revealed that the drug slightly reduced adipocyte hypertrophy, however not sufficient so as to induce weight loss. Most importantly, ASA was able to improve insulin resistance. Gene expression profiles of pro- and anti-inflammatory cytokines as well as the expression of macrophage and lymphocyte markers revealed that HFD led to a marked macrophage accumulation in the adipose tissue and an increase of several pro-inflammatory cytokines, a situation almost completely reverted after ASA administration. In addition, liver steatosis caused by HFD was completely abrogated by ASA treatment. SIGNIFICANCE: ASA can efficiently ameliorate pathological conditions usually associated with obesity by inhibiting the inflammatory process occurring in the adipose tissue.


Subject(s)
Adipose Tissue/drug effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspirin/therapeutic use , Diet, High-Fat/adverse effects , Insulin Resistance/physiology , Obesity/drug therapy , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Disease Models, Animal , Female , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/pathology , Treatment Outcome
18.
Am J Pathol ; 175(3): 1066-76, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19700751

ABSTRACT

We have recently observed that oral administration of D-glucose saves animals from lipopolysaccharide (LPS)-induced death. This effect is the likely consequence of glucose-induced activation of the sodium-dependent glucose transporter-1. In this study, we investigated possible hepatoprotective effects of glucose-induced, sodium-dependent, glucose transporter-1 activation. We show that oral administration of D-glucose, but not of either D-fructose or sucrose, prevents LPS-induced liver injury, as well as liver injury and death induced by an overdose of acetaminophen. In both of these models, physiological liver morphology is maintained and organ protection is confirmed by unchanged levels of the circulating markers of hepatotoxicity, such as alanine transaminase or lactate dehydrogenase. In addition, D-glucose was found to protect the liver from alpha-amanitin-induced liver injury. In this case, in contrast to the previously described models, a second signal had to be present in addition to glucose to achieve protective efficacy. Toll-like receptor 4 stimulation that was induced by low doses of LPS was identified as such a second signal. Eventually, the protective effect of orally administered glucose on liver injury induced by LPS, overdose of acetaminophen, or alpha-amanitin was shown to be mediated by the anti-inflammatory cytokine interleukin-10. These findings, showing glucose-induced protective effects in several animal models of liver injury, might be relevant in view of possible therapeutic interventions against different forms of acute hepatic injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Glucose/administration & dosage , Liver Diseases/prevention & control , Acetaminophen/adverse effects , Administration, Oral , Alpha-Amanitin/toxicity , Animals , Fructose/administration & dosage , Galactosamine/adverse effects , Interleukin-10 , Intestinal Mucosa/metabolism , Lipopolysaccharides/toxicity , Liver Diseases/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Sodium-Glucose Transporter 1/metabolism , Sucrose/administration & dosage , Toll-Like Receptor 4/metabolism
19.
Nutr Cancer ; 62(2): 237-42, 2010.
Article in English | MEDLINE | ID: mdl-20099198

ABSTRACT

Different studies have focused on the effects of phytoestrogens-supplemented diets on mammary gland morphogenesis and breast cancer risk; however, particular dieting behaviors and food choices may result in a reduction of the natural source of phytoestrogens. The evaluation of a reduced phytoestrogens intake effect by depletion without modifying other dietary ingredients is hard. Since lignans, the largest contributors to phytoestrogens intake in Western diets, are metabolized into bioactive compounds by gut bacteria, long-term antibiotic treatments, inducing intestinal microflora disruption, may reduce enterolactone availability. To elucidate the effect of phytoestrogens lack on mammary tissue morphogenesis, female FVB mice were treated with gentamicin or metronidazole/ciprofloxacin from the age of 6 to 7 wk. After 21 wk, enterolactone urine levels were 120.07 +/- 20.5 ng/ml in untreated mice, 30.4 +/- 24.46 ng/ml in metronidazole/ciprofloxacin-treated mice, and 3.29 +/- 4.38 ng/ml in gentamicin-treated mice. Histological analysis revealed no significant alterations of mammary morphology in metronidazole/ciprofloxacin-treated mice, whereas gentamicin-treated mice showed increase of ducts number and duct-tree branching vs. controls. These findings indicate that normal mammary tissue size and shape are maintained even in the presence of low levels of lignans and suggest that only a complete depletion of these compounds induced significant alterations of mammary gland structure.


Subject(s)
Lignans/administration & dosage , Lignans/metabolism , Mammary Glands, Animal/growth & development , Morphogenesis/physiology , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/biosynthesis , 4-Butyrolactone/urine , Animals , Anti-Bacterial Agents/pharmacology , Apoptosis , Bacteria/drug effects , Bacteria/metabolism , Cell Division , Ciprofloxacin/pharmacology , Female , Gentamicins/pharmacology , Intestines/microbiology , Lignans/biosynthesis , Lignans/urine , Mammary Glands, Animal/cytology , Metronidazole/pharmacology , Mice , Phytoestrogens/administration & dosage , Phytoestrogens/metabolism
20.
J Immunol ; 181(9): 6132-9, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941203

ABSTRACT

Graft-vs-host disease (GVHD) is a major complication after allogeneic bone marrow transplantation. Different studies have demonstrated that intestinal bacterial breakdown products and loss of gastrointestinal tract integrity, both induced by conditioning regiments, are critical in the pathogenesis of acute GVHD. Using C57BL/6 knockout mice, we evaluated the role of TLR4 and TLR9, which recognize bacterial LPS and DNA, respectively, in the GVHD associated with allogeneic bone marrow transplantation. When myeloablative-irradiated TLR9 knockout (TLR9(-/-)) mice were used as graft recipients, survival and clinical score of acute GVHD were improved as compared with the wild-type recipient mice (18/30 vs 1/31 mice still alive at day 70 in a total of four experiments); while no differences were observed using recipient TLR4 knockout (TLR4(-/-)) mice. The reduced mortality and morbidity in TLR9(-/-) mice related with reduced stimulatory activity of TLR9(-/-) spleen APCs after conditioning and reduced proliferation of allogeneic donor T cells. Experiments using TLR9(+/+) into TLR9(-/-) and TLR9(-/-) into TLR9(+/+) chimeric mice as recipients indicated a critical role for nonhematopoietic TLR9(+/+) cells interacting with bacterial breakdown products released in myeloablated mice. Altogether these data reveal a novel important role of TLR9 in GVHD, a finding that might provide tools to reduce this complication of allogeneic transplantation.


Subject(s)
Bone Marrow Transplantation/immunology , Graft vs Host Disease/immunology , Toll-Like Receptor 9/physiology , Acute Disease , Adoptive Transfer , Animals , Cells, Cultured , Coculture Techniques , Female , Graft vs Host Disease/genetics , Graft vs Host Disease/mortality , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Radiation Chimera/genetics , Radiation Chimera/immunology , Severity of Illness Index , Survival Analysis , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL