Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Bioessays ; 42(4): e1900108, 2020 04.
Article in English | MEDLINE | ID: mdl-32105359

ABSTRACT

Studies performed in absence of gravitational constraint show that a living system is unable to choose between two different phenotypes, thus leading cells to segregate into different, alternative stable states. This finding demonstrates that the genotype does not determine by itself the phenotype but requires additional, physical constraints to finalize cell differentiation. Constraints belong to two classes: holonomic (independent of the system's dynamical states, as being established by the space-time geometry of the field) and non-holonomic (modified during those biological processes to which they contribute in shaping). This latter kind of "constraints", in which dynamics works on the constraint to recreate them, have emerged as critical determinants of self-organizing systems, by manifesting a "closure of constraints." Overall, the constraints act by harnessing the "randomness" represented by the simultaneous presence of equiprobable events restraining the system within one attractor. These results cast doubt on the mainstream scientific concept and call for a better understanding of causation in cell biology.


Subject(s)
Cellular Reprogramming/genetics , Epigenesis, Genetic , Genotype , Phenotype , Cell Engineering , Environment , Gravitation , Humans , Kinetics , Models, Theoretical
2.
Molecules ; 25(19)2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32992691

ABSTRACT

Inositol and its phosphate metabolites play a pivotal role in several biochemical pathways and gene expression regulation: inositol pyrophosphates (PP-IPs) have been increasingly appreciated as key signaling modulators. Fluctuations in their intracellular levels hugely impact the transfer of phosphates and the phosphorylation status of several target proteins. Pharmacological modulation of the proteins associated with PP-IP activities has proved to be beneficial in various pathological settings. IP7 has been extensively studied and found to play a key role in pathways associated with PP-IP activities. Three inositol hexakisphosphate kinase (IP6K) isoforms regulate IP7 synthesis in mammals. Genomic deletion or enzymic inhibition of IP6K1 has been shown to reduce cell invasiveness and migration capacity, protecting against chemical-induced carcinogenesis. IP6K1 could therefore be a useful target in anticancer treatment. Here, we summarize the current understanding that established IP6K1 and the other IP6K isoforms as possible targets for cancer therapy. However, it will be necessary to determine whether pharmacological inhibition of IP6K is safe enough to begin clinical study. The development of safe and selective inhibitors of IP6K isoforms is required to minimize undesirable effects.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Delivery Systems , Enzyme Inhibitors/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Phosphotransferases (Phosphate Group Acceptor)/antagonists & inhibitors , Antineoplastic Agents/chemistry , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Enzyme Inhibitors/chemistry , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Neoplasm Proteins/metabolism , Neoplasms/chemically induced , Neoplasms/enzymology , Neoplasms/pathology , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Signal Transduction/drug effects
3.
Int J Mol Sci ; 20(9)2019 Apr 30.
Article in English | MEDLINE | ID: mdl-31052313

ABSTRACT

Some yet unidentified factors released by both oocyte and embryonic microenvironments demonstrated to be non-permissive for tumor development and display the remarkable ability to foster cell/tissue reprogramming, thus ultimately reversing the malignant phenotype. In the present study we observed how molecular factors extracted from Zebrafish embryos during specific developmental phases (20 somites) significantly antagonize proliferation of breast cancer cells, while reversing a number of prominent aspects of malignancy. Embryo extracts reduce cell proliferation, enhance apoptosis, and dramatically inhibit both invasiveness and migrating capabilities of cancer cells. Counteracting the invasive phenotype is a relevant issue in controlling tumor spreading and metastasis. Moreover, such effect is not limited to cancerous cells as embryo extracts were also effective in inhibiting migration and invasiveness displayed by normal breast cells undergoing epithelial-mesenchymal transition upon TGF-ß1 stimulation. The reversion program involves the modulation of E-cadherin/ß-catenin pathway, cytoskeleton remodeling with dramatic reduction in vinculin, as well as downregulation of TCTP and the concomitant increase in p53 levels. Our findings highlight that-contrary to the prevailing current "dogma", which posits that neoplastic cells are irreversibly "committed"-the malignant phenotype can ultimately be "reversed", at least partially, in response to environmental morphogenetic influences.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Embryo, Nonmammalian/chemistry , Tissue Extracts/pharmacology , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation , Humans , Phenotype , Tumor Protein, Translationally-Controlled 1 , Zebrafish , beta Catenin/metabolism
4.
J Pineal Res ; 64(4): e12467, 2018 May.
Article in English | MEDLINE | ID: mdl-29338098

ABSTRACT

Through activation of the ERK pathway, nicotine, in both normal MCF-10A and low-malignant breast cancer cells (MCF7), promotes increased motility and invasiveness. Melatonin antagonizes both these effects by inhibiting almost completely ERK phosphorylation. As melatonin has no effect on nonstimulated cells, it is likely that melatonin can counteract ERK activation only downstream of nicotine-induced activation. This finding suggests that melatonin hampers ERK phosphorylation presumably by targeting a still unknown intermediate factor that connects nicotine stimulation to ERK phosphorylation. Furthermore, downstream of ERK activation, melatonin significantly reduces fascin and calpain activation while restoring normal vinculin levels. Melatonin also counteracts nicotine effects by reshaping the overall cytoskeleton architecture and abolishing invasive membrane protrusion. In addition, melatonin decreases nicotine-dependent ROCK1/ROCK2 activation, thus further inhibiting cell contractility and motility. Melatonin actions are most likely attributable to ERK inhibition, although melatonin could display other ERK-independent effects, namely through a direct modulation of additional molecular and structural factors, including coronin, cofilin, and cytoskeleton components.


Subject(s)
Adenocarcinoma/pathology , Breast Neoplasms/pathology , Cell Movement/drug effects , MAP Kinase Signaling System/drug effects , Melatonin/pharmacology , Humans , MCF-7 Cells , Neoplasm Invasiveness/pathology , Nicotine/toxicity , Nicotinic Agonists/toxicity , Phosphorylation
5.
Cell Mol Life Sci ; 74(21): 4015-4025, 2017 11.
Article in English | MEDLINE | ID: mdl-28785807

ABSTRACT

The long-recognized fact that oxidative stress within mitochondria is a hallmark of mitochondrial dysfunction has stimulated the development of mitochondria-targeted antioxidant therapies. Melatonin should be included among the pharmacological agents able to modulate mitochondrial functions in cancer, given that a number of relevant melatonin-dependent effects are triggered by targeting mitochondrial functions. Indeed, melatonin may modulate the mitochondrial respiratory chain, thus antagonizing the cancer highly glycolytic bioenergetic pathway of cancer cells. Modulation of the mitochondrial respiratory chain, together with Ca2+ release and mitochondrial apoptotic effectors, may enhance the spontaneous or drug-induced apoptotic processes. Given that melatonin may efficiently counteract the Warburg effect while stimulating mitochondrial differentiation and mitochondrial-based apoptosis, it is argued that the pineal neurohormone could represent a promising new perspective in cancer treatment strategy.


Subject(s)
Antioxidants/pharmacology , Melatonin/pharmacology , Mitochondria/metabolism , Neoplasms/prevention & control , Animals , Humans , Mitochondria/drug effects , Neoplasms/metabolism
6.
Int J Mol Sci ; 19(9)2018 Sep 17.
Article in English | MEDLINE | ID: mdl-30227688

ABSTRACT

Melatonin is a lipophilic hormone synthesized and secreted mainly in the pineal gland, acting as a neuroendocrine transducer of photoperiodic information during the night. In addition to this activity, melatonin has shown an antioxidant function and a key role as regulator of physiological processes related to human reproduction. Melatonin is involved in the normal outcome of pregnancy, beginning with the oocyte quality, continuing with embryo implantation, and finishing with fetal development and parturition. Melatonin has been shown to act directly on several reproductive events, including folliculogenesis, oocyte maturation, and corpus luteum (CL) formation. The molecular mechanism of action has been investigated through several studies which provide solid evidence on the connections between maternal melatonin secretion and embryonic and fetal development. Melatonin administration, reducing oxidative stress and directly acting on its membrane receptors, melatonin thyroid hormone receptors (MT1 and MT2), displays effects on the earliest phases of pregnancy and during the whole gestational period. In addition, considering the reported positive effects on the outcomes of compromised pregnancies, melatonin supplementation should be considered as an important tool for supporting fetal development, opening new opportunities for the management of several reproductive and gestational pathologies.


Subject(s)
Embryo Implantation , Fetal Development , Melatonin/metabolism , Animals , Antioxidants/administration & dosage , Antioxidants/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Embryo Implantation/drug effects , Female , Fetal Development/drug effects , Humans , Melatonin/administration & dosage , Melatonin/pharmacology , Melatonin/therapeutic use , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Pregnancy , Pregnancy Outcome
7.
Exp Cell Res ; 345(1): 37-50, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27237097

ABSTRACT

Inositol displays multi-targeted effects on many biochemical pathways involved in epithelial-mesenchymal transition (EMT). As Akt activation is inhibited by inositol, we investigated if such effect could hamper EMT in MDA-MB-231 breast cancer cells. In cancer cells treated with pharmacological doses of inositol E-cadherin was increased, ß-catenin was redistributed behind cell membrane, and metalloproteinase-9 was significantly reduced, while motility and invading capacity were severely inhibited. Those changes were associated with a significant down-regulation of PI3K/Akt activity, leading to a decrease in downstream signaling effectors: NF-kB, COX-2, and SNAI1. Inositol-mediated inhibition of PS1 leads to lowered Notch 1 release, thus contributing in decreasing SNAI1 levels. Overall, these data indicated that inositol inhibits the principal molecular pathway supporting EMT. Similar results were obtained in ZR-75, a highly metastatic breast cancer line. These findings are coupled with significant changes on cytoskeleton. Inositol slowed-down vimentin expression in cells placed behind the wound-healing edge and stabilized cortical F-actin. Moreover, lamellipodia and filopodia, two specific membrane extensions enabling cell migration and invasiveness, were no longer detectable after inositol addiction. Additionally, fascin and cofilin, two mandatory required components for F-actin assembling within cell protrusions, were highly reduced. These data suggest that inositol may induce an EMT reversion in breast cancer cells, suppressing motility and invasiveness through cytoskeleton modifications.


Subject(s)
Breast Neoplasms/pathology , Cytoskeleton/pathology , Epithelial Cells/pathology , Inositol/pharmacology , Mesoderm/pathology , Amyloid Precursor Protein Secretases/metabolism , Cadherins/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cytoskeleton/drug effects , Down-Regulation/drug effects , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Female , Humans , Immunoblotting , Mesoderm/drug effects , Microfilament Proteins/metabolism , NF-kappa B/metabolism , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Presenilin-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vimentin/metabolism , Wound Healing/drug effects , beta Catenin/metabolism , rho-Associated Kinases/metabolism
8.
Int J Mol Sci ; 18(10)2017 Oct 20.
Article in English | MEDLINE | ID: mdl-29053604

ABSTRACT

Communities eating a western-like diet, rich in fat, sugar and significantly deprived of fibers, share a relevant increased risk of both metabolic and cancerous diseases. Even more remarkable is that a low-fiber diet lacks some key components-as phytates and inositols-for which a mechanistic link has been clearly established in the pathogenesis of both cancer and metabolic illness. Reduced bioavailability of inositol in living organisms could arise from reduced food supply or from metabolism deregulation. Inositol deregulation has been found in a number of conditions mechanistically and epidemiologically associated to high-glucose diets or altered glucose metabolism. Indeed, high glucose levels hinder inositol availability by increasing its degradation and by inhibiting both myo-Ins biosynthesis and absorption. These underappreciated mechanisms may likely account for acquired, metabolic deficiency in inositol bioavailability.


Subject(s)
Inositol/deficiency , Metabolic Diseases/chemically induced , Biological Availability , Humans , Inositol/pharmacokinetics , Nutritional Status
9.
Methods Mol Biol ; 2769: 87-98, 2024.
Article in English | MEDLINE | ID: mdl-38315391

ABSTRACT

The ectopic xenograft mouse model of cancer is a commonly employed tool for in vivo investigations, particularly for studying cell tumorigenicity and testing the efficacy and tolerability of systemic or local anti-cancer therapies. The model displays advantageous features with an easy-access to visualize and monitor tumor growth in real-time with a caliper. Although the tumor development occurs in an ectopic location, the histology of the tumor resembles that of human cancer upon pathological examination. This suggests that when human malignant cells are transplanted into immunocompromised mice, they can educate and attract murine cells from the surrounding environment to recapitulate a tumor structure. The experimental protocol for ectopic xenograft models is straightforward, making them reproducible, cost-effective, and conductive to shorter experimental durations. Here, we detail the utilization of ectopic xenograft models in studying biliary tract cancers (BTC), which involves subcutaneously grafting human BTC cell lines originating from different biliary tree locations onto immunocompromised nude mice.


Subject(s)
Biliary Tract Neoplasms , Humans , Animals , Mice , Mice, Nude , Heterografts , Xenograft Model Antitumor Assays , Cell Line, Tumor , Biliary Tract Neoplasms/metabolism , Biliary Tract Neoplasms/pathology , Models, Theoretical
10.
Curr Oncol ; 30(4): 4185-4196, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37185432

ABSTRACT

During the last decade, immunotherapy has radically changed perspectives on anti-tumor treatments. However, solid tumor treatment by immunotherapy has not met expectations. Indeed, poor clinical response to treatment has highlighted the need to understand and avoid immunotherapy resistance. Cholangiocarcinoma (CCA) is the second cause of hepatic cancer-related deaths because of drug inefficacy and chemo-resistance in a majority of patients. Thus, intense research is ongoing to better understand the mechanisms involved in the chemo-resistance processes. The tumor microenvironment (TME) may be involved in tumor therapy resistance by limiting drug access. Indeed, cells such as cancer-associated fibroblasts (CAFs) alter TME by producing in excess an aberrant extracellular matrix (ECM). Interestingly, CAFs are the dominant stromal component in CCA that secrete large amounts of stiff ECM. Stiff ECM could contribute to immune exclusion by limiting anti-tumor T-cells drop-in. Herein, we summarize features, functions, and interactions among CAFs, tumor-associated ECM, and immune cells in TME. Moreover, we discuss the strategies targeting CAFs and the remodeling of the ECM to improve immunotherapy and drug therapies.


Subject(s)
Bile Duct Neoplasms , Cancer-Associated Fibroblasts , Cholangiocarcinoma , Humans , Cholangiocarcinoma/therapy , Extracellular Matrix , Bile Duct Neoplasms/therapy , Bile Ducts, Intrahepatic , Tumor Microenvironment
11.
Cancer Lett ; 520: 48-56, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34229060

ABSTRACT

The deregulation of PI3K/Akt signaling is among the most causes in inducing the acquisition of a metastatic phenotype in breast cancer cells, leading to Epithelial-Mesenchymal Transition (EMT). Inhibition of the PI3K/Akt pathway is known to be beneficial in the clinical setting. However, the activation of secondary pathways and toxicity profiles of available inhibitors, hindering optimal therapeutic results. Preliminary studies showed that myo-Inositol inhibits the PI3K/Akt pathway by exerting a pleiotropic anti-tumor action. Herein, we demonstrate that myo-Inositol triggers a prompt and profound remodeling of delineated expression pattern in triple-negative breast cancer cells (MDA-MB-231). Consequently, it inhibits metastasis and tumor progression through miR-125a-5p transcription and the subsequent inhibition of IP6K1. In contrast, hormone-responsive breast cancer cells (MCF-7) are insensitive to myo-Inositol. This is due to the persistence of MDM2 synthesis promoted by estrogen-dependent pathways. Conversely, the counteraction of estrogen effects recovered the sensitivity to myo-Inositol in the hormone-responsive model. Overall, these results identify a novel axis primed by miR-125a-5p to downregulate IP6K1 gene that inhibits metastasis. Thus, administration of myo-Inositol can activate this axis as a molecular target therapy in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , MicroRNAs/genetics , Neoplasms, Hormone-Dependent/drug therapy , Phosphotransferases (Phosphate Group Acceptor)/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Inositol/pharmacology , MCF-7 Cells , Neoplasm Metastasis , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Signal Transduction/drug effects
12.
Science ; 374(6564): 216-224, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34618582

ABSTRACT

The microbiota comprises the microorganisms that live in close contact with the host, with mutual benefit for both counterparts. The contribution of the gut microbiota to the emergence of castration-resistant prostate cancer (CRPC) has not yet been addressed. We found that androgen deprivation in mice and humans promotes the expansion of defined commensal microbiota that contributes to the onset of castration resistance in mice. Specifically, the intestinal microbial community in mice and patients with CRPC was enriched for species capable of converting androgen precursors into active androgens. Ablation of the gut microbiota by antibiotic therapy delayed the emergence of castration resistance even in immunodeficient mice. Fecal microbiota transplantation (FMT) from CRPC mice and patients rendered mice harboring prostate cancer resistant to castration. In contrast, tumor growth was controlled by FMT from hormone-sensitive prostate cancer patients and Prevotella stercorea administration. These results reveal that the commensal gut microbiota contributes to endocrine resistance in CRPC by providing an alternative source of androgens.


Subject(s)
Androgens/biosynthesis , Bacteria/metabolism , Gastrointestinal Microbiome/physiology , Host Microbial Interactions , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/microbiology , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/genetics , Cell Line, Tumor , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasms, Experimental , Prevotella/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Symbiosis , Xenograft Model Antitumor Assays
13.
Drug Discov Today ; 25(4): 731-738, 2020 04.
Article in English | MEDLINE | ID: mdl-32027971

ABSTRACT

The widely recognized problems of pharmacological strategies based on killing cancer cells demand a rethink of therapeutic approaches. Tumor reversion strategies that aim to shift cancer cells to a healthy differentiated state are a promising alternative. Although many studies have firmly demonstrated the possibility of reverting cancer to a normal differentiated state, we are still unable (with the exception of retinoic acid in a form of leukemia) to revert cancer cells to a stable differentiated healthy state. Here, we review the main biological bases of redifferentiation strategies and provide a description of the most promising research avenues.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Neoplasms/drug therapy , Animals , Humans , Neoplasms/pathology , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL