Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Theor Biol ; 575: 111645, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37863423

ABSTRACT

Recent studies at individual cell resolution have revealed phenotypic heterogeneity in nominally clonal tumor cell populations. The heterogeneity affects cell growth behaviors, which can result in departure from the idealized uniform exponential growth of the cell population. Here we measured the stochastic time courses of growth of an ensemble of populations of HL60 leukemia cells in cultures, starting with distinct initial cell numbers to capture a departure from the uniform exponential growth model for the initial growth ("take-off"). Despite being derived from the same cell clone, we observed significant variations in the early growth patterns of individual cultures with statistically significant differences in growth dynamics, which could be explained by the presence of inter-converting subpopulations with different growth rates, and which could last for many generations. Based on the hypothesis of existence of multiple subpopulations, we developed a branching process model that was consistent with the experimental observations.


Subject(s)
Population Growth , Cell Cycle , Cell Proliferation , Clone Cells , Phenotype , Stochastic Processes
2.
Int J Mol Sci ; 23(16)2022 Aug 13.
Article in English | MEDLINE | ID: mdl-36012339

ABSTRACT

Ovarian cancer represents one of the most malignant gynecological cancers worldwide, with an overall 5-year survival rate, being locked in the 25-30% range in the last decade. Cancer immunotherapy is currently one of the most intensively investigated and promising therapeutic strategy and as such, is expected to provide in the incoming years significant benefits for ovarian cancer treatment as well. Here, we provide a detailed survey on the highly pleiotropic oncosuppressive roles played by the human RNASET2 gene, whose protein product has been consistently reported to establish a functional crosstalk between ovarian cancer cells and key cellular effectors of the innate immune system (the monocyte/macrophages lineage), which is in turn able to promote the recruitment to the cancer tissue of M1-polarized, antitumoral macrophages. This feature, coupled with the ability of T2 ribonucleases to negatively affect several cancer-related parameters in a cell-autonomous manner on a wide range of ovarian cancer experimental models, makes human RNASET2 a very promising candidate to develop a "multitasking" therapeutic approach for innovative future applications for ovarian cancer treatment.


Subject(s)
Ovarian Neoplasms , Ribonucleases , Tumor Suppressor Proteins , Female , Genes, Tumor Suppressor , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ribonucleases/genetics , Ribonucleases/metabolism , Tumor Suppressor Proteins/genetics
3.
Cell Tissue Res ; 368(2): 337-351, 2017 05.
Article in English | MEDLINE | ID: mdl-28070637

ABSTRACT

In recent years, several studies have demonstrated that the RNASET2 gene is involved in the control of tumorigenicity in ovarian cancer cells. Furthermore, a role in establishing a functional cross-talk between cancer cells and the surrounding tumor microenvironment has been unveiled for this gene, based on its ability to act as an inducer of the innate immune response. Although several studies have reported on the molecular features of RNASET2, the details on the mechanisms by which this evolutionarily conserved ribonuclease regulates the immune system are still poorly defined. In the effort to clarify this aspect, we report here the effect of recombinant human RNASET2 injection and its role in regulating the innate immune response after bacterial challenge in an invertebrate model, the medicinal leech. We found that recombinant RNASET2 injection induces fibroplasias, connective tissue remodeling and the recruitment of numerous infiltrating cells expressing the specific macrophage markers CD68 and HmAIF1. The RNASET2-mediated chemotactic activity for macrophages has been further confirmed by using a consolidated experimental approach based on injection of the Matrigel biomatrice (MG) supplemented with recombinant RNASET2 in the leech body wall. One week after injection, a large number of CD68+ and HmAIF-1+ macrophages massively infiltrated MG sponges. Finally, in leeches challenged with lipopolysaccharides (LPS) or with the environmental bacteria pathogen Micrococcus nishinomiyaensis, numerous macrophages migrating to the site of inoculation expressed high levels of endogenous RNASET2. Taken together, these results suggest that RNASET2 is likely involved in the initial phase of the inflammatory response in leeches.


Subject(s)
Connective Tissue/pathology , Hirudo medicinalis/physiology , Inflammation/pathology , Recombinant Proteins/pharmacology , Ribonucleases/pharmacology , Tumor Suppressor Proteins/pharmacology , Acid Phosphatase/metabolism , Animals , Cell Proliferation/drug effects , Collagen/metabolism , Connective Tissue/drug effects , Cryoultramicrotomy , Drug Combinations , Enzyme Assays , Fluorescent Antibody Technique , Hirudo medicinalis/anatomy & histology , Hirudo medicinalis/drug effects , Hirudo medicinalis/ultrastructure , Humans , Laminin/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Proteoglycans/metabolism
4.
Proc Natl Acad Sci U S A ; 110(20): 8140-5, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23630276

ABSTRACT

In recent years, the role played by the stromal microenvironment has been given growing attention in order to achieve a full understanding of cancer initiation and progression. Because cancer is a tissue-based disease, the integrity of tissue architecture is a major constraint toward cancer growth. Indeed, a large contribution of the natural resistance to cancer stems from stromal microenvironment components, the dysregulation of which can facilitate cancer occurrence. For instance, recent experimental evidence has highlighted the involvement of stromal cells in ovarian carcinogenesis, as epitomized by ovarian xenografts obtained by a double KO of the murine Dicer and Pten genes. Likewise, we reported the role of an ancient extracellular RNase, called Ribonuclease T2 (RNASET2), within the ovarian stromal microenvironment. Indeed, hyperexpression of RNASET2 is able to control tumorigenesis by recruiting macrophages (mostly of the anticancer M1 subtype) at the tumor sites. We present biological data obtained by RNASET2 silencing in the poorly tumorigenetic and highly RNASET2-expressing human OVCAR3 cell line. RNASET2 knockdown was shown to stimulate in vivo tumor growth early after microinjection of OVCAR3 cells in nude mice. Moreover, we have investigated by molecular profiling the in vivo expression signature of human and mouse cell xenografts and disclosed the activation of pathways related to activation of the innate immune response and modulation of ECM components. Finally, we provide evidence for a role of RNASET2 in triggering an in vitro chemotactic response in macrophages. These results further highlight the critical role played by the microenvironment in RNASET2-mediated ovarian tumor suppression, which could eventually contribute to better clarify the pathogenesis of this disease.


Subject(s)
Endoribonucleases/physiology , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Animals , Cell Line, Tumor , Chemotaxis , Endoribonucleases/genetics , Female , Gene Expression Profiling , Gene Knockdown Techniques , Gene Silencing , Humans , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Nude , Neoplasm Transplantation , Phylogeny , Polymerase Chain Reaction , U937 Cells
5.
Proc Natl Acad Sci U S A ; 108(3): 1104-9, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21189302

ABSTRACT

A recent body of evidence indicates an active role for stromal (mis)-regulation in the progression of neoplasias. Within this conceptual framework, genes belonging to the growing but still poorly characterized class of tumor antagonizing/malignancy suppressor genes (TAG/MSG) seem to play a crucial role in the regulation of the cross-talk between stromal and epithelial cells by controlling malignant growth in vivo without affecting any cancer-related phenotype in vitro. Here, we have functionally characterized the human RNASET2 gene, which encodes the first human member of the widespread Rh/T2/S family of extracellular RNases and was recently found to be down-regulated at the transcript level in several primary ovarian tumors or cell lines and in melanoma cell lines. Although we could not detect any activity for RNASET2 in several functional in vitro assays, a remarkable control of ovarian tumorigenesis could be detected in vivo. Moreover, the control of ovarian tumorigenesis mediated by this unique tumor suppressor gene occurs through modification of the cellular microenvironment and the induction of immunocompetent cells of the monocyte/macrophage lineage. Taken together, the data presented in this work strongly indicate RNASET2 as a previously unexplored member of the growing family of tumor-antagonizing genes.


Subject(s)
Macrophages/immunology , Ovarian Neoplasms/genetics , Ribonucleases/immunology , Tumor Suppressor Proteins/immunology , Analysis of Variance , Animals , Cell Line, Tumor , Female , Humans , Immunohistochemistry , In Situ Hybridization , In Vitro Techniques , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Ribonucleases/genetics , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays
6.
Biol Cell ; 104(1): 13-21, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22188480

ABSTRACT

BACKGROUND INFORMATION: The ribonucleases (RNases) constitute a heterogeneous group of enzymes, which exert diverse and specific biological functions. Several RNases have been shown to control gene expression and cell differentiation. RNASET2, a novel member of the Rh/T2/S family of RNases, exerts micro-environmental control of malignancy in different experimental models with a general onco-suppressor activity, since it prevents cancer proliferation. Indeed, RNASET2 was found to be downregulated at the transcript level in several primary ovarian tumours or cell lines and in melanoma cell lines. Although recent works shed light on the biological role of RNASET2 in delaying tumour growth, its trafficking within the cell is still poorly understood. RNASET2 seems to play diverse biological roles including turnover of tRNA in yeast as well as rRNA degradation in zebrafish. RESULTS: Here, we have studied the intracellular trafficking of RNASET2 in mammalian cells. RNASET2 co-localizes with markers for the trans-Golgi network (TGN), which is the central sorting and processing station of the secretory pathway. Moreover, using the temperature-sensitive vesicular stomatitis glycoprotein, we demonstrate that RNASET2 undergoes delivery to the plasma membrane. In contrast to other RNA-interacting proteins, RNASET2 does not accumulate in stress granules upon metabolic stress in mammalian cells. Surprisingly, RNASET2 shows co-localization with processing bodies (P-bodies), which increases upon metabolic stress. Finally, cells lacking RNASET2 show a reduced numbers of P-bodies. CONCLUSIONS: In this study, we have identified two distinct cellular pools of RNASET2-containing granules. One pool undergoes membrane delivery using the TGN, and it is released to the extracellular environment. The second pool is recruited into P-bodies, suggesting a possible involvement of RNASET2 in P-body formation in mammalian cells.


Subject(s)
Ribonucleases/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , HeLa Cells , Humans , Protein Transport/physiology , Ribonucleases/genetics , Tumor Suppressor Proteins/genetics , trans-Golgi Network/metabolism
7.
ArXiv ; 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37904742

ABSTRACT

Recent studies at individual cell resolution have revealed phenotypic heterogeneity in nominally clonal tumor cell populations. The heterogeneity affects cell growth behaviors, which can result in departure from the idealized uniform exponential growth of the cell population. Here we measured the stochastic time courses of growth of an ensemble of populations of HL60 leukemia cells in cultures, starting with distinct initial cell numbers to capture a departure from the uniform exponential growth model for the initial growth ("take-off"). Despite being derived from the same cell clone, we observed significant variations in the early growth patterns of individual cultures with statistically significant differences in growth dynamics, which could be explained by the presence of inter-converting subpopulations with different growth rates, and which could last for many generations. Based on the hypothesis of existence of multiple subpopulations, we developed a branching process model that was consistent with the experimental observations.

8.
Biotechnol Appl Biochem ; 58(1): 39-49, 2011.
Article in English | MEDLINE | ID: mdl-21446958

ABSTRACT

We report the expression of recombinant RNASET2, the only human member of the Rh/T2/S family of acid ribonucleases, in the yeast Pichia pastoris and the baculovirus-insect cell heterologous systems. In both models, the yield of recombinant protein was comparable and ranged between 5 mg/L (for a catalytically impaired mutant version of RNASET2) and 30 mg/L for the wild-type protein. Thus, the produced protein version rather than the expression system used appears to influence protein yield after optimization of culture conditions. The recombinant protein was found to undergo heterogeneous glycosylation in both systems, particularly in P. pastoris. Most importantly, the wild-type protein purified from both systems was found to be catalytically competent. The expression of recombinant RNASET2 in both systems will allow the implementation of functional assays in vivo and in vitro to better define the antioncogenic properties of this member of the Rh/T2/S ribonuclease family.


Subject(s)
Baculoviridae/metabolism , Gene Expression Regulation, Neoplastic , Pichia/metabolism , Ribonucleases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Baculoviridae/genetics , Base Sequence , Biocatalysis , Cells, Cultured , Cloning, Molecular , Glycosylation , Humans , Molecular Sequence Data , Mutation , Pichia/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribonucleases/genetics , Tumor Suppressor Proteins/genetics
9.
Cancers (Basel) ; 12(3)2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32197460

ABSTRACT

Human RNASET2 acts as a powerful oncosuppressor protein in in vivo xenograft-based murine models of human cancer. Secretion of RNASET2 in the tumor microenvironment seems involved in tumor suppression, following recruitment of M1-polarized macrophages. Here, we report a murine Rnaset2-based syngeneic in vivo assay. BALB/c mice were injected with parental, empty vector-transfected or murine Rnaset2-overexpressing mouse C51 or TS/A syngeneic cells and tumor growth pattern and immune cells distribution in tumor mass were investigated. Compared to control cells, mouse Rnaset2-expressing C51 cells showed strong delayed tumor growth. CD86+ M1 macrophages were massively recruited in Rnaset2-expressing C51-derived tumors, with concomitant inhibition of MDSCs and CD206+ M2 macrophages recruitment. At later times, a relevant expansion of intra-tumor CD8+ T cells was also observed. After re-challenge with C51 parental cells, most mice previously injected with Rnaset2-expressing C51 cells still rejected C51 tumor cells, suggesting a Rnaset2-mediated T cell adaptive immune memory response. These results point at T2 RNases as evolutionary conserved oncosuppressors endowed with the ability to inhibit cancer growth in vivo through rebalance of intra-tumor M1/M2 macrophage ratio and concomitant recruitment of adaptive anti-tumor CD8+ T cells.

10.
Cardiovasc Res ; 116(8): 1458-1472, 2020 07 01.
Article in English | MEDLINE | ID: mdl-31688894

ABSTRACT

AIMS: Increased Ankyrin Repeat Domain 1 (ANKRD1) levels linked to gain of function mutations have been associated to total anomalous pulmonary venous return and adult cardiomyopathy occurrence in humans. The link between increased ANKRD1 level and cardiac structural and functional disease is not understood. To get insight into this problem, we have generated a gain of function ANKRD1 mouse model by overexpressing ANKRD1 in the myocardium. METHODS AND RESULTS: Ankrd1 is expressed non-homogeneously in the embryonic myocardium, with a dynamic nucleo-sarcomeric localization in developing cardiomyocytes. ANKRD1 transgenic mice present sinus venosus defect, which originates during development by impaired remodelling of early embryonic heart. Adult transgenic hearts develop diastolic dysfunction with preserved ejection fraction, which progressively evolves into heart failure, as shown histologically and haemodynamically. Transgenic cardiomyocyte structure, sarcomeric assembly, and stability are progressively impaired from embryonic to adult life. Postnatal transgenic myofibrils also present characteristic functional alterations: impaired compliance at neonatal stage and impaired lusitropism in adult hearts. Altogether, our combined analyses suggest that impaired embryonic remodelling and adult heart dysfunction in ANKRD1 transgenic mice present a common ground of initial cardiomyocyte defects, which are exacerbated postnatally. Molecular analysis showed transient activation of GATA4-Nkx2.5 transcription in early transgenic embryos and subsequent dynamic transcriptional modulation within titin gene. CONCLUSIONS: ANKRD1 is a fine mediator of cardiomyocyte response to haemodynamic load in the developing and adult heart. Increased ANKRD1 levels are sufficient to initiate an altered cellular phenotype, which is progressively exacerbated into a pathological organ response by the high ventricular workload during postnatal life. Our study defines for the first time a unifying picture for ANKRD1 role in heart development and disease and provides the first mechanistic link between ANKRD1 overexpression and cardiac disease onset.


Subject(s)
Heart Septal Defects, Atrial/metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Function, Left , Animals , Diastole , Female , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Heart Septal Defects, Atrial/genetics , Heart Septal Defects, Atrial/pathology , Heart Septal Defects, Atrial/physiopathology , Homeobox Protein Nkx-2.5/genetics , Homeobox Protein Nkx-2.5/metabolism , Male , Mice, Transgenic , Muscle Proteins/genetics , Myocardium/pathology , Nuclear Proteins/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Repressor Proteins/genetics , Up-Regulation , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
11.
Int J Oncol ; 34(4): 1099-108, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287968

ABSTRACT

Antisense transcription has long been recognized as a mechanism involved in the regulation of gene expression. Therefore, several human diseases associated with abnormal patterns of gene expression might display antisense RNA-mediated pathogenetic mechanisms. Such issue could be particularly relevant for cancer pathogenesis, since deregulated gene expression has long been established as a hallmark of cancer cells. Herein, we report on a bioinformatic search for antisense transcription in two cancer-associated regions of human chromosome 6 (6q21 and 6q27). Natural antisense transcripts (NATs) for several genes in both genomic regions were predicted in silico and subsequently validated by strand-specific RT-PCR. Detailed experimental validation by quantitative real-time RT-PCR of five putative cancer related sense-antisense transcript pairs revealed a single candidate tumor suppressor gene (RPS6KA2) whose expression levels display marked cancer-related changes that are likely mediated by its antisense RNA in a breast cancer cell line model.


Subject(s)
Chromosomes, Human, Pair 6 , Neoplasms/genetics , RNA, Antisense/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Computational Biology/methods , Genes, Tumor Suppressor , Genome, Human , Humans , Introns , Models, Biological , Models, Genetic , Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Oligonucleotides, Antisense , RNA, Antisense/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Protein S6 Kinases, 90-kDa/biosynthesis
12.
Front Immunol ; 10: 2587, 2019.
Article in English | MEDLINE | ID: mdl-31749812

ABSTRACT

The link between cancer development or progression and immune system dysregulation has long been established. Virtually every cell type belonging to both the innate and adaptive immune system has been reported to be involved in a complex interplay that might culminate into either a pro- or anti-tumorigenic response. Among the cellular components of the innate immune system, cells belonging to the monocyte/macrophage lineage have been consistently shown to play a key role in the tumorigenic process. The most advanced human tumors are reported to be strongly infiltrated with Tumor-Associated Macrophages (TAMs) endowed with the ability to contribute to tumor growth and dissemination. However, given their widely acknowledged functional plasticity, macrophages can display anti-tumor properties as well. Based on these premises, experimental approaches to promote the in vivo macrophage shift from pro-tumor to anti-tumor phenotype represent one of the most promising research field aimed at developing immune system-mediated tumor suppressive therapies. In this context, the human RNASET2 oncosuppressor gene has emerged as a potential tool for macrophage-mediated tumor suppression. A growing body of experimental evidence has been reported to suggest a role for this gene in the regulation of macrophage activity in both in vitro and in vivo experimental models. Moreover, several recent reports suggest a role for this gene in a broad range of cell types involved in immune response, pointing at RNASET2 as a putative regulator of several functional features within the immune system.


Subject(s)
Ribonucleases/immunology , Tumor Suppressor Proteins/immunology , Animals , Humans , Immunity, Innate , Macrophages/immunology , Monocytes/immunology , Ribonucleases/genetics , Tumor Suppressor Proteins/genetics
13.
Cancers (Basel) ; 11(2)2019 Feb 22.
Article in English | MEDLINE | ID: mdl-30813308

ABSTRACT

RNASET2 is an extracellular ribonuclease endowed with a marked antitumorigenic role in several carcinomas, independent from its catalytic activity. Besides its antitumorigenic role by the recruitment to the tumor mass of immune cells from the monocyte/macrophage lineage, RNASET2 is induced by cellular stress and involved in actin cytoskeleton remodeling affecting cell interactions with the extracellular matrix (ECM). Here, we aimed to investigate the effects of RNASET2 expression modulation on cell phenotype and behavior in epithelial ovarian cancer (EOC) cellular models. In silico analysis on two publicly available datasets of gene expression from EOC patients (n = 392) indicated that increased RNASET2 transcript levels are associated with longer overall survival. In EOC biopsies (n = 101), analyzed by immunohistochemistry, RNASET2 was found heterogeneously expressed among tumors with different clinical⁻pathological characteristics and, in some cases, its expression localized to tumor-associated ECM. By characterizing in vitro two models of EOC cells in which RNASET2 was silenced or overexpressed, we report that RNASET2 expression negatively affects growth capability by conferring a peculiar cell phenotype upon the interaction of EOC cells with the ECM, resulting in decreased src activation. Altogether, these data suggest that drugs targeting activated src might represent a therapeutic approach for RNASET2-expressing EOCs.

14.
J Innate Immun ; 11(2): 150-167, 2019.
Article in English | MEDLINE | ID: mdl-30368505

ABSTRACT

Recent studies demonstrated that allograft inflammatory factor-1 (AIF-1) and RNASET2 act as chemoattractants for macrophages and modulate the inflammatory processes in both vertebrates and invertebrates. The expression of these proteins significantly increases after bacterial infection; however, the mechanisms by which they regulate the innate immune response are still poorly defined. Here, we evaluate the effect of bacterial lipopolysaccharide injection on the expression pattern of these genes and the interrelation between them during innate immune response in the medicinal leech, an invertebrate model with a simple anatomy and a marked similarity with vertebrates in inflammatory processes. Collectively, prokaryotic-eukaryotic co-cultures and in vivo infection assays suggest that RNASET2 and AIF-1 play a crucial role in orchestrating a functional cross-talk between granulocytes and macrophages in leeches, resulting in the activation of an effective response against pathogen infection. RNASET2, firstly released by granulocytes, likely plays an early antibacterial role. Subsequently, AIF-1+ RNASET2-recruited macrophages further recruit other macrophages to potentiate the antibacterial inflammatory response. These experimental data are in keeping with the notion of RNA-SET2 acting as an alarmin-like molecule whose role is to locally transmit a "danger" signal (such as a bacterial infection) to the innate immune system in order to trigger an appropriate host response.


Subject(s)
Calcium-Binding Proteins/metabolism , Endoribonucleases/metabolism , Escherichia coli Infections/immunology , Escherichia coli/immunology , Hirudo medicinalis/immunology , Macrophages/immunology , Neutrophils/immunology , Alarmins/metabolism , Animals , Cells, Cultured , Immunity, Innate , Lipopolysaccharides/immunology
15.
Hum Mutat ; 29(4): 468-74, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18273862

ABSTRACT

Total anomalous pulmonary venous return (TAPVR) is a congenital heart defect in which the pulmonary veins fail to enter the left atrium and drain instead into the right atrium or one of its venous tributaries. Although a genetic basis for TAPVR has long been recognized, no single gene involved in the pathogenesis of this disease has been identified to date. We previously reported a TAPVR patient bearing a de novo 10;21 balanced translocation. In this work, we cloned both translocation breakpoints from this patient and mapped the ANKRD1 gene, encoding a cardiac transcriptional regulator, 130 kb proximally to the breakpoint on chromosome 10. In situ hybridization analysis performed on murine embryos showed ANKRD1 expression in the developing pulmonary veins, suggesting a possible role for this gene in TAPVR pathogenesis. Moreover, ANKRD1 expression levels were found to be highly increased in lymphoblastoid cell lines derived from both the translocation-bearing proband and a second independent sporadic TAPVR patient, suggesting that disruption of the normal ANKRD1 expression pattern is associated with TAPVR. Finally, a nonconservative missense mutation in the ANKRD1 gene was found in a third sporadic TAPVR patient. In vitro calpain-mediated degradation assays, coupled to reporter gene analysis in transfected HeLa cells, strongly suggested that this mutation enhances both the stability of the ANKRD1/CARP protein and its transcriptional repression activity upon the cardiac-specific atrial natriuretic factor (ANF) promoter. Taken together, these results define ANKRD1 as a possible candidate gene for TAPVR pathogenesis.


Subject(s)
Heart Defects, Congenital/genetics , Muscle Proteins/genetics , Mutation, Missense , Nuclear Proteins/genetics , Pulmonary Veins/abnormalities , Repressor Proteins/genetics , Animals , Base Sequence , Cell Line , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 21/genetics , DNA/genetics , Female , Gene Expression , HeLa Cells , Heart Defects, Congenital/metabolism , Humans , Male , Mice , Muscle Proteins/metabolism , Nuclear Proteins/metabolism , Pedigree , Pregnancy , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Transfection , Translocation, Genetic
16.
Oncol Res ; 17(2): 69-74, 2008.
Article in English | MEDLINE | ID: mdl-18543608

ABSTRACT

The RNASET2 gene, mapped in 6q27, was previously found to exert control of tumorigenesis in an ovarian cancer system. We present here results indicating a similar control in a melanoma cancer model. Thus, this gene is most likely involved in a common general pathway of tumorigenesis. Moreover, its antitumorigenic activity is manifested in vivo but not in vitro, suggesting that this gene belongs to the growing category of tumor antagonizing/malignancy suppressor genes. A possible role of RNASET2 in the activation of a senescence program, whose responsible locus was mapped in the same chromosomal 6q27 region, seems to be inconsistent with our data.


Subject(s)
Disease Models, Animal , Genes, Tumor Suppressor/physiology , Genetic Therapy , Melanoma, Experimental/therapy , Ribonucleases/genetics , Tumor Suppressor Proteins/genetics , Animals , Blotting, Northern , Cell Proliferation , Chromosomes, Human, Pair 6/genetics , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Ultrastruct Pathol ; 32(6): 246-51, 2008.
Article in English | MEDLINE | ID: mdl-19117266

ABSTRACT

BACE2 is a protease homologous to BACE1 protein, an enzyme involved in the amyloid formation of Alzheimer disease (AD). However, despite the high homology between these two proteins, the biological role of BACE2 is still controversial, even though a few studies have suggested a pathogenetic role in sporadic inclusion-body myositis and hereditary inclusion-body myopathy, which are characterized by vacuolization of muscular fibers with intracellular deposits of proteins similar to those found in the brain of AD patients. Although BACE2 has also been identified in the pancreas, its function remains unknown and its specific localization in different pancreatic cell types has not been definitively ascertained. For these reasons, the authors have investigated the cellular and subcellular localization of BACE2 in normal rodent pancreases. BACE2 immunoreactivity was found in secretory granules of beta cells, co-stored with insulin and IAPP, while it was lacking in the other endocrine and exocrine cell types. The presence of BACE2 in secretory granules of beta cells suggests that it may play a role in diabetes-associated amyloidogenesis.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Insulin-Secreting Cells/enzymology , Secretory Vesicles/enzymology , Amyloid Precursor Protein Secretases/ultrastructure , Animals , Aspartic Acid Endopeptidases/ultrastructure , Immunohistochemistry , Insulin-Secreting Cells/ultrastructure , Mice , Microscopy, Electron, Transmission , Rats , Secretory Vesicles/ultrastructure
18.
Sci Rep ; 8(1): 17903, 2018 Dec 12.
Article in English | MEDLINE | ID: mdl-30538266

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

19.
Immunol Lett ; 203: 102-111, 2018 11.
Article in English | MEDLINE | ID: mdl-30218741

ABSTRACT

Macrophages represent key inflammatory cellular effectors of the innate immune response. Despite being widely acknowledged as professional phagocytes, the functional roles played by these cells have been progressively widened over the years to encompass regulation of the adaptive immune system, stimulation or suppression of cancer cell growth and tissue remodeling. These diverse functional features have led to the concept of "macrophage plasticity", i.e. the ability of these cells to express a wide range of phenotypes endowed with different functional roles. Several activation programs have been described for mammalian macrophages, based mainly on their differential transcriptional profiles. Based on established in vitro experimental conditions, many researchers currently refer to the M1 (or M1-like) and M2 (or M2-like) terms to describe the two extremes of a rather broad spectrum of polarization states that macrophages can experience in vivo. In light of the widely recognized opposite roles of M1-like and M2-like macrophages on cancer growth, and our largely incomplete knowledge of the cellular and molecular mechanisms underlying the establishment of the M1-like versus M2-like balance within a tumor mass, we report here results from in vitro assays pointing at the human RNASET2 gene as a potential regulator of the balance between M1-like/M2-like macrophage polarization. Not only do our results confirm previous in vivo data, thus further supporting a role for this pleiotropic protein in the innate immune system, but they also define RNASET2 as a new molecular target with potential applications for in vivo reprogramming of macrophage polarization, an increasingly appraised anticancer strategy.


Subject(s)
Macrophage Activation , Macrophages/immunology , Ribonucleases/immunology , Tumor Suppressor Proteins/immunology , Cells, Cultured , Female , Humans , Male
20.
Hum Pathol ; 79: 66-76, 2018 09.
Article in English | MEDLINE | ID: mdl-29763721

ABSTRACT

Ribonuclease T2 (RNASET2) is a pleiotropic and polyfunctional protein, which exerts several different activities in neoplastic cells since the early steps of tumor development. Besides having an antitumorigenic activity, RNASET2 inhibits both bFGF-induced and VEGF-induced angiogenesis and has a role as a stress-response, alarmin-like, protein. In this study, we investigated RNASET2 expression in well-differentiated and poorly differentiated neuroendocrine neoplasms of the lung (Lu-NENs), which are known to show clear-cut differences in morphology, biology and clinical behavior. In addition, we explored possible relationships between RNASET2 expression and a series of immunohistochemical markers related to hypoxic stress, apoptosis, proliferation and angiogenesis. Our results showed a significantly higher expression of RNASET2, HIF-1α, and its target CA IX in poorly differentiated than in well-differentiated Lu-NENs, the former also showing higher proliferation and apoptotic rates, as well as a lower microvessel density (MVD) than the latter. Moreover, we were able to demonstrate in vitro an overexpression of RNASET2 in consequence of the activation of HIF-1α. In conclusion, we suggest that in poorly differentiated Lu-NENs, RNASET2 expression may be induced by HIF-1α, behaving as an alarmin-like molecule. In this aggressive group of cancers, which have highly deregulated proliferation pathways, RNASET2 fails to exert the growth-inhibiting effects described in other types of neoplasms. Its increased expression, however, may contribute to the typical phenotypic alterations seen in poorly differentiated Lu-NENs, such as the high apoptotic rate and the extensive necrosis, and may also enhance the low MVD observed in these neoplasms.


Subject(s)
Carcinoid Tumor/blood supply , Carcinoid Tumor/enzymology , Cell Differentiation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/blood supply , Lung Neoplasms/enzymology , Microvessels/pathology , Neuroendocrine Tumors/blood supply , Neuroendocrine Tumors/enzymology , Ribonucleases/metabolism , Tumor Suppressor Proteins/metabolism , Antigens, Neoplasm/metabolism , Apoptosis , Carbonic Anhydrase IX/metabolism , Carcinoid Tumor/pathology , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung Neoplasms/pathology , MCF-7 Cells , Microvessels/metabolism , Necrosis , Neovascularization, Pathologic , Neuroendocrine Tumors/pathology , Ribonucleases/genetics , Tumor Hypoxia , Tumor Microenvironment , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL