14 results on '"Anna Ranghetti"'
Search Results
2. miR-511-3p Modulates Genetic Programs of Tumor-Associated Macrophages
- Author
-
Mario Leonardo Squadrito, Ferdinando Pucci, Laura Magri, Davide Moi, Gregor D. Gilfillan, Anna Ranghetti, Andrea Casazza, Massimiliano Mazzone, Robert Lyle, Luigi Naldini, and Michele De Palma
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Expression of the mannose receptor (MRC1/CD206) identifies macrophage subtypes, such as alternatively activated macrophages (AAMs) and M2-polarized tumor-associated macrophages (TAMs), which are endowed with tissue-remodeling, proangiogenic, and protumoral activity. However, the significance of MRC1 expression for TAM's protumoral activity is unclear. Here, we describe and characterize miR-511-3p, an intronic microRNA (miRNA) encoded by both mouse and human MRC1 genes. By using sensitive miRNA reporter vectors, we demonstrate robust expression and bioactivity of miR-511-3p in MRC1+ AAMs and TAMs. Unexpectedly, enforced expression of miR-511-3p tuned down the protumoral gene signature of MRC1+ TAMs and inhibited tumor growth. Our findings suggest that transcriptional activation of Mrc1 in TAMs evokes a genetic program orchestrated by miR-511-3p, which limits rather than enhances their protumoral functions. Besides uncovering a role for MRC1 as gatekeeper of TAM's protumoral genetic programs, these observations suggest that endogenous miRNAs may operate to establish thresholds for inflammatory cell activation in tumors.
- Published
- 2012
- Full Text
- View/download PDF
3. Gene expression profiles identify inflammatory signatures in dendritic cells.
- Author
-
Anna Torri, Ottavio Beretta, Anna Ranghetti, Francesca Granucci, Paola Ricciardi-Castagnoli, and Maria Foti
- Subjects
Medicine ,Science - Abstract
Dendritic cells (DCs) constitute a heterogeneous group of antigen-presenting leukocytes important in activation of both innate and adaptive immunity. We studied the gene expression patterns of DCs incubated with reagents inducing their activation or inhibition. Total RNA was isolated from DCs and gene expression profiling was performed with oligonucleotide microarrays. Using a supervised learning algorithm based on Random Forest, we generated a molecular signature of inflammation from a training set of 77 samples. We then validated this molecular signature in a testing set of 38 samples. Supervised analysis identified a set of 44 genes that distinguished very accurately between inflammatory and non inflammatory samples. The diagnostic performance of the signature genes was assessed against an independent set of samples, by qRT-PCR. Our findings suggest that the gene expression signature of DCs can provide a molecular classification for use in the selection of anti-inflammatory or adjuvant molecules with specific effects on DC activity.
- Published
- 2010
- Full Text
- View/download PDF
4. Correction: Gene Expression Profiles Identify Inflammatory Signatures in Dendritic Cells.
- Author
-
Anna Torri, Ottavio Beretta, Anna Ranghetti, Francesca Granucci, Paola Ricciardi-Castagnoli, and Maria Foti
- Subjects
Medicine ,Science - Published
- 2010
- Full Text
- View/download PDF
5. Targeted inducible delivery of immunoactivating cytokines reprograms glioblastoma microenvironment and inhibits growth in mouse models
- Author
-
Filippo Birocchi, Melania Cusimano, Federico Rossari, Stefano Beretta, Paola M. V. Rancoita, Anna Ranghetti, Stefano Colombo, Barbara Costa, Peter Angel, Francesca Sanvito, Marcella Callea, Rossana Norata, Linda Chaabane, Tamara Canu, Antonello Spinelli, Marco Genua, Renato Ostuni, Ivan Merelli, Nadia Coltella, and Luigi Naldini
- Subjects
Disease Models, Animal ,Mice ,Brain Neoplasms ,Cell Line, Tumor ,Tumor Microenvironment ,Animals ,Cytokines ,Interferon-alpha ,General Medicine ,Glioblastoma ,Interleukin-12 - Abstract
Glioblastoma multiforme (GBM) is the most common and lethal brain tumor characterized by a strongly immunosuppressive tumor microenvironment (TME) that represents a barrier also for the development of effective immunotherapies. The possibility to revert this hostile TME by immunoactivating cytokines is hampered by the severe toxicity associated with their systemic administration. Here, we exploited a lentiviral vector–based platform to engineer hematopoietic stem cells ex vivo with the aim of releasing, via their tumor-infiltrating monocyte/macrophage progeny, interferon-α (IFN-α) or interleukin-12 (IL-12) at the tumor site with spatial and temporal selectivity. Taking advantage of a syngeneic GBM mouse model, we showed that inducible release of IFN-α within the TME achieved robust tumor inhibition up to eradication and outperformed systemic treatment with the recombinant protein in terms of efficacy, tolerability, and specificity. Single-cell RNA sequencing of the tumor immune infiltrate revealed reprogramming of the immune microenvironment toward a proinflammatory and antitumoral state associated with loss of a macrophage subpopulation shown to be associated with poor prognosis in human GBM. The spatial and temporal control of IL-12 release was critical to overcome an otherwise lethal hematopoietic toxicity while allowing to fully exploit its antitumor activity. Overall, our findings demonstrate a potential therapeutic approach for GBM and set the bases for a recently launched first-in-human clinical trial in patients with GBM.
- Published
- 2022
6. Interferon gene therapy reprograms the leukemia microenvironment inducing protective immunity to multiple tumor antigens
- Author
-
Margherita Norelli, Renato Ostuni, Giulia Escobar, Barbara Camisa, Attilio Bondanza, Bernhard Gentner, Chiara Brombin, Davide Cittaro, Andrea Annoni, Tiziana Plati, Marco Genua, Luigi Naldini, Luigi Barbarossa, Giulia Barbiera, Anna Ranghetti, Escobar, G., Barbarossa, L., Barbiera, G., Norelli, M., Genua, M., Ranghetti, A., Plati, T., Camisa, B., Brombin, C., Cittaro, D., Annoni, A., Bondanza, A., Ostuni, R., Gentner, B., and Naldini, L.
- Subjects
0301 basic medicine ,Male ,Genetic enhancement ,medicine.medical_treatment ,T-Lymphocytes ,General Physics and Astronomy ,Immunotherapy, Adoptive ,0302 clinical medicine ,Interferon ,hemic and lymphatic diseases ,Tumor Microenvironment ,lcsh:Science ,Cells, Cultured ,Regulation of gene expression ,Multidisciplinary ,Gene Expression Regulation, Leukemic ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,3. Good health ,Leukemia ,030220 oncology & carcinogenesis ,Female ,medicine.drug ,Transgene ,Science ,Mice, Transgenic ,Gene delivery ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Antigens, Neoplasm ,Cell Line, Tumor ,medicine ,Animals ,Tumor microenvironment ,business.industry ,Animal ,Immunity ,General Chemistry ,Immunotherapy ,Genetic Therapy ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,T-Lymphocyte ,Cancer research ,lcsh:Q ,Interferons ,sense organs ,business - Abstract
Immunotherapy is emerging as a new pillar of cancer treatment with potential to cure. However, many patients still fail to respond to these therapies. Among the underlying factors, an immunosuppressive tumor microenvironment (TME) plays a major role. Here we show that monocyte-mediated gene delivery of IFNα inhibits leukemia in a mouse model. IFN gene therapy counteracts leukemia-induced expansion of immunosuppressive myeloid cells and imposes an immunostimulatory program to the TME, as shown by bulk and single-cell transcriptome analyses. This reprogramming promotes T-cell priming and effector function against multiple surrogate tumor-specific antigens, inhibiting leukemia growth in our experimental model. Durable responses are observed in a fraction of mice and are further increased combining gene therapy with checkpoint blockers. Furthermore, IFN gene therapy strongly enhances anti-tumor activity of adoptively transferred T cells engineered with tumor-specific TCR or CAR, overcoming suppressive signals in the leukemia TME. These findings warrant further investigations on the potential development of our gene therapy strategy towards clinical testing., An immune suppressive tumor microenvironment (TME) is a limitation for immunotherapy. Here the authors show that, in a B cell acute lymphoblastic leukemia mouse model, gene-based delivery of IFNα reprograms the leukemia-induced immunosuppressive TME into immunostimulatory and enhances T-cell responses.
- Published
- 2018
7. miRNA-126 Orchestrates an Oncogenic Program in B Cell Precursor Acute Lymphoblastic Leukemia
- Author
-
Francesco Boccalatte, Anna Ranghetti, Fabio Ciceri, Eric R. Lechman, Jose Manuel Garcia-Manteiga, Bernhard Gentner, Luigi Naldini, Silvia Nucera, Maurilio Ponzoni, Tiziana Plati, Fabrizio Benedicenti, Eugenio Montini, Andrea Calabria, Alice Giustacchini, Davide Cittaro, Cristiana Fanciullo, John E. Dick, Nucera, Silvia, Giustacchini, Alice, Boccalatte, Francesco, Calabria, Andrea, Fanciullo, Cristiana, Plati, Tiziana, Ranghetti, Anna, Garcia Manteiga, Jose, Cittaro, Davide, Benedicenti, Fabrizio, Lechman, Eric R., Dick, John E., Ponzoni, Maurilio, Ciceri, Fabio, Montini, Eugenio, Gentner, Bernhard, and Naldini, Luigi
- Subjects
0301 basic medicine ,Cancer Research ,Cellular differentiation ,Apoptosis ,Biology ,Mice ,03 medical and health sciences ,Downregulation and upregulation ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,microRNA ,medicine ,Animals ,Humans ,Progenitor cell ,B cell ,Regulation of gene expression ,Cell Cycle ,Hematopoietic Stem Cell Transplantation ,Cell Differentiation ,Neoplasms, Experimental ,Cell Biology ,Cell cycle ,Hematopoietic Stem Cells ,medicine.disease ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,B-cell leukemia ,Cancer research ,Tumor Suppressor Protein p53 ,Signal Transduction - Abstract
MicroRNA (miRNA)-126 is a known regulator of hematopoietic stem cell quiescence. We engineered murine hematopoiesis to express miRNA-126 across all differentiation stages. Thirty percent of mice developed monoclonal B cell leukemia, which was prevented or regressed when a tetracycline-repressible miRNA-126 cassette was switched off. Regression was accompanied by upregulation of cell-cycle regulators and B cell differentiation genes, and downregulation of oncogenic signaling pathways. Expression of dominant-negative p53 delayed blast clearance upon miRNA-126 switch-off, highlighting the relevance of p53 inhibition in miRNA-126 addiction. Forced miRNA-126 expression in mouse and human progenitors reduced p53 transcriptional activity through regulation of multiple p53-related targets. miRNA-126 is highly expressed in a subset of human B-ALL, and antagonizing miRNA-126 in ALL xenograft models triggered apoptosis and reduced disease burden.
- Published
- 2016
8. Abstract PR13: Development of chimeric forms of IFN-alpha for 'on demand' in vivo cancer gene therapy
- Author
-
Melania Cusimano, Peter Angel, Barbara Costa, Anna Ranghetti, Tiziano Di Tomaso, Filippo Birocchi, Nadia Coltella, and Luigi Naldini
- Subjects
Cancer Research ,Tumor microenvironment ,education.field_of_study ,Genetic enhancement ,Melanoma ,medicine.medical_treatment ,Immunology ,Population ,Immunotherapy ,Biology ,medicine.disease ,Fusion protein ,In vivo ,Glioma ,medicine ,Cancer research ,education - Abstract
The immunosuppressive tumor microenvironment is a major hurdle for cancer therapy. Our lab previously developed a cell and gene therapy strategy for the tumor-targeted delivery of IFN-alpha based on the transduction of hematopoietic stem/progenitor cells (HSPC) with lentiviral vectors (LV) expressing the IFN-alpha gene under the control of the TIE2 enhancer/promoter. The specificity of this strategy is ensured by the restricted expression of IFN-alpha in a population of HSPC-derived tumor-infiltrating macrophages expressing the TIE2 receptor (TEM), which are recruited at the tumor site. TEM-mediated delivery of IFN-alpha has shown efficacy in different tumor models of both solid and hematopoietic origin, including a model of human glioma in immunodeficient mice. New data obtained in a more relevant immunocompetent mouse model of orthotopic glioma closely reproducing several features of the human disease show dramatic tumor inhibition in mice treated with IFN-alpha gene therapy. We are currently characterizing the contribution of IFN-induced antiangiogenic and modulatory activity on the innate and adaptive immune system underlying such response. To further improve the therapeutic index of our strategy, we are developing inducible strategies to superimpose a timer and a rheostatic switch on the amount of IFN-alpha secreted in the tumor microenvironment. Indeed, whereas a sustained cytokine output could ensure efficacy and long-term protection from tumor recurrence, it may raise concerns for long-term effects, especially in case of cancer eradication. By fusing a destabilizing domain (DD) to a protein of interest (POI), the former can confer its instability to the latter. This destabilization can be rescued in a reversible and dose-dependent manner with the addition of a small molecule specifically binding to the DD. To apply this technology to our strategy, we have designed and in vitro tested different fusion proteins of IFN-alpha (DD-IFN-alpha) with or without the addition of flexible or cleavable linkers and selected them for their capacity to be stabilized in presence of their specific ligand in vitro. Through this approach, we have identified fusion proteins with low basal activity and high fold induction upon ligand treatment. These novel regulated forms of IFN-alpha are functional and their specific activities are comparable to the wild type. Based on preliminary evidence of in vivo ligand-regulated IFN release, we are now testing the safety and efficacy of our new platforms in inducing antitumor responses in melanoma, colon, and glioma models of cancer. This abstract is also being presented as Poster B31. Citation Format: Filippo Birocchi, Melania Cusimano, Anna Ranghetti, Tiziano Di Tomaso, Barbara Costa, Peter Angel, Nadia Coltella, Luigi Naldini. Development of chimeric forms of IFN-alpha for “on demand” in vivo cancer gene therapy [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr PR13.
- Published
- 2020
9. Novel members of the family Micromonosporaceae, Rugosimonospora acidiphila gen. nov., sp. nov. and Rugosimonospora africana sp. nov
- Author
-
Manfred Rohde, Linda Cavaletti, Alessandra Mezzelani, Paolo Monciardini, Ruggiero Bamonte, Peter Schumann, Anna Ranghetti, Stefano Donadio, Margherita Sosio, and Vicuron Pharmaceuticals, 21040 Gerenzano, Italy. paolo.monciardini@ktedogen.com
- Subjects
DNA, Bacterial ,Molecular Sequence Data ,DNA, Ribosomal ,Microbiology ,chemistry.chemical_compound ,RNA, Ribosomal, 16S ,Phylogeny ,Soil Microbiology ,Ecology, Evolution, Behavior and Systematics ,chemistry.chemical_classification ,biology ,Fatty Acids ,Fatty acid ,Micromonosporaceae ,General Medicine ,16S ribosomal RNA ,biology.organism_classification ,Type species ,Biochemistry ,chemistry ,Peptidoglycan ,Actinomycetales ,Diaminopimelic acid ,Bacteria - Abstract
Two novel Gram-positive-staining, acidophilic strains were isolated from soil samples. Both show typical features of filamentous actinomycetes. On the basis of 16S rRNA gene sequence analysis, the strains are members of the family Micromonosporaceae. The two strains contain hydroxydiaminopimelic acid, glycine, alanine and glutamic acid in the peptidoglycan. Fatty acid profiles clearly differentiate the two strains: cyclohexyl C(17 : 0), i-C(16 : 0) and ai-C(17 : 0) are predominant in Delta1(T), while the major components for Delta3(T) are ai-C(17 : 0) and i-C(16 : 0). The two strains also differ in their major menaquinones, MK-9(H(8), H(4), H(6)) for Delta1(T) and MK-9(H(8), H(6)) for Delta3(T), and in phospholipid patterns; Delta1(T) displays phosphatidylglycerol, diphosphatidylglycerol, phosphatidylinositol, phosphatidylethanolamine, methyl phosphatidylethanolamine and an unknown aminophospholipid, while Delta3(T) also contains minor amounts of several unknown phospholipids in addition to these phospholipids. The whole-cell sugars of both strains are galactose, arabinose and xylose. The G+C content of the DNA is 72.7 mol% for Delta1(T) and 71.9 mol% for Delta3(T). On the basis of chemotaxonomic, physiological and phylogenetic data, we propose Rugosimonospora gen. nov. to accommodate the two strains, with the description of Rugosimonospora acidiphila gen. nov., sp. nov. (the type species; type strain Delta1(T) =DSM 45227(T) =NBRC 104874(T)) and Rugosimonospora africana sp. nov. (type strain Delta3(T) =DSM 45228(T) =NBRC 104875(T)).
- Published
- 2009
10. Angiopoietin 2 expression in the cornea and its control of corneal neovascularisation
- Author
-
Chiara Giacomini, Paolo Rama, Roberta Mazzieri, Davide Moi, Claudio Doglioni, Fabio Bignami, Luigi Naldini, Anna Ranghetti, Giulio Ferrari, Ferrari, Giulio, Giacomini, Chiara, Bignami, Fabio, Moi, Davide, Ranghetti, Anna, Doglioni, C, Naldini, Luigi, Rama, Paolo, and Mazzieri, Roberta
- Subjects
0301 basic medicine ,CD31 ,Pathology ,medicine.medical_specialty ,Endothelium ,Cell Count ,Dermoscopy ,Angiopoietin-2 ,Cornea ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Stroma ,medicine ,Animals ,Humans ,Corneal Neovascularization ,business.industry ,Anatomy ,Immunohistochemistry ,eye diseases ,Sensory Systems ,Epithelium ,Lymphangiogenesis ,Ophthalmology ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Lymphatic system ,030221 ophthalmology & optometry ,Female ,sense organs ,Bowman Membrane ,business ,Biomarkers - Abstract
http://hdl.handle.net/20.500.11768/96868 Purpose To define proangiogenic angiopoietin 2 (ANG2) expression and role(s) in human and mouse vascularised corneas. Further, to evaluate the effect of ANG2 inhibition on corneal neovascularisation (CNV). Methods CNV was induced in FVB mice by means of intrastromal suture placement. One group of animals was sacrificed 10 days later; corneas were immunostained for ANG2 and compared with (i) mouse non-vascularised corneas and (ii) human vascularised and non-vascularised corneas. A second group of CNV animals was treated systemically with an anti-ANG2 antibody. After 10 days, the corneas were whole-mounted, stained for CD31 and LYVE1 and lymphatic/blood vessels quantified. In another set of experiments, the corneal basal Bowman membrane was either (i) removed or (ii) left in place. After 2 or 10 days the corneas were removed and immunostained for collagen IV, ANG2, CD31, LYVE1, CD11b and MRC1 markers. Results In human beings and mice, ANG2 is expressed only in the epithelium, and, mildly, in the endothelium, of the avascular cornea. Instead, it is expressed in the epithelium, endothelium and stroma of vascularised corneas. Disruption of the Bowman membrane is associated with a significant increase of (i) ANG2 stromal expression and (ii) proangiogenic macrophage infiltration in the corneal stroma. Finally, blocking ANG2 significantly reduced hemangiogenesis, lymphangiogenesis and macrophage infiltration. Conclusions Balancing proper healing and good vision is crucial in the cornea, constantly exposed to potential injuries. In this paper, we suggest the existence of a mechanism regulating the onset of inflammation (and associated CNV) depending on injury severity. ZB 0 Z8 0 ZR 0 ZS 0
- Published
- 2015
11. Abstract B181: Genetic engineering of tumor-infiltrating monocytes to inhibit primary and metastatic breast cancer
- Author
-
Anna Ranghetti, Davide Moi, Moustafa Sherif, Roberta Mazzieri, Luigi Naldini, Giulia Escobar, and Ryan Galea
- Subjects
Cancer Research ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Immunology ,Cancer ,Bone metastasis ,Acquired immune system ,medicine.disease ,Metastatic breast cancer ,Immune system ,Breast cancer ,Cancer immunotherapy ,medicine ,business - Abstract
The establishment of an immunosuppressive microenvironment is one of the hallmarks of cancer and the major impediment to the successful application of cancer immunotherapy. By exploiting the tumor homing ability of a subpopulation of monocytes we turned them into an efficient vehicle for the tumor-targeted delivery of a potent immune-stimulatory molecule: interferon-alpha (IFNα);. We are now combining the ability of IFNα to reverse the immunosuppressive tumor microenvironment with established and new cancer immunotherapies for the treatment of advanced breast cancer, including disseminated lung and bone metastasis. We identified and characterized a specific subpopulation of pro-tumoral macrophages characterized by the expression of the Angiopoietin receptor TIE2 (TEMs: TIE2-expressing monocytes/macrophages) and endowed with pro-angiogenic and immunosuppressive activities. As a strategy aimed at reversing the immunosuppressive microenvironment, we exploited the tumor homing ability and selective expression of the TIE2 receptor to turn TEMs into IFNα delivery vehicles by engineering hematopoietic stem cells (HSCs) to express an IFNα transgene under the control of the Tie2 promoter. To progress this strategy to clinical application we recently developed a humanized vector expressing a human IFNα gene, and have studied the feasibility and safety of engineering human HSCs for its expression in their TEM progeny. Moreover, to demonstrate the efficacy of our human IFNα delivery platform, we challenged human hematochimeric mice with a human breast cancer cell line. Type I interferons (IFNs) are pleiotropic cytokines involved in innate and adaptive immunity that have been shown to promote anti-tumor immune responses. The broad anti-tumor biological activities of type I IFNs have provided the rationale for testing administration of exogenous IFNα as an anti-cancer treatment, which has proven effective against several solid and hematological malignancies. However, clinical use of IFNα has since declined because of the substantial toxicity associated with systemic administration and the limited efficacy at the maximal tolerated doses, thus calling for safer and more effective delivery strategies. Our cell- and gene-based IFNα delivery strategy strongly inhibited primary breast cancer tumors and breast cancer lung metastasis in mouse and human hematochimeric models by promoting the recruitment and activation of both innate and adaptive immune cells with no evident signs of toxicity. Moreover, our studies validated the feasibility, safety and therapeutic potential of a new HSC-based gene therapy strategy to treat established tumors, and could, therefore, revive interest in testing high-dose chemotherapy followed by autologous HSC transplantation in advanced breast cancer patients. Importantly, we think that the multiple activities of type I IFNs in the complex network of cell interactions that lead to activation and deployment of immune responses may represent a valid strategy to promote and improve the outcome of cancer immunotherapy for the treatment of advanced breast cancer including lung and bone metastasis. Preliminary data on new therapeutic strategies combining TEM-mediated delivery of IFNα with monoclonal antibodies against inhibitory checkpoint molecules such as PD1 and CTLA4, will be presented. Citation Format: Giulia Escobar, Davide Moi, Ryan Galea, Moustafa Sherif, Anna Ranghetti, Luigi Naldini, Roberta Mazzieri. Genetic engineering of tumor-infiltrating monocytes to inhibit primary and metastatic breast cancer. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B181.
- Published
- 2016
12. miR-511-3p modulates genetic programs of tumor-associated macrophages
- Author
-
Luigi Naldini, Davide Moi, Anna Ranghetti, Mario Leonardo Squadrito, Massimiliano Mazzone, Laura Magri, Ferdinando Pucci, Michele De Palma, Andrea Casazza, Gregor D. Gilfillan, Robert Lyle, Squadrito, M. L., Pucci, F., Magri, L., Moi, D., Gilfillan, G. D., Ranghetti, A., Casazza, A, Mazzone, M., Lyle, R., Naldini, L., and De Palma, M.
- Subjects
Inbred C57BL ,Mice ,0302 clinical medicine ,Lectins ,Neoplasms ,Receptors ,RNA Precursors ,Macrophage ,Animals ,Base Pairing ,Base Sequence ,Bone Marrow Cells ,Cell Line ,Cell Line, Tumor ,Down-Regulation ,Gene Expression Profiling ,Gene Expression Regulation, Neoplastic ,Hematopoiesis ,Humans ,Immunophenotyping ,Lectins, C-Type ,Macrophages ,Mannose Receptor ,Mannose-Binding Lectins ,Mice, Inbred C57BL ,MicroRNAs ,Molecular Sequence Data ,Nucleic Acid Conformation ,Receptors, Cell Surface ,rho-Associated Kinases ,Receptor ,lcsh:QH301-705.5 ,Regulation of gene expression ,0303 health sciences ,Tumor ,C-Type ,030220 oncology & carcinogenesis ,Cell Surface ,Mannose receptor ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,stomatognathic system ,microRNA ,Gene ,030304 developmental biology ,Neoplastic ,Gene signature ,Molecular biology ,Gene expression profiling ,lcsh:Biology (General) ,Gene Expression Regulation ,Cancer research - Abstract
Expression of the mannose receptor (MRC1/CD206) identifies macrophage subtypes, such as alternatively activated macrophages (AAMs) and M2-polarized tumor-associated macrophages (TAMs), which are endowed with tissue-remodeling, proangiogenic, and protumoral activity. However, the significance of MRC1 expression for TAM's protumoral activity is unclear. Here, we describe and characterize miR-511-3p, an intronic microRNA (miRNA) encoded by both mouse and human MRC1 genes. By using sensitive miRNA reporter vectors, we demonstrate robust expression and bioactivity of miR-511-3p in MRC1(+) AAMs and TAMs. Unexpectedly, enforced expression of miR-511-3p tuned down the protumoral gene signature of MRC1(+) TAMs and inhibited tumor growth. Our findings suggest that transcriptional activation of Mrc1 in TAMs evokes a genetic program orchestrated by miR-511-3p, which limits rather than enhances their protumoral functions. Besides uncovering a role for MRC1 as gatekeeper of TAM's protumoral genetic programs, these observations suggest that endogenous miRNAs may operate to establish thresholds for inflammatory cell activation in tumors. ispartof: Cell reports vol:1 issue:2 pages:141-154 ispartof: location:United States status: published
- Published
- 2012
13. Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells
- Author
-
Roberta Mazzieri, Jeffrey L. Brown, Michele De Palma, Luigi Naldini, Anna Ranghetti, Alvise Berti, Bernhard Gentner, Erika Zonari, Ferdinando Pucci, Davide Moi, Letterio S. Politi, Mazzieri, R, Pucci, F, Moi, D, Zonari, E, Ranghetti, A, Berti, A, Politi, L, Gentner, B, Brown, Jl, Naldini, Luigi, and De Palma, M.
- Subjects
Cell signaling ,Cancer Research ,Angiogenesis ,Cell Communication ,Metastasis ,Neovascularization ,Angiopoietin-2 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Medicine ,Animals ,Humans ,Myeloid Cells ,Neoplasm Metastasis ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Gene knockdown ,biology ,Neovascularization, Pathologic ,business.industry ,Macrophages ,Endothelial Cells ,Mammary Neoplasms, Experimental ,Receptor Protein-Tyrosine Kinases ,Neoplasms, Experimental ,Cell Biology ,medicine.disease ,Adenoma, Islet Cell ,Angiopoietin receptor ,Receptor, TIE-2 ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Neuroendocrine Tumors ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,embryonic structures ,Cancer research ,biology.protein ,cardiovascular system ,medicine.symptom ,business - Abstract
SummaryTumor-infiltrating myeloid cells convey proangiogenic programs that counteract the efficacy of antiangiogenic therapy. Here, we show that blocking angiopoietin-2 (ANG2), a TIE2 ligand and angiogenic factor expressed by activated endothelial cells (ECs), regresses the tumor vasculature and inhibits progression of late-stage, metastatic MMTV-PyMT mammary carcinomas and RIP1-Tag2 pancreatic insulinomas. ANG2 blockade did not inhibit recruitment of MRC1+ TIE2-expressing macrophages (TEMs) but impeded their upregulation of Tie2, association with blood vessels, and ability to restore angiogenesis in tumors. Conditional Tie2 gene knockdown in TEMs was sufficient to decrease tumor angiogenesis. Our findings support a model wherein the ANG2-TIE2 axis mediates cell-to-cell interactions between TEMs and ECs that are important for tumor angiogenesis and can be targeted to induce effective antitumor responses.
- Published
- 2011
14. A Mechanistic Role For Mir-126, a Hematopoietic Stem Cell Microrna, In Acute Leukemias
- Author
-
Eric R. Lechman, Luigi Naldini, Bernhard Gentner, Francesco Boccalatte, Tiziana Plati, Alice Giustacchini, Anna Ranghetti, John E. Dick, Maurilio Ponzoni, Cristiana Fanciullo, Massimo Bernardi, Luca Vago, and Silvia Nucera
- Subjects
Acute leukemia ,biology ,Immunology ,Hematopoietic stem cell ,Cell Biology ,Hematology ,CD38 ,medicine.disease ,Biochemistry ,CD19 ,medicine.anatomical_structure ,Acute lymphocytic leukemia ,medicine ,Cancer research ,biology.protein ,Stem cell ,Progenitor cell ,B cell - Abstract
We have recently shown that miR-126 expression faithfully identifies the engrafting fraction of bone marrow (BM) and cord blood (Gentner et al, Sci Transl Med 2010), in which it regulates hematopoietic stem cell (HSC) pool size by modulating cell cycle progression (Lechman et al., Cell Stem Cell 2012). Antagonizing miR-126 expands HSC by enhancing PI3K/AKT signaling, without causing their exhaustion or transformation. By reconstituting mice with lentivirally transduced BM cells ectopically expressing miR-126, we noted differential effects in HSC and progenitors: while miR-126 overexpressing (126OE) HSC were more quiescent and outcompeted by HSC with physiologic miR-126 expression, progenitor subsets were increased in number and proliferated more upon 126OE. In particular, B lymphopoiesis was enhanced, with the appearance of 126OE-vector marked, CD45(low)CD19+Ig(-) B cell progenitors in the peripheral blood. This abnormal population was transiently observed in all 126OE (n=45), but in none of the control mice (n=30), and BM B cell precursors and progenitors from 126OE mice showed significantly reduced levels of apoptosis, as well as a differentiation block at the immature B cell stage. At least part of this effect was due to interference of miR-126 with p53 activation. Consequentially, 20-30% of 126OE mice (as compared to 0% of control mice) followed over 1 year developed vector-marked, high-grade B-cell neoplasms which resembled diffuse large B-cell lymphoma, lymphoblastic lymphoma or acute lymphoblastic leukemia (ALL). Strikingly, by using a tetracycline-regulated, conditional miR-126OE vector, we observed full regression of the expanded abnormal B cell population after switching miR-126 expression off by doxycycline administration, demonstrating that miR-126 is an oncogenic driver in hematologic malignancies. To establish the relevance of miR-126 in human disease, we studied primary samples from patients with acute leukemia. We measured miR-126 expression in blasts from 12 adult patients with acute lymphoblastic leukemia (ALL). miR-126 was highly expressed in all studied cases of ALL (Phil+: n=7, Phil-: n=5), often surpassing the levels found in normal HSC. Unlike in HSC, miR-126 expression was independent from expression of its host gene EGFL7, suggesting an ALL-specific regulation of the miR-126 locus. By exploiting a lentiviral miR-126 reporter vector, we quantified miR-126 activity with single cell resolution after xenotransplantation of miR-126 reporter vector transduced ALL blasts into NSG mice. In 7 out of 12 ALL cases, we identified distinct subpopulations exhibiting different levels of miR-126 activity, and miR-126 (hi) ALL cells were more frequently contained in the CD34+ cell fraction. Moreover, high miR-126 expression correlated with faster and higher ALL engraftment in NSG mice. In acute myeloid leukemia (AML), miR-126 is significantly enriched in leukemic stem cells (LSC) where it governs quiescence and chemotherapy resistance (see abstract by Lechman et al), and analogous studies in ALL are ongoing. To further substantiate the role of miR-126 in AML, we studied miR-126 activity in 11 primary AML cases (4 AML with CBF, core binding factor, mutations; 7 AML with normal karyotype) using the reporter vector technology and the NSG xenotransplantation model. Globally, we found highest miR-126 activity in CBF-mutated AML. However, there was substantial heterogeneity in subpopulations of each single disease. Significantly higher miR-126 levels were found in CD34+, and particularly in CD34+CD38- AML cells, a fraction that is enriched for LSC. To see whether there is a correlation between miR-126 expression and disease progression, we measured miR-126 levels in sorted blasts from paired diagnosis/relapse (n=6) and diagnosis/chemotherapy-refractory (n=8) patient samples. Strikingly, we found a significant up-regulation of miR-126 in the disease progression sample, suggesting a mechanistic role for miR-126 in AML persistence after chemotherapy. In summary, these data support a clinically relevant role of miR-126 in the pathogenesis, treatment response and progession of acute leukemias. Authors AG and SN contributed equally, and LN and BG contributed equally. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.