12 results on '"Tripodo, C"'
Search Results
2. Inhibition of DNA damage response at telomeres improves the detrimental phenotypes of Hutchinson–Gilford Progeria Syndrome
- Author
-
Giovanna Lattanzi, Fabrizio d'Adda di Fagagna, Peh Fern Ong, Corey Winston Jones-Weinert, Oliver Dreesen, Claudio Tripodo, Agustin Sola-Carvajal, Maria Eriksson, Francesca Rossiello, Emelie Wallén Arzt, Gwladys Revêchon, Valeria Cancila, Julio Aguado, Aguado J., Sola-Carvajal A., Cancila V., Revechon G., Ong P.F., Jones-Weinert C.W., Wallen Arzt E., Lattanzi G., Dreesen O., Tripodo C., Rossiello F., Eriksson M., and d'Adda di Fagagna F.
- Subjects
0301 basic medicine ,Genome instability ,RNA, Untranslated ,DNA Repair ,General Physics and Astronomy ,Cellular homeostasis ,Antisense oligonucleotide therapy ,Mice ,0302 clinical medicine ,Progeria ,Homeostasis ,lcsh:Science ,Cellular Senescence ,Skin ,Multidisciplinary ,integumentary system ,Telomere ,Progerin ,Lamin Type A ,3. Good health ,Cell biology ,Telomeres ,Phenotype ,Premature aging ,congenital, hereditary, and neonatal diseases and abnormalities ,DNA repair ,Science ,Double-strand DNA breaks ,Biology ,Settore MED/08 - Anatomia Patologica ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,medicine ,DNA damage, Hutchinson-Gilford Progeria Syndrome ,Animals ,Cell Proliferation ,nutritional and metabolic diseases ,General Chemistry ,Oligonucleotides, Antisense ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Mutation ,lcsh:Q ,030217 neurology & neurosurgery ,Lamin ,DNA Damage - Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a genetic disorder characterized by premature aging features. Cells from HGPS patients express progerin, a truncated form of Lamin A, which perturbs cellular homeostasis leading to nuclear shape alterations, genome instability, heterochromatin loss, telomere dysfunction and premature entry into cellular senescence. Recently, we reported that telomere dysfunction induces the transcription of telomeric non-coding RNAs (tncRNAs) which control the DNA damage response (DDR) at dysfunctional telomeres. Here we show that progerin-induced telomere dysfunction induces the transcription of tncRNAs. Their functional inhibition by sequence-specific telomeric antisense oligonucleotides (tASOs) prevents full DDR activation and premature cellular senescence in various HGPS cell systems, including HGPS patient fibroblasts. We also show in vivo that tASO treatment significantly enhances skin homeostasis and lifespan in a transgenic HGPS mouse model. In summary, our results demonstrate an important role for telomeric DDR activation in HGPS progeroid detrimental phenotypes in vitro and in vivo., Hutchinson–Gilford progeria syndrome causes premature aging. Here the authors show that activation of the DNA damage response at dysfunctional telomeres and transcription of telomeric non-coding RNAs contributes to the pathogenesis, which can be ameliorated by treatment with sequence-specific telomeric antisense oligonucleotides.
- Published
- 2019
3. C1q acts in the tumour microenvironment as a cancer-promoting factor independently of complement activation
- Author
-
Damiano Rami, Roberta Bulla, Claudio Tripodo, Marina Botto, Carla Guarnotta, Guang Sheng Ling, Paolo Durigutto, Sonia Zorzet, Chiara Agostinis, Francesco Tedesco, Bulla, Roberta, Tripodo, Claudio, Rami, Damiano, Ling, Guang Sheng, Agostinis, Chiara, Guarnotta, Carla, Zorzet, Sonia, Durigutto, Paolo, Botto, Marina, Tedesco, Francesco, Bulla, R., Tripodo, C., Rami, D., Ling, G., Agostinis, C., Guarnotta, C., Zorzet, S., Durigutto, P., Botto, M., Tedesco, F., and Wellcome Trust
- Subjects
Genetics and Molecular Biology (all) ,0301 basic medicine ,PROTEIN ,General Physics and Astronomy ,MELANOMA ,Apoptosis ,Inbred C57BL ,Biochemistry ,DISEASE ,Animals ,Cell Line, Tumor ,Cell Movement ,Cell Proliferation ,Complement Activation ,Complement C1q ,Complement C3 ,Complement C5 ,Humans ,Mice ,Mice, Inbred C57BL ,Mice, Knockout ,Neoplasms ,Biochemistry, Genetics and Molecular Biology (all) ,Chemistry (all) ,Physics and Astronomy (all) ,fluids and secretions ,immune system diseases ,IMMUNE-RESPONSE ,skin and connective tissue diseases ,Complement component 5 ,Tumor ,Multidisciplinary ,3. Good health ,Cell biology ,Multidisciplinary Sciences ,DEFICIENCY ,medicine.anatomical_structure ,Science & Technology - Other Topics ,Human ,Knockout ,Science ,chemical and pharmacologic phenomena ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,TROPHOBLAST INVASION ,MECHANISMS ,Cell Line ,03 medical and health sciences ,Classical complement pathway ,Immune system ,INFLAMMATION ,medicine ,Science & Technology ,Animal ,Cell growth ,EFFECTOR SYSTEM ,Apoptosi ,General Chemistry ,Complement system ,030104 developmental biology ,Cancer cell ,Neoplasm ,Bone marrow ,ANTIBODY THERAPY - Abstract
Complement C1q is the activator of the classical pathway. However, it is now recognized that C1q can exert functions unrelated to complement activation. Here we show that C1q, but not C4, is expressed in the stroma and vascular endothelium of several human malignant tumours. Compared with wild-type (WT) or C3- or C5-deficient mice, C1q-deficient (C1qa−/−) mice bearing a syngeneic B16 melanoma exhibit a slower tumour growth and prolonged survival. This effect is not attributable to differences in the tumour-infiltrating immune cells. Tumours developing in WT mice display early deposition of C1q, higher vascular density and an increase in the number of lung metastases compared with C1qa−/− mice. Bone marrow (BM) chimeras between C1qa−/− and WT mice identify non-BM-derived cells as the main local source of C1q that can promote cancer cell adhesion, migration and proliferation. Together these findings support a role for locally synthesized C1q in promoting tumour growth., C1q is known to initiate the activation of the complement classical pathway. Here, the authors show the C1q is expressed in the tumour microenvironment and can promote cancer cell migration and adhesion in a complement activation-independent manner.
- Published
- 2016
4. Unsupervised representation learning of chromatin images identifies changes in cell state and tissue organization in DCIS.
- Author
-
Zhang X, Venkatachalapathy S, Paysan D, Schaerer P, Tripodo C, Uhler C, and Shivashankar GV
- Subjects
- Humans, Female, Biomarkers, Tumor metabolism, Biomarkers, Tumor genetics, Unsupervised Machine Learning, Image Processing, Computer-Assisted methods, Tissue Array Analysis, Neoplasm Staging, Carcinoma, Intraductal, Noninfiltrating pathology, Carcinoma, Intraductal, Noninfiltrating genetics, Carcinoma, Intraductal, Noninfiltrating metabolism, Chromatin metabolism, Breast Neoplasms pathology, Breast Neoplasms genetics, Breast Neoplasms metabolism
- Abstract
Ductal carcinoma in situ (DCIS) is a pre-invasive tumor that can progress to invasive breast cancer, a leading cause of cancer death. We generate a large-scale tissue microarray dataset of chromatin images, from 560 samples from 122 female patients in 3 disease stages and 11 phenotypic categories. Using representation learning on chromatin images alone, without multiplexed staining or high-throughput sequencing, we identify eight morphological cell states and tissue features marking DCIS. All cell states are observed in all disease stages with different proportions, indicating that cell states enriched in invasive cancer exist in small fractions in normal breast tissue. Tissue-level analysis reveals significant changes in the spatial organization of cell states across disease stages, which is predictive of disease stage and phenotypic category. Taken together, we show that chromatin imaging represents a powerful measure of cell state and disease stage of DCIS, providing a simple and effective tumor biomarker., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. Spatially-resolved transcriptomics reveal macrophage heterogeneity and prognostic significance in diffuse large B-cell lymphoma.
- Author
-
Liu M, Bertolazzi G, Sridhar S, Lee RX, Jaynes P, Mulder K, Syn N, Hoppe MM, Fan S, Peng Y, Thng J, Chua R, Jayalakshmi, Batumalai Y, De Mel S, Poon L, Chan EHL, Lee J, Hue SS, Chang ST, Chuang SS, Chandy KG, Ye X, Pan-Hammarström Q, Ginhoux F, Chee YL, Ng SB, Tripodo C, and Jeyasekharan AD
- Subjects
- Humans, Prognosis, Gene Expression Profiling, Transcriptome, Germinal Center pathology, Tumor Microenvironment genetics, Lymphoma, Large B-Cell, Diffuse pathology
- Abstract
Macrophages are abundant immune cells in the microenvironment of diffuse large B-cell lymphoma (DLBCL). Macrophage estimation by immunohistochemistry shows varying prognostic significance across studies in DLBCL, and does not provide a comprehensive analysis of macrophage subtypes. Here, using digital spatial profiling with whole transcriptome analysis of CD68+ cells, we characterize macrophages in distinct spatial niches of reactive lymphoid tissues (RLTs) and DLBCL. We reveal transcriptomic differences between macrophages within RLTs (light zone /dark zone, germinal center/ interfollicular), and between disease states (RLTs/ DLBCL), which we then use to generate six spatially-derived macrophage signatures (MacroSigs). We proceed to interrogate these MacroSigs in macrophage and DLBCL single-cell RNA-sequencing datasets, and in gene-expression data from multiple DLBCL cohorts. We show that specific MacroSigs are associated with cell-of-origin subtypes and overall survival in DLBCL. This study provides a spatially-resolved whole-transcriptome atlas of macrophages in reactive and malignant lymphoid tissues, showing biological and clinical significance., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. Author Correction: Mutant p53 sustains serine-glycine synthesis and essential amino acids intake promoting breast cancer growth.
- Author
-
Tombari C, Zannini A, Bertolio R, Pedretti S, Audano M, Triboli L, Cancila V, Vacca D, Caputo M, Donzelli S, Segatto I, Vodret S, Piazza S, Rustighi A, Mantovani F, Belletti B, Baldassarre G, Blandino G, Tripodo C, Bicciato S, Mitro N, and Del Sal G
- Published
- 2023
- Full Text
- View/download PDF
7. Mutant p53 sustains serine-glycine synthesis and essential amino acids intake promoting breast cancer growth.
- Author
-
Tombari C, Zannini A, Bertolio R, Pedretti S, Audano M, Triboli L, Cancila V, Vacca D, Caputo M, Donzelli S, Segatto I, Vodret S, Piazza S, Rustighi A, Mantovani F, Belletti B, Baldassarre G, Blandino G, Tripodo C, Bicciato S, Mitro N, and Del Sal G
- Subjects
- Female, Humans, Amino Acids metabolism, Amino Acids, Essential, Glycine, Large Neutral Amino Acid-Transporter 1 genetics, Serine, Breast Neoplasms pathology, Tumor Suppressor Protein p53 genetics
- Abstract
Reprogramming of amino acid metabolism, sustained by oncogenic signaling, is crucial for cancer cell survival under nutrient limitation. Here we discovered that missense mutant p53 oncoproteins stimulate de novo serine/glycine synthesis and essential amino acids intake, promoting breast cancer growth. Mechanistically, mutant p53, unlike the wild-type counterpart, induces the expression of serine-synthesis-pathway enzymes and L-type amino acid transporter 1 (LAT1)/CD98 heavy chain heterodimer. This effect is exacerbated by amino acid shortage, representing a mutant p53-dependent metabolic adaptive response. When cells suffer amino acids scarcity, mutant p53 protein is stabilized and induces metabolic alterations and an amino acid transcriptional program that sustain cancer cell proliferation. In patient-derived tumor organoids, pharmacological targeting of either serine-synthesis-pathway and LAT1-mediated transport synergizes with amino acid shortage in blunting mutant p53-dependent growth. These findings reveal vulnerabilities potentially exploitable for tackling breast tumors bearing missense TP53 mutations., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
8. Epigenomic landscape of human colorectal cancer unveils an aberrant core of pan-cancer enhancers orchestrated by YAP/TAZ.
- Author
-
Della Chiara G, Gervasoni F, Fakiola M, Godano C, D'Oria C, Azzolin L, Bonnal RJP, Moreni G, Drufuca L, Rossetti G, Ranzani V, Bason R, De Simone M, Panariello F, Ferrari I, Fabbris T, Zanconato F, Forcato M, Romano O, Caroli J, Gruarin P, Sarnicola ML, Cordenonsi M, Bardelli A, Zucchini N, Ceretti AP, Mariani NM, Cassingena A, Sartore-Bianchi A, Testa G, Gianotti L, Opocher E, Pisati F, Tripodo C, Macino G, Siena S, Bicciato S, Piccolo S, and Pagani M
- Subjects
- Gene Expression Regulation, Neoplastic, Histone Code, Humans, Models, Genetic, Organoids metabolism, RNA-Seq, Single-Cell Analysis, Transcriptional Coactivator with PDZ-Binding Motif Proteins, Tumor Cells, Cultured, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing genetics, Colorectal Neoplasms genetics, Enhancer Elements, Genetic, Epigenesis, Genetic, Trans-Activators genetics, Transcription Factors genetics
- Abstract
Cancer is characterized by pervasive epigenetic alterations with enhancer dysfunction orchestrating the aberrant cancer transcriptional programs and transcriptional dependencies. Here, we epigenetically characterize human colorectal cancer (CRC) using de novo chromatin state discovery on a library of different patient-derived organoids. By exploring this resource, we unveil a tumor-specific deregulated enhancerome that is cancer cell-intrinsic and independent of interpatient heterogeneity. We show that the transcriptional coactivators YAP/TAZ act as key regulators of the conserved CRC gained enhancers. The same YAP/TAZ-bound enhancers display active chromatin profiles across diverse human tumors, highlighting a pan-cancer epigenetic rewiring which at single-cell level distinguishes malignant from normal cell populations. YAP/TAZ inhibition in established tumor organoids causes extensive cell death unveiling their essential role in tumor maintenance. This work indicates a common layer of YAP/TAZ-fueled enhancer reprogramming that is key for the cancer cell state and can be exploited for the development of improved therapeutic avenues.
- Published
- 2021
- Full Text
- View/download PDF
9. Mutant p53 induces Golgi tubulo-vesiculation driving a prometastatic secretome.
- Author
-
Capaci V, Bascetta L, Fantuz M, Beznoussenko GV, Sommaggio R, Cancila V, Bisso A, Campaner E, Mironov AA, Wiśniewski JR, Ulloa Severino L, Scaini D, Bossi F, Lees J, Alon N, Brunga L, Malkin D, Piazza S, Collavin L, Rosato A, Bicciato S, Tripodo C, Mantovani F, and Del Sal G
- Subjects
- Animals, Biopsy, Breast Neoplasms pathology, Cell Line, Tumor, Cell Transformation, Neoplastic metabolism, Female, Fibroblasts, Gene Expression Regulation, Neoplastic, Golgi Apparatus metabolism, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Li-Fraumeni Syndrome pathology, Mice, Microtubules metabolism, Microtubules pathology, Mutation, Primary Cell Culture, Secretory Vesicles metabolism, Secretory Vesicles pathology, Signal Transduction genetics, Skin cytology, Skin pathology, Tumor Microenvironment genetics, Xenograft Model Antitumor Assays, Breast Neoplasms genetics, Cell Transformation, Neoplastic genetics, Golgi Apparatus pathology, Li-Fraumeni Syndrome genetics, MicroRNAs metabolism, Tumor Suppressor Protein p53 genetics
- Abstract
TP53 missense mutations leading to the expression of mutant p53 oncoproteins are frequent driver events during tumorigenesis. p53 mutants promote tumor growth, metastasis and chemoresistance by affecting fundamental cellular pathways and functions. Here, we demonstrate that p53 mutants modify structure and function of the Golgi apparatus, culminating in the increased release of a pro-malignant secretome by tumor cells and primary fibroblasts from patients with Li-Fraumeni cancer predisposition syndrome. Mechanistically, interacting with the hypoxia responsive factor HIF1α, mutant p53 induces the expression of miR-30d, which in turn causes tubulo-vesiculation of the Golgi apparatus, leading to enhanced vesicular trafficking and secretion. The mut-p53/HIF1α/miR-30d axis potentiates the release of soluble factors and the deposition and remodeling of the ECM, affecting mechano-signaling and stromal cells activation within the tumor microenvironment, thereby enhancing tumor growth and metastatic colonization.
- Published
- 2020
- Full Text
- View/download PDF
10. IRSp53 controls plasma membrane shape and polarized transport at the nascent lumen in epithelial tubules.
- Author
-
Bisi S, Marchesi S, Rizvi A, Carra D, Beznoussenko GV, Ferrara I, Deflorian G, Mironov A, Bertalot G, Pisati F, Oldani A, Cattaneo A, Saberamoli G, Pece S, Viale G, Bachi A, Tripodo C, Scita G, and Disanza A
- Subjects
- Actins metabolism, Cell Polarity genetics, Cell Polarity physiology, Epithelial Cells metabolism, Female, Humans, Morphogenesis genetics, Morphogenesis physiology, Nerve Tissue Proteins genetics, Protein Transport genetics, Protein Transport physiology, Sialoglycoproteins genetics, Sialoglycoproteins metabolism, rab GTP-Binding Proteins genetics, Cell Membrane metabolism, Nerve Tissue Proteins metabolism, rab GTP-Binding Proteins metabolism
- Abstract
It is unclear whether the establishment of apical-basal cell polarity during the generation of epithelial lumens requires molecules acting at the plasma membrane/actin interface. Here, we show that the I-BAR-containing IRSp53 protein controls lumen formation and the positioning of the polarity determinants aPKC and podocalyxin. Molecularly, IRSp53 acts by regulating the localization and activity of the small GTPase RAB35, and by interacting with the actin capping protein EPS8. Using correlative light and electron microscopy, we further show that IRSp53 ensures the shape and continuity of the opposing plasma membrane of two daughter cells, leading to the formation of a single apical lumen. Genetic removal of IRSp53 results in abnormal renal tubulogenesis, with altered tubular polarity and architectural organization. Thus, IRSp53 acts as a membrane curvature-sensing platform for the assembly of multi-protein complexes that control the trafficking of apical determinants and the integrity of the luminal plasma membrane.
- Published
- 2020
- Full Text
- View/download PDF
11. Inhibition of DNA damage response at telomeres improves the detrimental phenotypes of Hutchinson-Gilford Progeria Syndrome.
- Author
-
Aguado J, Sola-Carvajal A, Cancila V, Revêchon G, Ong PF, Jones-Weinert CW, Wallén Arzt E, Lattanzi G, Dreesen O, Tripodo C, Rossiello F, Eriksson M, and d'Adda di Fagagna F
- Subjects
- Animals, Cell Line, Cell Proliferation, Cellular Senescence, DNA Repair, Disease Models, Animal, Homeostasis, Lamin Type A genetics, Lamin Type A metabolism, Mice, Mutation genetics, Oligonucleotides, Antisense metabolism, Phenotype, RNA, Untranslated genetics, RNA, Untranslated metabolism, Skin pathology, DNA Damage, Progeria pathology, Telomere metabolism
- Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a genetic disorder characterized by premature aging features. Cells from HGPS patients express progerin, a truncated form of Lamin A, which perturbs cellular homeostasis leading to nuclear shape alterations, genome instability, heterochromatin loss, telomere dysfunction and premature entry into cellular senescence. Recently, we reported that telomere dysfunction induces the transcription of telomeric non-coding RNAs (tncRNAs) which control the DNA damage response (DDR) at dysfunctional telomeres. Here we show that progerin-induced telomere dysfunction induces the transcription of tncRNAs. Their functional inhibition by sequence-specific telomeric antisense oligonucleotides (tASOs) prevents full DDR activation and premature cellular senescence in various HGPS cell systems, including HGPS patient fibroblasts. We also show in vivo that tASO treatment significantly enhances skin homeostasis and lifespan in a transgenic HGPS mouse model. In summary, our results demonstrate an important role for telomeric DDR activation in HGPS progeroid detrimental phenotypes in vitro and in vivo.
- Published
- 2019
- Full Text
- View/download PDF
12. C1q acts in the tumour microenvironment as a cancer-promoting factor independently of complement activation.
- Author
-
Bulla R, Tripodo C, Rami D, Ling GS, Agostinis C, Guarnotta C, Zorzet S, Durigutto P, Botto M, and Tedesco F
- Subjects
- Animals, Apoptosis, Cell Line, Tumor, Cell Movement, Cell Proliferation, Complement C1q genetics, Complement C3 genetics, Complement C3 metabolism, Complement C5 genetics, Complement C5 metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Complement Activation physiology, Complement C1q metabolism, Neoplasms metabolism
- Abstract
Complement C1q is the activator of the classical pathway. However, it is now recognized that C1q can exert functions unrelated to complement activation. Here we show that C1q, but not C4, is expressed in the stroma and vascular endothelium of several human malignant tumours. Compared with wild-type (WT) or C3- or C5-deficient mice, C1q-deficient (C1qa(-/-)) mice bearing a syngeneic B16 melanoma exhibit a slower tumour growth and prolonged survival. This effect is not attributable to differences in the tumour-infiltrating immune cells. Tumours developing in WT mice display early deposition of C1q, higher vascular density and an increase in the number of lung metastases compared with C1qa(-/-) mice. Bone marrow (BM) chimeras between C1qa(-/-) and WT mice identify non-BM-derived cells as the main local source of C1q that can promote cancer cell adhesion, migration and proliferation. Together these findings support a role for locally synthesized C1q in promoting tumour growth.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.