50 results on '"Mattia Bugatti"'
Search Results
2. Data from A Population of TIM4+FOLR2+ Macrophages Localized in Tertiary Lymphoid Structures Correlates to an Active Immune Infiltrate Across Several Cancer Types
- Author
-
William Vermi, Federica Benvenuti, Julie Helft, Yoann Missolo-Koussou, Nicoletta Caronni, Sara Picinoli, Irene Pezzali, Laura Ardighieri, Matilde Monti, Francesco Missale, Marco Bergamini, and Mattia Bugatti
- Abstract
TIM4 has previously been associated with antitumor immunity, yet the pattern of expression and the function of this receptor across human cancer tissues remain poorly explored. Here we combined extensive immunolabeling of human tissues with in silico analysis of pan-cancer transcriptomic data sets to explore the clinical significance of TIM4 expression. Our results unveil that TIM4 is expressed on a fraction of cavity macrophages (CATIM4+MΦ) of carcinoma patients. Moreover, we uncover a high expression of TIM4 on macrophages of the T-cell zone of the carcinoma-associated tertiary lymphoid structures (TLSTIM4+MΦ). In silico analysis of a pan-cancer data set revealed a positive correlation between TIM4 expression and markers of B cells, effector CD8+ T cells, and a 12-chemokine signature defining tertiary lymphoid structure. In addition, TLSTIM4+MΦ were enriched in cancers displaying microsatellite instability and high CD8+ T-cell infiltration, confirming their association with immune-reactive tumors. Both CATIM4+MΦ and TLSTIM4+MΦ express FOLR2, a marker of tissue-resident MΦ. However, CATIM4+MΦ had a higher expression of the immunosuppressive molecules TREM2, IL10, and TGFβ as compared with TLSTIM4+MΦ. By analyzing a scRNA sequence data set of tumor-associated myeloid cells, we identified two TIM4+FOLR2+ clusters coherent with CATIM4+MΦ and TLSTIM4+MΦ. We defined specific gene signatures for each subset and found that the CATIM4+ MΦ signature was associated with worse patient survival. In contrast, TLSTIM4+MΦ gene signature positively correlates with a better prognosis. Together, these data illustrate that TIM4 marks two distinct macrophage populations with distinct phenotypes and tissue localization and that may have opposing roles in tumor immunity.
- Published
- 2023
3. Supplementary Table from A Population of TIM4+FOLR2+ Macrophages Localized in Tertiary Lymphoid Structures Correlates to an Active Immune Infiltrate Across Several Cancer Types
- Author
-
William Vermi, Federica Benvenuti, Julie Helft, Yoann Missolo-Koussou, Nicoletta Caronni, Sara Picinoli, Irene Pezzali, Laura Ardighieri, Matilde Monti, Francesco Missale, Marco Bergamini, and Mattia Bugatti
- Abstract
Supplementary Table from A Population of TIM4+FOLR2+ Macrophages Localized in Tertiary Lymphoid Structures Correlates to an Active Immune Infiltrate Across Several Cancer Types
- Published
- 2023
4. Supplementary Data from The Atypical Receptor CCRL2 Is Essential for Lung Cancer Immune Surveillance
- Author
-
Silvano Sozzani, Alberto Mantovani, Barbara Bottazzi, Annunciata Vecchi, Federica Benvenuti, Giovanni Bernardini, Valentina Salvi, Mattia Bugatti, Stefano Calza, William Vermi, Andrea Ponzetta, Tiziana Schioppa, Francesca Sozio, and Annalisa Del Prete
- Abstract
Supplementary Figures
- Published
- 2023
5. Data from The Atypical Receptor CCRL2 Is Essential for Lung Cancer Immune Surveillance
- Author
-
Silvano Sozzani, Alberto Mantovani, Barbara Bottazzi, Annunciata Vecchi, Federica Benvenuti, Giovanni Bernardini, Valentina Salvi, Mattia Bugatti, Stefano Calza, William Vermi, Andrea Ponzetta, Tiziana Schioppa, Francesca Sozio, and Annalisa Del Prete
- Abstract
CCRL2 is a nonsignaling seven-transmembrane domain receptor. CCRL2 binds chemerin, a protein that promotes chemotaxis of leukocytes, including macrophages and natural killer (NK) cells. In addition, CCRL2 controls the inflammatory response in different pathologic settings, such as hypersensitivity, inflammatory arthritis, and experimental autoimmune encephalitis. Here, we investigated the role of CCRL2 in the regulation of lung cancer–related inflammation. The genetic deletion of Ccrl2 promoted tumor progression in urethane-induced and in KrasG12D/+/p53LoxP lung tumor mouse models. Similarly, a Kras-mutant lung tumor displayed enhanced growth in Ccrl2-deficient mice. This phenotype was associated with a reduced inflammatory infiltrate characterized by the impaired recruitment of several leukocyte populations including NK cells. Bone marrow chimeras showed that CCRL2 expression by the nonhematopoietic cell compartment was responsible for the increased tumor formation observed in Kras-mutant Ccrl2-deficient mice. In human and mouse lungs, CCRL2 was expressed by a fraction of CD31+ endothelial cells, where it could control NK infiltration. Elevated CCRL2 expression in biopsies from human lung adenocarcinoma positively correlated with clinical outcome. These results provide evidence for a crucial role of CCRL2 in shaping an anti–lung tumor immune response.
- Published
- 2023
6. Supplementary Figure from A Population of TIM4+FOLR2+ Macrophages Localized in Tertiary Lymphoid Structures Correlates to an Active Immune Infiltrate Across Several Cancer Types
- Author
-
William Vermi, Federica Benvenuti, Julie Helft, Yoann Missolo-Koussou, Nicoletta Caronni, Sara Picinoli, Irene Pezzali, Laura Ardighieri, Matilde Monti, Francesco Missale, Marco Bergamini, and Mattia Bugatti
- Abstract
Supplementary Figure from A Population of TIM4+FOLR2+ Macrophages Localized in Tertiary Lymphoid Structures Correlates to an Active Immune Infiltrate Across Several Cancer Types
- Published
- 2023
7. Supplementary Data from A Population of TIM4+FOLR2+ Macrophages Localized in Tertiary Lymphoid Structures Correlates to an Active Immune Infiltrate Across Several Cancer Types
- Author
-
William Vermi, Federica Benvenuti, Julie Helft, Yoann Missolo-Koussou, Nicoletta Caronni, Sara Picinoli, Irene Pezzali, Laura Ardighieri, Matilde Monti, Francesco Missale, Marco Bergamini, and Mattia Bugatti
- Abstract
Supplementary Data from A Population of TIM4+FOLR2+ Macrophages Localized in Tertiary Lymphoid Structures Correlates to an Active Immune Infiltrate Across Several Cancer Types
- Published
- 2023
8. Supplementary Figures from Collapse of the Plasmacytoid Dendritic Cell Compartment in Advanced Cutaneous Melanomas by Components of the Tumor Cell Secretome
- Author
-
William Vermi, Fabio Facchetti, Ausilia Manganoni, Paolo Bergese, Aldo Scarpa, Rosanna Verardi, Camillo Almici, Michele Simbolo, Camillo Farisoglio, Ester Fonsatti, Michele Maio, Mattia Bugatti, Giulio Rossi, Mariella Chiudinelli, Stefano Calza, Laura Melocchi, Luisa Benerini, Francesca Consoli, Lucia Paolini, Daniele Moratto, Matilde Monti, and Raffaella Vescovi
- Abstract
Supplementary Figures
- Published
- 2023
9. Data from Collapse of the Plasmacytoid Dendritic Cell Compartment in Advanced Cutaneous Melanomas by Components of the Tumor Cell Secretome
- Author
-
William Vermi, Fabio Facchetti, Ausilia Manganoni, Paolo Bergese, Aldo Scarpa, Rosanna Verardi, Camillo Almici, Michele Simbolo, Camillo Farisoglio, Ester Fonsatti, Michele Maio, Mattia Bugatti, Giulio Rossi, Mariella Chiudinelli, Stefano Calza, Laura Melocchi, Luisa Benerini, Francesca Consoli, Lucia Paolini, Daniele Moratto, Matilde Monti, and Raffaella Vescovi
- Abstract
Melanoma is an immunogenic neoplasm infiltrated by T cells, although these adaptive T cells usually fail to eradicate the tumor. Plasmacytoid dendritic cells (PDCs) are potent regulators of the adaptive immune response and can eliminate melanoma cells via TLR-mediated effector functions. The PDC compartment is maintained by progressively restricted bone marrow progenitors. Terminally differentiated PDCs exit the bone marrow into the circulation, then home to lymph nodes and inflamed peripheral tissues. Infiltration by PDCs is documented in various cancers. However, their role within the melanoma immune contexture is not completely known. We found that in locoregional primary cutaneous melanoma (PCM), PDC infiltration was heterogeneous, occurred early, and was recurrently localized at the invasive margin, the site where PDCs interact with CD8+ T cells. A reduced PDC density was coupled with an increased Breslow thickness and somatic mutations at the NRAS p.Q61 codon. Compared with what was seen in PCM, high numbers of PDCs were found in regional lymph nodes, as also identified by in silico analysis. In contrast, in metastatic melanoma patients, PDCs were mostly absent in the tumor tissues and were significantly reduced in the circulation, particularly in the advanced M1c group. Exposure of circulating PDCs to melanoma cell supernatant (SN-mel) depleted of extracellular vesicles resulted in significant PDC death. SN-mel exposure also resulted in a defect of PDC differentiation from CD34+ progenitors. These findings indicate that soluble components released by melanoma cells support the collapse of the PDC compartment, with clinical implications for refining TLR agonist–based trials.
- Published
- 2023
10. Supplementary Tables from Collapse of the Plasmacytoid Dendritic Cell Compartment in Advanced Cutaneous Melanomas by Components of the Tumor Cell Secretome
- Author
-
William Vermi, Fabio Facchetti, Ausilia Manganoni, Paolo Bergese, Aldo Scarpa, Rosanna Verardi, Camillo Almici, Michele Simbolo, Camillo Farisoglio, Ester Fonsatti, Michele Maio, Mattia Bugatti, Giulio Rossi, Mariella Chiudinelli, Stefano Calza, Laura Melocchi, Luisa Benerini, Francesca Consoli, Lucia Paolini, Daniele Moratto, Matilde Monti, and Raffaella Vescovi
- Abstract
Supplementary tables
- Published
- 2023
11. Data from slan+ Monocytes and Macrophages Mediate CD20-Dependent B-cell Lymphoma Elimination via ADCC and ADCP
- Author
-
Marco A. Cassatella, Silvia Lonardi, Luisa Lorenzi, Fabio Facchetti, Alberto Zamò, Giuseppe Todeschini, Lara Furlani, Giuseppe Rossi, Alessandra Tucci, Piera Balzarini, Stefano Pileri, Claudio Agostinelli, Stefano Calza, Mattia Bugatti, Sara Costa, Federica Calzetti, Cristina Tecchio, Giulia Finotti, Alessandra Micheletti, and William Vermi
- Abstract
Terminal tissue differentiation and function of slan+ monocytes in cancer is largely unexplored. Our recent studies demonstrated that slan+ monocytes differentiate into a distinct subset of dendritic cells (DC) in human tonsils and that slan+ cells colonize metastatic carcinoma-draining lymph nodes. Herein, we report by retrospective analysis of multi-institutional cohorts that slan+ cells infiltrate various types of non-Hodgkin lymphomas (NHL), particularly the diffuse large B-cell lymphoma (DLBCL) group, including the most aggressive, nodal and extranodal, forms. Nodal slan+ cells displayed features of either immature DC or macrophages, in the latter case ingesting tumor cells and apoptotic bodies. We also found in patients with DLBCL that peripheral blood slan+ monocytes, but not CD14+ monocytes, increased in number and displayed highly efficient rituximab-mediated antibody-dependent cellular cytotoxicity, almost equivalent to that exerted by NK cells. Notably, slan+ monocytes cultured in conditioned medium from nodal DLBCL (DCM) acquired a macrophage-like phenotype, retained CD16 expression, and became very efficient in rituximab-mediated antibody-dependent cellular phagocytosis (ADCP). Macrophages derived from DCM-treated CD14+ monocytes performed very efficient rituximab-mediated ADCP, however, using different FcγRs from those used by slan+ macrophages. Our observations shed new light on the complexity of the immune microenvironment of DLBCL and demonstrate plasticity of slan+ monocytes homing to cancer tissues. Altogether, data identify slan+ monocytes and macrophages as prominent effectors of antibody-mediated tumor cell targeting in patients with DLBCL.Significance: slan+ monocytes differentiate into macrophages that function as prominent effectors of antibody-mediated tumor cell targeting in lymphoma.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/13/3544/F1.large.jpg. Cancer Res; 78(13); 3544–59. ©2018 AACR.
- Published
- 2023
12. Supplementary Figures from slan+ Monocytes and Macrophages Mediate CD20-Dependent B-cell Lymphoma Elimination via ADCC and ADCP
- Author
-
Marco A. Cassatella, Silvia Lonardi, Luisa Lorenzi, Fabio Facchetti, Alberto Zamò, Giuseppe Todeschini, Lara Furlani, Giuseppe Rossi, Alessandra Tucci, Piera Balzarini, Stefano Pileri, Claudio Agostinelli, Stefano Calza, Mattia Bugatti, Sara Costa, Federica Calzetti, Cristina Tecchio, Giulia Finotti, Alessandra Micheletti, and William Vermi
- Abstract
The file contains 8 supplementary Figures displaying results in support of the data described in the results section.
- Published
- 2023
13. Figures S1-S13 from NK-cell Editing Mediates Epithelial-to-Mesenchymal Transition via Phenotypic and Proteomic Changes in Melanoma Cell Lines
- Author
-
Massimo Vitale, Segundo González, Lorenzo Moretta, William Vermi, Maria Cristina Mingari, Marcella Marconi, Enrico Munari, Mattia Bugatti, Gabriella Pietra, Alejandro López-Soto, Mirna Balsamo, Enrique J. DeAndrés-Galiana, Andrea Petretto, Juan L. Fernández-Martínez, Chiara Lavarello, Claudia Cantoni, Monica Parodi, and Leticía Huergo-Zapico
- Abstract
Fig S1-S3: Effect of NK cells on melanoma cell lines. Fig S4: Outline of transwell experiments. Fig S5: Effects of blocking mAbs on NK-induced EMT. Fig S6-S7: effects of EMT on NK-receptor ligand expression. Fig S8: effects of NK cells on melanoma cell susceptibility to drugs or g-radiation. Fig S9: Supplemental proteomic data. Fig S10,12: IHC. Fig S11: Effect of NK cells on autophagy. Fig S13: Effect of NK cells on two epithelial cell lines
- Published
- 2023
14. Table S3 from NK-cell Editing Mediates Epithelial-to-Mesenchymal Transition via Phenotypic and Proteomic Changes in Melanoma Cell Lines
- Author
-
Massimo Vitale, Segundo González, Lorenzo Moretta, William Vermi, Maria Cristina Mingari, Marcella Marconi, Enrico Munari, Mattia Bugatti, Gabriella Pietra, Alejandro López-Soto, Mirna Balsamo, Enrique J. DeAndrés-Galiana, Andrea Petretto, Juan L. Fernández-Martínez, Chiara Lavarello, Claudia Cantoni, Monica Parodi, and Leticía Huergo-Zapico
- Abstract
Supplementary Table S3
- Published
- 2023
15. Table S2 from NK-cell Editing Mediates Epithelial-to-Mesenchymal Transition via Phenotypic and Proteomic Changes in Melanoma Cell Lines
- Author
-
Massimo Vitale, Segundo González, Lorenzo Moretta, William Vermi, Maria Cristina Mingari, Marcella Marconi, Enrico Munari, Mattia Bugatti, Gabriella Pietra, Alejandro López-Soto, Mirna Balsamo, Enrique J. DeAndrés-Galiana, Andrea Petretto, Juan L. Fernández-Martínez, Chiara Lavarello, Claudia Cantoni, Monica Parodi, and Leticía Huergo-Zapico
- Abstract
Table S2
- Published
- 2023
16. Table S1 from NK-cell Editing Mediates Epithelial-to-Mesenchymal Transition via Phenotypic and Proteomic Changes in Melanoma Cell Lines
- Author
-
Massimo Vitale, Segundo González, Lorenzo Moretta, William Vermi, Maria Cristina Mingari, Marcella Marconi, Enrico Munari, Mattia Bugatti, Gabriella Pietra, Alejandro López-Soto, Mirna Balsamo, Enrique J. DeAndrés-Galiana, Andrea Petretto, Juan L. Fernández-Martínez, Chiara Lavarello, Claudia Cantoni, Monica Parodi, and Leticía Huergo-Zapico
- Abstract
Table S1
- Published
- 2023
17. Supplementary Tables from slan+ Monocytes and Macrophages Mediate CD20-Dependent B-cell Lymphoma Elimination via ADCC and ADCP
- Author
-
Marco A. Cassatella, Silvia Lonardi, Luisa Lorenzi, Fabio Facchetti, Alberto Zamò, Giuseppe Todeschini, Lara Furlani, Giuseppe Rossi, Alessandra Tucci, Piera Balzarini, Stefano Pileri, Claudio Agostinelli, Stefano Calza, Mattia Bugatti, Sara Costa, Federica Calzetti, Cristina Tecchio, Giulia Finotti, Alessandra Micheletti, and William Vermi
- Abstract
10 Tables displaying the clinical features of patient cohorts analyzed in this study, as well as correlation analyses among the clinical features and slan+ cell content.
- Published
- 2023
18. SI Materials and Methods from NK-cell Editing Mediates Epithelial-to-Mesenchymal Transition via Phenotypic and Proteomic Changes in Melanoma Cell Lines
- Author
-
Massimo Vitale, Segundo González, Lorenzo Moretta, William Vermi, Maria Cristina Mingari, Marcella Marconi, Enrico Munari, Mattia Bugatti, Gabriella Pietra, Alejandro López-Soto, Mirna Balsamo, Enrique J. DeAndrés-Galiana, Andrea Petretto, Juan L. Fernández-Martínez, Chiara Lavarello, Claudia Cantoni, Monica Parodi, and Leticía Huergo-Zapico
- Abstract
Supplementary Materials and Methods
- Published
- 2023
19. The prometastatic relevance of tumor‐infiltrating B lymphocytes in laryngeal squamous cell carcinoma
- Author
-
Francesco Missale, Mattia Bugatti, Filippo Marchi, Giulio E Mandelli, Maria Bruni, Giulia Palmerini, Matilde Monti, Anna M Bozzola, Giorgio Arena, Luca Guastini, Maurizio Boggio, Giampiero Parrinello, Giorgio Peretti, and William Vermi
- Subjects
Immunology ,Immunology and Allergy ,General Nursing - Published
- 2023
20. Adalimumab for interleukin‐1β‐mediated chronic non‐scarring scalp folliculitis: Case report and literature review
- Author
-
William Vermi, Marina Venturini, Piergiacomo Calzavara-Pinton, Mattia Bugatti, Simone Soglia, and Vincenzo Maione
- Subjects
medicine.medical_specialty ,medicine.drug_class ,Interleukin-1beta ,Antibiotics ,Folliculitis ,Inflammation ,Dermatology ,Disease ,neutrophils ,Adalimumab ,Humans ,Medicine ,Scalp folliculitis ,Scalp ,integumentary system ,business.industry ,hair ,General Medicine ,medicine.disease ,body regions ,Interleukin 1β ,Scalp Dermatoses ,inflammation ,medicine.symptom ,business ,adalimumab ,folliculitis ,medicine.drug - Abstract
Chronic non-scarring scalp folliculitis is a little-known entity included within the spectrum of scalp folliculitis, a group of diseases sharing clinical features but with heterogeneity in terms of residual scarring (always absent in chronic non-scarring scalp folliculitis), microbiology, and response to antibiotics. Chronic non-scarring scalp folliculitis is most likely an inflammatory disease within the group of neutrophilic dermatoses. The recognition of the inflammatory nature of this disease may pave the way for the use of new therapies, directly targeting pathogenic molecules. Herein, we report the first case of chronic non-scarring scalp folliculitis treated by adalimumab.
- Published
- 2021
21. The modulation of iron metabolism affects the Rhabdomyosarcoma tumor growth in vitro and in vivo
- Author
-
Michela Asperti, Luca Cantamessa, Magdalena Gryzik, Mattia Bugatti, Silvia Codenotti, Andrea Denardo, William Vermi, Alessandro Fanzani, and Maura Poli
- Subjects
General Medicine ,General Biochemistry, Genetics and Molecular Biology - Abstract
Rhabdomyosarcoma (RMS) is an aggressive rare neoplasm that derives from mesenchymal cells, which frequently develops resistance to the current therapies and the formation of metastases. Thus, new therapies are needed. The alteration of iron metabolism in cancer cells was effective in reducing the progression of many tumors but not yet investigated in RMS. Here we investigated the effect of iron modulation in RMS both in vitro and in vivo. We first characterized the most used RMS cell lines representing the most common subtypes, embryonal (ERMS, RD cells) and alveolar (ARMS, RH30 cells), for their iron metabolism, in basal condition and in response to its modulation. Then we investigated the effects of both iron overload and chelation strategies in vitro and in vivo. RMS cell lines expressed iron-related proteins, even if at lower levels compared to hepatic cell lines and they are correctly modulated in response to iron increase and deprivation. Interestingly, the treatment with different doses of ferric ammonium citrate (FAC, as iron source) and with deferiprone (DFP, as iron chelator), significantly affected the cell viability of RD and RH30. Moreover, iron supplementation (in the form of iron dextran) or iron chelation (in the form of DFP) were also effective in vivo in inhibiting the tumor mass growth both derived from RD and RH30 with iron chelation treatment the most effective one. All the data suggest that the iron modulation could be a promising approach to overcome the RMS tumor growth. The mechanism of action seems to involve the apoptotic cell death for both iron supplementation and chelation with the concomitant induction of ferroptosis in the case of iron supplementation.
- Published
- 2022
22. Pathology of macrophage activation syndrome in humanized NSGS mice
- Author
-
Zev A. Binder, Mattia Bugatti, James C. Tarrant, Charles-Antoine Assenmacher, Gwenn Danet-Desnoyers, Enrico Radaelli, Natalie Hoepp, Xiaochuan Shan, Brona N Ranieri, Joost van den Oord, Donald M. O'Rourke, and William Vermi
- Subjects
Male ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Pathology ,medicine.medical_specialty ,Chimeric mouse ,Myeloid ,040301 veterinary sciences ,Transgene ,CD34 ,Antigens, CD34 ,Spleen ,DNA-Activated Protein Kinase ,Mice, SCID ,NSGS ,Humanized model ,Inflammasome ,0403 veterinary science ,Mice ,03 medical and health sciences ,Mice, Inbred NOD ,NSG-SGM3 ,medicine ,Animals ,Humans ,HLH ,Macrophage activation syndrome ,PDX model ,030304 developmental biology ,Stem Cell Factor ,0303 health sciences ,General Veterinary ,business.industry ,Macrophages ,Hematopoietic Stem Cell Transplantation ,Granulocyte-Macrophage Colony-Stimulating Factor ,04 agricultural and veterinary sciences ,Hematopoietic Stem Cells ,medicine.disease ,Recombinant Proteins ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Bone marrow ,business ,Histiocytosis ,Interleukin Receptor Common gamma Subunit - Abstract
“Humanized” immunodeficient mice generated via the transplantation of CD34+ human hematopoietic stem cells (hHSC) are an important preclinical model system. The triple transgenic NOD.Cg-PrkdcscidIl2rgtm1Wjl Tg(CMV-IL3,CSF2,KITLG)1Eav/MloySzJ (NSGS) mouse line is increasingly used as recipient for CD34+ hHSC engraftment. NSGS mice combine the features of the highly immunodeficient NSG mice with transgenic expression of the human myeloid stimulatory cytokines GM-CSF, IL-3, and Kit ligand. While generating humanized NSGS (huNSGS) mice from two independent cohorts, we encountered a fatal macrophage activation syndrome (MAS)-like phenotype resulting from the transplantation of CD34+ hHSC. huNSGS mice exhibiting this phenotype declined clinically starting at approximately 10 weeks following CD34+ hHSC engraftment, with all mice requiring euthanasia by 16 weeks. Gross changes comprised small, irregular liver, splenomegaly, cardiomegaly, and generalized pallor. Hematological abnormalities included severe thrombocytopenia and anemia. Pathologically, huNSGS spontaneously developed a disseminated histiocytosis with infiltrates of activated macrophages and hemophagocytosis predominantly affecting the liver, spleen, bone marrow, and pancreas. The infiltrates were chimeric with a mixture of human and mouse macrophages. Immunohistochemistry suggested activation of the inflammasome in both human and murine macrophages. Active Epstein-Barr virus infection was not a feature. Although the affected mice exhibited robust chimerism of the spleen and bone marrow, the phenotype often developed in the face of low chimerism of the peripheral blood. Given the high penetrance and early lethality associated with the MAS-like phenotype here described, we urge caution when considering the use of huNSGS mice for the development of long-term studies.
- Published
- 2021
23. Immunohistochemical Detection of SARS-CoV-2 Antigens by Single and Multiple Immunohistochemistry
- Author
-
Silvia, Lonardi, Mattia, Bugatti, Arianna, Valzelli, and Fabio, Facchetti
- Subjects
Epitopes ,COVID-19 Testing ,SARS-CoV-2 ,COVID-19 ,Humans ,Immunohistochemistry - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can be demonstrated in tissue sections by immunohistochemistry (IHC), which has the power to localize in bright field specific antigens in cells and tissues. The use of double or triple immunostains is capable of highlighting which cells are infected and/or the relationship of infected cell with other cells and tissue structures. In addition, immunoenzymatic multi-staining permits the simultaneous identification, localization, and enumeration of different cellular epitopes. Moreover, this method improves analytical precision, decreasing the time required for morphometric quantification, maximizing the information obtained from a single slide of paraffin-embedded tissue.
- Published
- 2022
24. Integrated Biomarker Analysis Reveals L1CAM as a Potential Stratification Marker for No Specific Molecular Profile High-Risk Endometrial Carcinoma
- Author
-
Antonella Ravaggi, Davide Capoferri, Laura Ardighieri, Iacopo Ghini, Federico Ferrari, Chiara Romani, Mattia Bugatti, Laura Zanotti, Stephanie Vrede, Germana Tognon, Johanna M. A. Pijnenborg, Enrico Sartori, Stefano Calza, Eliana Bignotti, and Franco Odicino
- Subjects
Cancer Research ,molecular classification ,Oncology ,L1CAM ,NSMP ,prognosis ,high-risk endometrial carcinoma ,platinum-based adjuvant chemotherapy ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] - Abstract
Contains fulltext : 287744.pdf (Publisher’s version ) (Open Access) Histopathologic assessment of high-risk endometrial cancer (EC) suffers from intersubject variability and poor reproducibility. The pragmatic classification in four molecular subgroups helps to overcome these limits, showing a significant prognostic value. The "no specific molecular profile" (NSMP) is the most heterogeneous EC subgroup, requiring further characterization to better guide its clinical management. DNA sequencing of POLE exonuclease domain and immunohistochemistry for PMS2, MSH6, and p53 were performed in order to stratify a cohort of 94 high-risk EC patients in the four molecular subgroups. Moreover, a panel of seven additional biomarkers was tested. Patients were found to be 16% POLE-mutated, 36% mismatch repair-deficient, 27% p53-abnormal, and 21% NSMP. In the multivariable model, molecular groups confirmed their significant association with disease-specific survival and progression-free survival, with p53-abnormal and NSMP endometrial cancer characterized by poor outcomes. Among the additional evaluated biomarkers, L1CAM was the only one with a significant prognostic value within the NSMP subgroup. NSMP/L1CAM-positive patients experienced the worst outcome and were "early-relapsing" after platinum-based chemotherapy, with a significantly shorter platinum-free interval compared to L1CAM-negative patients. L1CAM appears to be a promising candidate as a prognostic and predictive biomarker in the high-risk NSMP subgroup, which is actually known to lack specific molecular markers.
- Published
- 2022
25. L1CAM expression as a predictor of platinum response in high-risk endometrial carcinoma
- Author
-
Chiara Romani, Davide Capoferri, Casper Reijnen, Silvia Lonardi, Antonella Ravaggi, Martina Ratti, Mattia Bugatti, Laura Zanotti, Germana Tognon, Enrico Sartori, Franco Odicino, Stefano Calza, Johanna M. A. Pijnenborg, and Eliana Bignotti
- Subjects
Cancer Research ,precision medicine ,Neural Cell Adhesion Molecule L1 ,Prognosis ,Carboplatin ,Endometrial Neoplasms ,Women's cancers Radboud Institute for Health Sciences [Radboudumc 17] ,high-risk endometrial cancer ,Oncology ,L1CAM ,platinum-based adjuvant treatment ,Biomarkers, Tumor ,Humans ,Female ,Carcinoma, Endometrioid ,Neoplasm Staging ,Platinum - Abstract
Contains fulltext : 251438.pdf (Publisher’s version ) (Open Access) For high-risk endometrial cancer (EC) patients, adjuvant chemotherapy is recommended to improve outcome. Yet, predictive biomarkers for response to platinum-based chemotherapy (Pt-aCT) are currently lacking. We tested expression of L1 cell-adhesion molecule (L1CAM), a well-recognised marker of poor prognosis in EC, in tumour samples from high-risk EC patients, to explore its role as a predictive marker of Pt-aCT response. L1CAM expression was determined using RT-qPCR and immunohistochemistry in a cohort of high-risk EC patients treated with Pt-aCT and validated in a multicentric independent cohort. The association between L1CAM and clinicopathologic features and L1CAM additive value in predicting platinum response were determined. The effect of L1CAM gene silencing on response to carboplatin was functionally tested on primary L1CAM-expressing cells. Increased L1CAM expression at both genetic and protein level correlated with high-grade, non-endometrioid histology and poor response to platinum treatment. A predictive model adding L1CAM to prognostic clinical variables significantly improved platinum response prediction (C-index 78.1%, P = .012). In multivariate survival analysis, L1CAM expression was significantly associated with poor outcome (HR: 2.03, P = .019), potentially through an indirect effect, mediated by its influence on response to chemotherapy. In vitro, inhibition of L1CAM significantly increased cell sensitivity to carboplatin, supporting a mechanistic link between L1CAM expression and response to platinum in EC cells. In conclusion, we have demonstrated the role of L1CAM in the prediction of response to Pt-aCT in two independent cohorts of high-risk EC patients. L1CAM is a promising candidate biomarker to optimise decision making in high-risk patients who are eligible for Pt-aCT.
- Published
- 2022
26. Self-Renewal of Macrophages: Tumor-Released Factors and Signaling Pathways
- Author
-
Serena Filiberti, Mariapia Russo, Silvia Lonardi, Mattia Bugatti, William Vermi, Cathy Tournier, and Emanuele Giurisato
- Subjects
Medicine (miscellaneous) ,General Biochemistry, Genetics and Molecular Biology - Abstract
Macrophages are the most abundant immune cells of the tumor microenvironment (TME) and have multiple important functions in cancer. During tumor growth, both tissue-resident macrophages and newly recruited monocyte-derived macrophages can give rise to tumor-associated macrophages (TAMs), which have been associated with poor prognosis in most cancers. Compelling evidence indicate that the high degree of plasticity of macrophages and their ability to self-renew majorly impact tumor progression and resistance to therapy. In addition, the microenvironmental factors largely affect the metabolism of macrophages and may have a major influence on TAMs proliferation and subsets functions. Thus, understanding the signaling pathways regulating TAMs self-renewal capacity may help to identify promising targets for the development of novel anticancer agents. In this review, we focus on the environmental factors that promote the capacity of macrophages to self-renew and the molecular mechanisms that govern TAMs proliferation. We also highlight the impact of tumor-derived factors on macrophages metabolism and how distinct metabolic pathways affect macrophage self-renewal.
- Published
- 2022
27. A ligand-insensitive UNC5B splicing isoform regulates angiogenesis by promoting apoptosis
- Author
-
Alex Pezzotta, Anna Pistocchi, Patrick Mehlen, Andrea Paradisi, Anna Di Matteo, Federico Forneris, Daniele Campolungo, Costanza Giampietro, Elisa Belloni, Gianluca Deflorian, Nina Volf, Antonella Chiapparino, Michael Rehman, William Vermi, Maria Paola Paronetto, Mattia Bugatti, Davide Pradella, Claudia Ghigna, Anne Eichmann, Matteo Campioni, Serena Zacchigna, Luigi Scietti, Paradisi, Andrea, Consiglio Nazionale delle Ricerche (CNR), Università degli Studi di Pavia = University of Pavia (UNIPV), IFOM, Istituto FIRC di Oncologia Molecolare (IFOM), Università degli Studi di Milano = University of Milan (UNIMI), San Raffaele Scientific Institute, Vita-Salute San Raffaele University and Center for Translational Genomics and Bioinformatics, Centre de Recherche en Cancérologie de Lyon (UNICANCER/CRCL), Centre Léon Bérard [Lyon]-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Développement Cancer et Thérapies Ciblées [Lyon] (LabEx DEVweCAN), Université de Lyon, Centre Léon Bérard [Lyon], University of Brescia, Washington University School of Medicine in St. Louis, Washington University in Saint Louis (WUSTL), Heidelberg University, Swiss Federal Laboratories for Materials Science and Technology [Dübendorf] (EMPA), International Centre for Genetic Engineering and Biotechnology (ICGEB) (Trieste), Yale School of Medicine [New Haven, Connecticut] (YSM), Università degli studi di Trieste = University of Trieste, IRCCS Ospedale Pediatrico Bambino Gesù [Roma], University of Rome 'Foro Italico', Paris-Centre de Recherche Cardiovasculaire (PARCC (UMR_S 970/ U970)), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Cité (UPCité), Pradella, D., Deflorian, G., Pezzotta, A., Di Matteo, A., Belloni, E., Campolungo, D., Paradisi, A., Bugatti, M., Vermi, W., Campioni, M., Chiapparino, A., Scietti, L., Forneris, F., Giampietro, C., Volf, N., Rehman, M., Zacchigna, S., Paronetto, M. P., Pistocchi, A., Eichmann, A., Mehlen, P., Ghigna, C., University of Pavia, University of Milan, Yale University School of Medicine, University of Trieste, and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP)
- Subjects
Netrin Receptor ,Angiogenesis ,Regulator ,General Physics and Astronomy ,Apoptosis ,RNA-binding protein ,RNA-Binding Protein ,0302 clinical medicine ,Netrin ,Morphogenesis ,RNA Isoforms ,MESH: Animals ,MESH: Endothelial Cells ,MESH: Nerve Tissue Proteins ,Zebrafish ,Colonic Neoplasm ,Endothelial Cell ,0303 health sciences ,Multidisciplinary ,Neovascularization, Pathologic ,Chemistry ,MESH: Alternative Splicing ,RNA-Binding Proteins ,RNA Isoform ,Netrin-1 ,Cell biology ,MESH: Survival Analysis ,030220 oncology & carcinogenesis ,embryonic structures ,Colonic Neoplasms ,RNA splicing ,Survival Analysi ,Netrin Receptors ,Human ,Gene isoform ,animal structures ,Blood Vessel ,Science ,Nerve Tissue Proteins ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Neuro-Oncological Ventral Antigen ,Morphogenesi ,Animals ,Humans ,Alternative Splicing ,Blood Vessels ,Endothelial Cells ,Survival Analysis ,MESH: Zebrafish ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Neovascularization ,030304 developmental biology ,Pathologic ,MESH: Colonic Neoplasms ,MESH: Humans ,Animal ,MESH: Apoptosis ,fungi ,Alternative splicing ,MESH: Blood Vessels ,General Chemistry ,MESH: Netrin Receptors ,MESH: RNA Isoforms ,MESH: Morphogenesis ,Exon skipping ,MESH: RNA-Binding Proteins ,nervous system ,MESH: Netrin-1 ,Nerve Tissue Protein ,MESH: Neovascularization, Pathologic ,Tumour angiogenesis - Abstract
The Netrin-1 receptor UNC5B is an axon guidance regulator that is also expressed in endothelial cells (ECs), where it finely controls developmental and tumor angiogenesis. In the absence of Netrin-1, UNC5B induces apoptosis that is blocked upon Netrin-1 binding. Here, we identify an UNC5B splicing isoform (called UNC5B-Δ8) expressed exclusively by ECs and generated through exon skipping by NOVA2, an alternative splicing factor regulating vascular development. We show that UNC5B-Δ8 is a constitutively pro-apoptotic splicing isoform insensitive to Netrin-1 and required for specific blood vessel development in an apoptosis-dependent manner. Like NOVA2, UNC5B-Δ8 is aberrantly expressed in colon cancer vasculature where its expression correlates with tumor angiogenesis and poor patient outcome. Collectively, our data identify a mechanism controlling UNC5B’s necessary apoptotic function in ECs and suggest that the NOVA2/UNC5B circuit represents a post-transcriptional pathway regulating angiogenesis., Nature Communications, 12, ISSN:2041-1723
- Published
- 2021
28. FXYD5 (Dysadherin) upregulation predicts shorter survival and reveals platinum resistance in high-grade serous ovarian cancer patients
- Author
-
Mattia Bugatti, Eliana Bignotti, Angela Gambino, Laura Zanotti, Antonella Ravaggi, Chiara Romualdi, Chiara Romani, Enrico Sartori, Renata A. Tassi, Germana Tognon, Fulvio Borella, Paola Todeschini, Laura Ardighieri, Franco Odicino, and Dionyssios Katsaros
- Subjects
Oncology ,Cancer Research ,Microarray ,Drug Resistance ,Ion Channels ,Carboplatin ,Transcriptome ,Prognostic markers ,0302 clinical medicine ,Ovarian carcinoma ,Receptors ,Ovarian Neoplasms ,Reverse Transcriptase Polymerase Chain Reaction ,Microfilament Proteins ,Middle Aged ,Prognosis ,Progression-Free Survival ,Neoplasm Proteins ,Up-Regulation ,Serous fluid ,030220 oncology & carcinogenesis ,Cell Surface ,Immunohistochemistry ,Female ,Aged ,Analysis of Variance ,Antineoplastic Agents ,Cystadenocarcinoma, Serous ,Humans ,Neoplasm Grading ,Receptors, Cell Surface ,Survival Analysis ,Drug Resistance, Neoplasm ,medicine.medical_specialty ,Cystadenocarcinoma ,Article ,03 medical and health sciences ,Ovarian cancer ,Internal medicine ,medicine ,Progression-free survival ,Survival analysis ,business.industry ,Proportional hazards model ,Serous ,Neoplasm ,business - Abstract
Background High-grade serous ovarian carcinoma (HGSOC) is generally associated with a very dismal prognosis. Nevertheless, patients with similar clinicopathological characteristics can have markedly different clinical outcomes. Our aim was the identification of novel molecular determinants influencing survival. Methods Gene expression profiles of extreme HGSOC survivors (training set) were obtained by microarray. Differentially expressed genes (DEGs) and enriched signalling pathways were determined. A prognostic signature was generated and validated on curatedOvarianData database through a meta-analysis approach. The best prognostic biomarker from the signature was confirmed by RT-qPCR and by immunohistochemistry on an independent validation set. Cox regression model was chosen for survival analysis. Results Eighty DEGs and the extracellular matrix-receptor (ECM-receptor) interaction pathway were associated to extreme survival. A 10-gene prognostic signature able to correctly classify patients with 98% of accuracy was identified. By an ‘in-silico’ meta-analysis, overexpression of FXYD domain-containing ion transport regulator 5 (FXYD5), also known as dysadherin, was confirmed in HGSOC short-term survivors compared to long-term ones. Its prognostic and predictive power was then successfully validated, both at mRNA and protein level, first on training than on validation sample set. Conclusion We demonstrated the possible involvement of FXYD5 and ECM-receptor interaction signal pathway in HCSOC survival and prognosis.
- Published
- 2019
29. Collapse of the Plasmacytoid Dendritic Cell Compartment in Advanced Cutaneous Melanomas by Components of the Tumor Cell Secretome
- Author
-
Mattia Bugatti, Ausilia Maria Manganoni, Matilde Monti, Camillo Almici, Aldo Scarpa, Ester Fonsatti, Francesca Consoli, Michele Maio, Luisa Benerini, Giulio Rossi, Daniele Moratto, Fabio Facchetti, Paolo Bergese, Rosanna Verardi, Raffaella Vescovi, Stefano Calza, William Vermi, Camillo Farisoglio, Michele Simbolo, Mariella Chiudinelli, Lucia Paolini, and Laura Melocchi
- Subjects
Adult ,Male ,Proto-Oncogene Proteins B-raf ,EXPRESSION ,RECRUITMENT ,0301 basic medicine ,Cancer Research ,Skin Neoplasms ,Immunology ,CD34 ,MICROENVIRONMENT ,Plasmacytoid dendritic cell ,Biology ,EARLY MARKER ,Proto-Oncogene Proteins p21(ras) ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Humans ,Melanoma ,Aged ,LACTATE-DEHYDROGENASE ,Aged, 80 and over ,LACTIC-ACID ,CUTTING EDGE ,LYMPH-NODES ,hemic and immune systems ,Dendritic Cells ,Middle Aged ,Acquired immune system ,medicine.disease ,PD-1 BLOCKADE ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Disease Progression ,Cancer research ,Female ,Bone marrow ,Chemokines ,Sentinel Lymph Node ,RESPONSES ,CD8 - Abstract
Melanoma is an immunogenic neoplasm infiltrated by T cells, although these adaptive T cells usually fail to eradicate the tumor. Plasmacytoid dendritic cells (PDCs) are potent regulators of the adaptive immune response and can eliminate melanoma cells via TLR-mediated effector functions. The PDC compartment is maintained by progressively restricted bone marrow progenitors. Terminally differentiated PDCs exit the bone marrow into the circulation, then home to lymph nodes and inflamed peripheral tissues. Infiltration by PDCs is documented in various cancers. However, their role within the melanoma immune contexture is not completely known. We found that in locoregional primary cutaneous melanoma (PCM), PDC infiltration was heterogeneous, occurred early, and was recurrently localized at the invasive margin, the site where PDCs interact with CD8+ T cells. A reduced PDC density was coupled with an increased Breslow thickness and somatic mutations at the NRAS p.Q61 codon. Compared with what was seen in PCM, high numbers of PDCs were found in regional lymph nodes, as also identified by in silico analysis. In contrast, in metastatic melanoma patients, PDCs were mostly absent in the tumor tissues and were significantly reduced in the circulation, particularly in the advanced M1c group. Exposure of circulating PDCs to melanoma cell supernatant (SN-mel) depleted of extracellular vesicles resulted in significant PDC death. SN-mel exposure also resulted in a defect of PDC differentiation from CD34+ progenitors. These findings indicate that soluble components released by melanoma cells support the collapse of the PDC compartment, with clinical implications for refining TLR agonist–based trials.
- Published
- 2019
30. Tissue-resident FOLR2+ macrophages associate with tumor-infiltrating CD8+ T cells and with increased survival of breast cancer patients
- Author
-
Christian P. Bromley, Eliane Piaggio, William Vermi, André Nicolas, Jérôme Galon, Sylvain Baulande, Christine Sedlik, Charles-Antoine Dutertre, Kotsias F, Fabien Reyal, Wilfrid Richer, Nicolás Gonzalo Núñez, Pierre Guermonprez, Gerber-Ferder Y, Julie Helft, Anne Vincent-Salomon, Laetitia Lesage, Yoann Missolo-Koussou, Sophie Viel, Rodrigo Nalio Ramos, Santiago Zelenay, Emmanuel Donnadieu, Sonia Lameiras, Jordan Denizeau, Florent Ginhoux, Mattia Bugatti, Mylène Bohec, Jimena Tosello, Buttard B, Didier Meseure, Niborski Ll, Pamela Caudana, Lene Vimeux, and Institut Curie, PSL Research University, INSERM U932.
- Subjects
0303 health sciences ,education.field_of_study ,[SDV]Life Sciences [q-bio] ,Population ,Mammary gland ,Cancer ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,medicine.disease ,[SDV.IMM.II]Life Sciences [q-bio]/Immunology/Innate immunity ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,medicine.anatomical_structure ,[SDV.IMM.IA]Life Sciences [q-bio]/Immunology/Adaptive immunology ,Immunity ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Cytotoxic T cell ,Macrophage ,education ,CD8 ,030304 developmental biology - Abstract
SUMMARYMacrophage infiltration is a hallmark of solid cancers and overall macrophage infiltration is correlated with lower patient survival and resistance to therapy. However, tumor-associated macrophages are phenotypically and functionally heterogeneous. Specific tumor-associated macrophage subsets might be endowed with antagonistic role on cancer progression and on the development of anti-tumor immunity. For instance, monocyte-derived TREM2+ tumor-associated macrophages have pro-tumorigenic and immunosuppressive functions. Here, we identify a discrete population of FOLR2+ tumor-associated macrophages positively correlating with patient survival in breast cancer. FOLR2+ macrophages are evolutionarily conserved across species and populate human and murine healthy mammary gland. Moreover, FOLR2+ macrophages co-localize with lymphoid aggregates containing CD8+ T cells in breast cancer and across ten other types of cancers. This study highlights antagonistic roles for tumor-associated macrophage subsets and paves the way for subset-specific therapeutic interventions in macrophages-based cancer therapies.
- Published
- 2021
31. Risk Assessment in Solitary Fibrous Tumor of the Uterine Corpus: Report of a Case and Systematic Review of the Literature
- Author
-
Franco Odicino, Federico Ferrari, Mattia Bugatti, Luisa Bercich, Monica Ragnoli, Laura Ardighieri, Enrico Sartori, Andrea Palicelli, and Iacopo Ghini
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Solitary fibrous tumor ,Neoplasms, Fibrous Tissue ,Uterus ,Risk Assessment ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Medicine ,Humans ,Fibroblastic Tumor ,business.industry ,Clinical course ,Myometrium ,Middle Aged ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Uterine corpus ,030220 oncology & carcinogenesis ,Solitary Fibrous Tumors ,Surgery ,Female ,Anatomy ,business - Abstract
Solitary fibrous tumor (SFT) is an uncommon fibroblastic tumor occurring preferentially in the pleura, with a variable clinical course. SFT can arise also in numerous extrathoracic sites and very rarely in the female genital tract, with only scarce reports of uterine SFT. We reported a new uterine SFT arising in a 45-year-old woman, and we performed a systematic review of SFT cases of the uterine corpus interrogating the electronic databases PubMed, Web of Science, and Scopus. We identified only 13 patients diagnosed with SFT of the uterine corpus, including our one. Complete clinical workout at disease presentation showed no evidence of extrauterine spread in all cases, except for 1 patient who presented with metastatic disease. Tumor recurrences/metastases occurred in a minority of the patients and were poorly related to clinicopathological risk factors and patients stratification based on different scoring systems. Since the long-term clinical behavior of uterine SFT is limited and poorly predictable, extended follow-up is recommended also for all cases arising in the uterine corpus.
- Published
- 2021
32. Persistent Infection of Human Mesenchymal Stromal Cells With Bartonella Henselae Exerts a Proangiogenic Effect
- Author
-
Tiziana Musso, Valentina Salvi, Rosaria Sparti, Sara Scutera, Stefania Mitola, Silvano Sozzani, Giorgia Piersigilli, Mattia Bugatti, Tiziana Schioppa, Daniela Alotto, and Elisabetta Grillo
- Subjects
Bartonella henselae ,biology ,Mesenchymal stem cell ,Cancer research ,biology.organism_classification - Abstract
Background B henselae is in humans the aetiologic agent of cat-scratch disease and of the vasculoproliferative disorders bacillary angiomatosis and bacillary peliosis. Although endothelial cells are crucial in the pathogenesis other cell types function as reservoir and contribute to pathological angiogenesis. Among them, mesenchymal stromal cells (MSCs) can sense pathogens and mount an appropriate cytokine/chemokine response through different Pattern Recognition Receptors (PRRs). MSCs exert direct antimicrobial effector function but may also shelter bacteria such as M. tuberculosis. Methods Adipose-derived MSCs were infected with B. henselae and analyzed for bacterial persistence by gentamicin protection assay, immunohistochemistry and immunofluorescence. Involvement of PRRs in bacterial infection was evaluated through gene and protein expression analysis. The effect of infection on MSC proliferation, apoptosis and release of soluble factors was assessed. The role of infected-MSC conditioned medium in promoting Bartonella infection of endothelial cells and angiogenesis was demonstrated using respectively gentamicin protection assay and different pro-angiogenic assays including spheroid, wound healing and morphogenesis. Results B. henselae can readily infect MSCs and survive in perinuclear bound vacuoles for up to 8 days. Bartonella infection stimulates MSC proliferation and upregulation of EGFR and of the two pattern recognition receptors (PRRs) TLR2 and NOD1. Specific inhibition of EGFR reduces bacterial internalization and treatment with anti-TLR2 neutralizing antibody or EGFR/NOD1 inhibitors significantly downmodulates CXCL8 production. Secretome analysis shows that, in addition to CXCL8, infected MSCs secrete higher levels of the proangiogenic factors VEGF, FGF-7, MMP-9, PIGF, serpin E1, TSP-1, uPA, IL-6, CCL5 and PDGF-D. Importantly, supernatants from B. henselae-infected MSCs increase the susceptibility of ECs to B. henselae infection while enhancing EC proangiogenic potential. Conclusions Altogether, these findings indicate that MSCs constitute a novel niche for B. henselae, which favors the persistence of vascular proliferative disorders.
- Published
- 2020
33. Tissue-resident FOLR2+ macrophages associate with CD8+ T cell infiltration in human breast cancer
- Author
-
Rodrigo Nalio Ramos, Yoann Missolo-Koussou, Yohan Gerber-Ferder, Christian P. Bromley, Mattia Bugatti, Nicolas Gonzalo Núñez, Jimena Tosello Boari, Wilfrid Richer, Laurie Menger, Jordan Denizeau, Christine Sedlik, Pamela Caudana, Fiorella Kotsias, Leticia L. Niborski, Sophie Viel, Mylène Bohec, Sonia Lameiras, Sylvain Baulande, Laëtitia Lesage, André Nicolas, Didier Meseure, Anne Vincent-Salomon, Fabien Reyal, Charles-Antoine Dutertre, Florent Ginhoux, Lene Vimeux, Emmanuel Donnadieu, Bénédicte Buttard, Jérôme Galon, Santiago Zelenay, William Vermi, Pierre Guermonprez, Eliane Piaggio, and Julie Helft
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2022
34. Tissue-resident FOLR2
- Author
-
Rodrigo, Nalio Ramos, Yoann, Missolo-Koussou, Yohan, Gerber-Ferder, Christian P, Bromley, Mattia, Bugatti, Nicolas Gonzalo, Núñez, Jimena, Tosello Boari, Wilfrid, Richer, Laurie, Menger, Jordan, Denizeau, Christine, Sedlik, Pamela, Caudana, Fiorella, Kotsias, Leticia L, Niborski, Sophie, Viel, Mylène, Bohec, Sonia, Lameiras, Sylvain, Baulande, Laëtitia, Lesage, André, Nicolas, Didier, Meseure, Anne, Vincent-Salomon, Fabien, Reyal, Charles-Antoine, Dutertre, Florent, Ginhoux, Lene, Vimeux, Emmanuel, Donnadieu, Bénédicte, Buttard, Jérôme, Galon, Santiago, Zelenay, William, Vermi, Pierre, Guermonprez, Eliane, Piaggio, and Julie, Helft
- Subjects
Lymphocytes, Tumor-Infiltrating ,Macrophages ,Humans ,Breast Neoplasms ,Female ,Breast ,Folate Receptor 2 ,CD8-Positive T-Lymphocytes ,Prognosis - Abstract
Macrophage infiltration is a hallmark of solid cancers, and overall macrophage infiltration correlates with lower patient survival and resistance to therapy. Tumor-associated macrophages, however, are phenotypically and functionally heterogeneous. Specific subsets of tumor-associated macrophage might be endowed with distinct roles on cancer progression and antitumor immunity. Here, we identify a discrete population of FOLR2
- Published
- 2020
35. Single-cell analyses of Crohn's disease tissues reveal intestinal intraepithelial T cells heterogeneity and altered subset distributions
- Author
-
Ekaterina Esaulova, Mattia Bugatti, Scott A. Jelinsky, Marina Cella, Natalia Jaeger, Ramnik J. Xavier, Jorge Schettini, Thomas A. Wynn, Blanda Di Luccia, Ramya Gamini, Maxim N. Artyomov, Charles V. Rosadini, William Vermi, Marco Colonna, Baylee Kinnett, and Shanrong Zhao
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,T cell ,Science ,T cells ,General Physics and Astronomy ,Systems analysis ,Inflammation ,Biology ,CD8-Positive T-Lymphocytes ,T-Lymphocytes, Regulatory ,General Biochemistry, Genetics and Molecular Biology ,Article ,Inflammatory bowel disease ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Crohn Disease ,RAR-related orphan receptor gamma ,T-Lymphocyte Subsets ,medicine ,Humans ,Lymphocyte Count ,Intraepithelial Lymphocytes ,Cells, Cultured ,Lamina propria ,Multidisciplinary ,Gene Expression Profiling ,General Chemistry ,Intestinal epithelium ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Intraepithelial lymphocyte ,Th17 Cells ,Mucosal immunology ,medicine.symptom ,Single-Cell Analysis ,CD8 - Abstract
Crohn’s disease (CD) is a chronic transmural inflammation of intestinal segments caused by dysregulated interaction between microbiome and gut immune system. Here, we profile, via multiple single-cell technologies, T cells purified from the intestinal epithelium and lamina propria (LP) from terminal ileum resections of adult severe CD cases. We find that intraepithelial lymphocytes (IEL) contain several unique T cell subsets, including NKp30+γδT cells expressing RORγt and producing IL-26 upon NKp30 engagement. Further analyses comparing tissues from non-inflamed and inflamed regions of patients with CD versus healthy controls show increased activated TH17 but decreased CD8+T, γδT, TFH and Treg cells in inflamed tissues. Similar analyses of LP find increased CD8+, as well as reduced CD4+T cells with an elevated TH17 over Treg/TFH ratio. Our analyses of CD tissues thus suggest a potential link, pending additional validations, between transmural inflammation, reduced IEL γδT cells and altered spatial distribution of IEL and LP T cell subsets., Crohn’s disease results from transmural inflammation in the gut, but analyses of local immune populations are still lacking. Here, the authors show, by combining multiple single-cell approaches, that intraepithelial and lamina propria T cells are heterogenous, show unique phenotypes, and exhibit altered subsets upon inflammation.
- Published
- 2020
36. A Rare Complex BRAF Mutation Involving Codon V600 and K601 in Primary Cutaneous Melanoma: Case Report
- Author
-
Francesca Consoli, Gianluca Barbieri, Matteo Picciolini, Daniela Medicina, Mattia Bugatti, Valeria Tovazzi, Barbara Liserre, Claudia Zambelli, Fausto Zorzi, Alfredo Berruti, Emanuele Giurisato, and William Vermi
- Subjects
0301 basic medicine ,PTEN ,Cancer Research ,Case Report ,V600E2, K601I, Melanoma, BRAF mutation V600, Next-generation sequencing, PTEN ,medicine.disease_cause ,lcsh:RC254-282 ,BRAF ,V600E2 ,03 medical and health sciences ,0302 clinical medicine ,BRAF mutation V600 ,medicine ,skin and connective tissue diseases ,MEK inhibitors ,Melanoma ,neoplasms ,Gene ,Mutation ,biology ,business.industry ,Kinase ,Point mutation ,BRAF inhibitors ,metastatic melanoma ,mutation ,NGS ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Next-generation sequencing ,Cancer research ,biology.protein ,Immunohistochemistry ,K601I ,business - Abstract
BRAF is one of the most common mutated kinases detected in human cancer, particularly in cases of primary cutaneous melanomas (PCM). Mutations of the BRAF proto-oncogene, at the p.V600 codon, has been detected in more than 50% of primary and metastatic melanoma cells in clinical samples. In addition to the most frequent BRAF p.V600E mutation, corresponding to the single base pair substitution c.1799T>A, rarer mutations, within and outside the V600 codon, have been described. Expectedly, BRAF and MEK inhibitors (or their combination) have been poorly explored as potential therapeutic strategies in metastatic melanomas harboring this rare mutation. By using a set of sequencing techniques and immunohistochemistry, this work reports the genomic and clinical features of two melanoma patients showing a rare complex mutation affecting codon V600 and K601 of the BRAF gene, leading to a V600E2; K601I change. Specifically, these two patients show a distinct clinical behavior and significantly differ in their responses to BRAF and MEK inhibitors. Indeed, although this treatment has proven to be effective and safe in both cases, the observed variability between the two patients resulted as a direct consequence of the baseline extent of brain involvement, intracranial treatment failure as well as on the PTEN status.
- Published
- 2020
37. Human neutrophils activated by TLR8 agonists, with or without IFNγ, synthesize and release EBI3, but not IL-12, IL-27, IL-35, or IL-39
- Author
-
Sara Gasperini, Fabio Arruda-Silva, Mattia Bugatti, William Vermi, Marco A. Cassatella, Francisco Bianchetto-Aguilera, Nicola Tamassia, Odile Devergne, Elisa Gardiman, and Frédérique Larousserie
- Subjects
0301 basic medicine ,EBI3 ,Neutrophils ,Immunology ,Biology ,Virus ,IL-27 ,Minor Histocompatibility Antigens ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,In vivo ,TNFα ,Immunology and Allergy ,Animals ,Humans ,TLR8 ,Interleukins ,Imidazoles ,Cell Biology ,Acquired immune system ,Molecular biology ,Interleukin-12 ,030104 developmental biology ,IL-12 ,Toll-Like Receptor 8 ,030220 oncology & carcinogenesis ,IL-35 ,Interleukin 12 ,TLR4 ,Tumor necrosis factor alpha - Abstract
The IL-12 family of cytokines plays crucial functions in innate and adaptive immunity. These cytokines include heterodimers sharing distinct α (IL-12A, IL-23A, and IL-27A) with two β (IL-12B and Epstein-Barr virus induced gene 3 [EBI3]) chains, respectively, IL-12 (IL-12B plus IL-12A) and IL-23 (IL-12B plus IL-23A) sharing IL-12B, IL-27 (EBI3 plus IL-27A), IL-35 (EBI3 plus IL-12A), and IL-39 (EBI3 plus IL-23A) sharing EBI3. In this context, we have recently reported that highly pure neutrophils incubated with TLR8 agonists produce functional IL-23. Previously, we showed that neutrophils incubated with LPS plus IFNγ for 20 h produce IL-12. Herein, we investigated whether highly pure, TLR8-activated, neutrophils produce EBI3, and in turn IL-27, IL-35, and IL-39, the IL-12 members containing it. We report that neutrophils incubated with TLR8 ligands, TNFα and, to a lesser extent, LPS, produce and release remarkable amounts of EBI3, but not IL-27A, consequently excluding the possibility for an IL-27 production. We also report a series of unsuccessful experiments performed to investigate whether neutrophil-derived EBI3 associates with IL-23A to form IL-39. Furthermore, we show that neutrophils incubated with IFNγ in combination with either TLR8 or TLR4 ligands express/produce neither IL-12, nor IL-35, due to the inability of IFNγ, contrary to previous findings, to activate IL12A transcription. Even IL-27 was undetectable in supernatants harvested from IFNγ plus R848-treated neutrophils, although they were found to accumulate IL27A transcripts. Finally, by immunohistochemistry experiments, EBI3-positive neutrophils were found in discrete pathologies only, including diverticulitis, cholecystitis, Gorham disease, and Bartonella Henselae infection, implying a specific role of neutrophil-derived EBI3 in vivo.
- Published
- 2020
38. Molecular Pathology Demonstration of SARS-CoV-2 in Cytotrophoblast from Placental Tissue with Chronic Histiocytic Intervillositis, Trophoblast Necrosis and COVID-19
- Author
-
Fabio Facchetti, David A. Schwartz, Mattia Bugatti, and Amerigo Santoro
- Subjects
placental pathology ,Pathology ,medicine.medical_specialty ,QH301-705.5 ,viruses ,syncytiotrophoblast ,Chronic histiocytic intervillositis ,Coronavirus placental infection ,COVID-19 ,Cytotrophoblast ,Molecular pathology ,Placental pathology ,Pregnancy ,SARS-CoV-2 ,SARS-CoV-2 in cytotrophoblast ,Syncytiotrophoblast ,cytotrophoblast ,Biology ,Article ,molecular pathology ,Placenta ,medicine ,Biology (General) ,skin and connective tissue diseases ,Molecular Biology ,reproductive and urinary physiology ,chronic histiocytic intervillositis ,fungi ,Trophoblast ,Cell Biology ,coronavirus placental infection ,Staining ,medicine.anatomical_structure ,embryonic structures ,Immunohistochemistry ,Chorionic villi ,pregnancy ,Developmental Biology - Abstract
A subset of placentas from pregnant women having the SARS-CoV-2 infection have been found to be infected with the coronavirus using molecular pathology methods including immunohistochemistry and RNA in situ hybridization. These infected placentas can demonstrate several unusual findings which occur together—chronic histiocytic intervillositis, trophoblast necrosis and positive staining of the syncytiotrophoblast for SARS-CoV-2. They frequently also have increased fibrin deposition, which can be massive in some cases. Syncytiotrophoblast is the most frequent fetal-derived cell type to be positive for SARS-CoV-2. It has recently been shown that in a small number of infected placentas, villous stromal macrophages, termed Hofbauer cells, and villous capillary endothelial cells can also stain positive for SARS-CoV-2. This report describes a placenta from a pregnant woman with SARS-CoV-2 that had chronic histiocytic intervillositis, trophoblast necrosis, increased fibrin deposition and positive staining of the syncytiotrophoblast for SARS-CoV-2. In addition, molecular pathology testing including RNAscope and immunohistochemistry for SARS-CoV-2 and double-staining immunohistochemistry using antibodies to E-cadherin and GATA3 revealed that cytotrophoblast cells stained intensely for SARS-CoV-2. All of the cytotrophoblast cells that demonstrated positive staining for SARS-CoV-2 were in direct physical contact with overlying syncytiotrophoblast that also stained positive for the virus. The pattern of cytotrophoblast staining for SARS-CoV-2 was patchy, and there were chorionic villi having diffuse positive staining of the syncytiotrophoblast for SARS-CoV-2, but without staining of cytotrophoblast. This first detailed description of cytotrophoblast involvement by SARS-CoV-2 adds another fetal cell type from infected placentas that demonstrate viral staining.
- Published
- 2021
39. Long Pentraxin-3 Follows and Modulates Bladder Cancer Progression
- Author
-
Marta Turati, Sara Rezzola, William Vermi, Sara Matarazzo, Marianna Cerasuolo, Marco Presta, Mattia Bugatti, Roberto Ronca, Laura Melocchi, Sara Taranto, Federica Maccarinelli, Arianna Giacomini, Luca Zammataro, and Elisabetta Grillo
- Subjects
0301 basic medicine ,Cancer Research ,Biology ,Fibroblast growth factor ,Bladder cancer ,FGF ,FGFR ,Long pentraxin-3 ,Metabolism ,Stemness ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,stemness ,0302 clinical medicine ,Immune system ,medicine ,Innate immune system ,Cell growth ,Cancer ,PTX3 ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,long pentraxin-3 ,Oncology ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,Cancer research ,bladder cancer ,metabolism - Abstract
Bladder tumors are a diffuse type of cancer. Long pentraxin-3 (PTX3) is a component of the innate immunity with pleiotropic functions in the regulation of immune response, tissue remodeling, and cancer progression. PTX3 may act as an oncosuppressor in different contexts, functioning as an antagonist of the fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) system, rewiring the immune microenvironment, or acting through mechanisms not yet fully clarified. In this study we used biopsies and data mining to assess that PTX3 is differentially expressed during the different stages of bladder cancer (BC) progression. BC cell lines, representative of different tumor grades, and transgenic/carcinogen-induced models were used to demonstrate in vitro and in vivo that PTX3 production by tumor cells decreases along the progression from low-grade to high-grade advanced muscle invasive forms (MIBC). In vitro and in vivo data revealed for the first time that PTX3 modulation and the consequent impairment of FGF/FGR systems in BC cells have a significant impact on different biological features of BC growth, including cell proliferation, motility, metabolism, stemness, and drug resistance. PTX3 exerts an oncosuppressive effect on BC progression and may represent a potential functional biomarker in BC evolution. Moreover, FGF/FGFR blockade has an impact on drug resistance and stemness features in BC.
- Published
- 2019
40. Heterogeneity of human intestinal intraepithelial T cells and their abnormal distribution in Crohn’s disease revealed at high resolution
- Author
-
Natalia Jaeger, Ramya Gamini, Marina Cella, Jorge L. Schettini, Mattia Bugatti, Shanrong Zhao, Charles V. Rosadini, Ekaterina Esaulova, Blanda Di Luccia, Baylee Kinnett, Willam Vermi, Maxim N. Artyomov, Thomas A Wynn, Ramnik J Xavier, Scott A. Jelinsky, and Marco Colonna
- Subjects
Immunology ,Immunology and Allergy - Abstract
Crohn’s disease (CD) is a chronic transmural inflammation of intestinal segments caused by dysregulated interaction between microbiome and gut immune system. Recurrent/relapsing CD and resistance to medical treatments result in complications requiring surgery. High-dimensional single-cell profiling approaches, such as scRNA-seq and mass cytometry, have been recently performed on intestinal specimens from patients with IBD and controls. However, most of these studies have analyzed whole mucosal biopsies or the lamina propria (LP) compartment, while few have addressed the intraepithelial lymphocytes (IEL) compartment. Here, we profiled T cells purified from the IEL and LP from terminal ileum resections of adult severe CD cases by single cell technologies. Our study defined a vast heterogeneity of T cell lineages in the IEL compartment. IEL included, among others, unique γδT cell subsets: NKp30+γδ T cells expressing RORγt, which produced IL-26 upon NKp30 engagement and a subset expressing PDGFD and CSF1, which may act on epithelial cells, IEL ILC1s, and macrophages, respectively. We have also observed long-lived memory TCF7+CD8+ T cells expressing DC chemoattractants and TFH subsets that may respond to distinct glutathione-conjugated lipids. CD IEL showed a significant increase of activated TH17, coupled with decreased CD8+ T cells, γδT cells, TFH, and Treg. Conversely, the LP showed increased CD8+ T cells and reduced CD4+ T cells with a relative increase of TH17 over Treg/TFH. Results provide an unbiased view of diversity of cell lineages and their functional states in the intestinal mucosa of controls and CD and identify an altered spatial distribution of T cell subsets between the IEL and the LP compartments.
- Published
- 2021
41. Identical TP53 mutations in pelvic carcinosarcomas and associated serous tubal intraepithelial carcinomas provide evidence of their clonal relationship
- Author
-
Antonella Ravaggi, Franco Odicino, Fabio Facchetti, Luigi Mori, Laura Ardighieri, Sara Conzadori, Marcella Falchetti, Carla Donzelli, and Mattia Bugatti
- Subjects
p53 ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Cystadenocarcinoma ,Bilateral ,Carcinosarcoma ,STIC ,TP53 ,Aged ,Carcinoma in Situ ,Cystadenocarcinoma, Serous ,Female ,Genes, p53 ,Humans ,Immunohistochemistry ,Middle Aged ,Mutation ,Ovarian Neoplasms ,Pelvic Neoplasms ,Precancerous Conditions ,2734 ,Molecular Biology ,Cell Biology ,Biology ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Peritoneum ,medicine ,Carcinoma ,Carcinoma in situ ,Serous ,Serous Tubal Intraepithelial Carcinoma ,General Medicine ,medicine.disease ,Serous fluid ,030104 developmental biology ,medicine.anatomical_structure ,Genes ,030220 oncology & carcinogenesis ,Fallopian tube - Abstract
Pelvic carcinosarcomas (PCSs) are rare aggressive biphasic tumors that localize in the ovary, fallopian tube, or peritoneum and present frequently as bilateral disease. We undertook a morphological, p53 immunohistochemical and TP53 gene mutational analysis study in a single institution cohort of 16 PCSs in order to investigate the nature of bilateral tumors and to shed light on their origin and pathogenesis. Of the 16 patients, 10 presented with bilateral disease, 6 with a carcinosarcoma in both adnexa, and the remaining cases with a carcinosarcoma in one adnexum and a carcinoma in the opposite. The carcinoma component showed high-grade serous features in 13/16 of cases (81 %). In 10 patients (63 %), a serous tubal intraepithelial carcinoma (STIC) was found, in one case bilateral, making a total of 11 STICs. STIC was found only in cases with a carcinoma component with high-grade serous features. All 10 bilateral tumors and all 11 PCS-associated STICs showed a similar p53 immunostaining pattern. At mutation analysis of the TP53 gene, all five bilateral PCS contained an identical mutation in both localizations. Furthermore, a TP53 mutation was found in 8 of 10 STICs, with an identical mutation in the associated PCS. The finding of similar p53 immunostaining in all bilateral cases and identical TP53 mutations in most PCS-associated STIC provides evidence for a clonal relation between these neoplastic lesions, supporting a metastatic nature of bilateral PCS and suggesting that they have an extraovarian origin in a STIC.
- Published
- 2016
42. Human Plasmacytoid Dendritic Cells and Cutaneous Melanoma
- Author
-
Raffaella Vescovi, Mattia Bugatti, Francesca Consoli, William Vermi, and Matilde Monti
- Subjects
Proto-Oncogene Proteins B-raf ,Skin Neoplasms ,Review ,cutaneous melanoma ,Immune system ,Interferon ,Immunity ,TLR ,medicine ,Humans ,Neoplasm Metastasis ,Receptor ,lcsh:QH301-705.5 ,Melanoma ,business.industry ,hemic and immune systems ,Dendritic Cells ,General Medicine ,MAP Kinase Kinase Kinases ,medicine.disease ,lcsh:Biology (General) ,Toll-Like Receptor 7 ,plasmacytoid dendritic cells ,Toll-Like Receptor 9 ,Interferon Type I ,Cancer cell ,Cutaneous melanoma ,Cancer research ,Immunotherapy ,Signal transduction ,business ,Signal Transduction ,medicine.drug - Abstract
The prognosis of metastatic melanoma (MM) patients has remained poor for a long time. However, the recent introduction of effective target therapies (BRAF and MEK inhibitors for BRAFV600-mutated MM) and immunotherapies (anti-CTLA-4 and anti-PD-1) has significantly improved the survival of MM patients. Notably, all these responses are highly dependent on the fitness of the host immune system, including the innate compartment. Among immune cells involved in cancer immunity, properly activated plasmacytoid dendritic cells (pDCs) exert an important role, bridging the innate and adaptive immune responses and directly eliminating cancer cells. A distinctive feature of pDCs is the production of high amount of type I Interferon (I-IFN), through the Toll-like receptor (TLR) 7 and 9 signaling pathway activation. However, published data indicate that melanoma-associated escape mechanisms are in place to hijack pDC functions. We have recently reported that pDC recruitment is recurrent in the early phases of melanoma, but the entire pDC compartment collapses over melanoma progression. Here, we summarize recent advances on pDC biology and function within the context of melanoma immunity.
- Published
- 2020
43. Tumor Infiltrating Neutrophils Are Enriched in Basal-Type Urothelial Bladder Cancer
- Author
-
Elena Caveggion, Matilde Monti, Sandra Belotti, Claudio Simeone, Luca Cristinelli, Luisa Benerini Gatta, Giulio Eugenio Mandelli, Stefano Calza, William Vermi, Laura Melocchi, Patrizia Scapini, Mattia Bugatti, Francesco Missale, Debora Bresciani, and Elisa Roca
- Subjects
Male ,STAT3 Transcription Factor ,Chemokine ,tumor-associated neutrophils ,CD3 Complex ,Cell Survival ,Neutrophils ,Chemokine CXCL1 ,T-Lymphocytes ,medicine.medical_treatment ,CD3 ,Chemokine CXCL2 ,CD66b ,Kaplan-Meier Estimate ,Article ,Basal (phylogenetics) ,Immune system ,Cell Movement ,Cell Line, Tumor ,Databases, Genetic ,basal ,medicine ,Humans ,Gene Silencing ,Interleukin 8 ,lcsh:QH301-705.5 ,Aged ,Retrospective Studies ,Aged, 80 and over ,Muscle Neoplasms ,Bladder cancer ,biology ,Interleukin-8 ,General Medicine ,Immunotherapy ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Carcinoembryonic Antigen ,CXCL1 ,bladder cancer ,lcsh:Biology (General) ,Urinary Bladder Neoplasms ,biology.protein ,Cancer research ,Female - Abstract
Background: Urothelial bladder cancers (UBCs) are distinct in two main molecular subtypes, namely basal and luminal type. Subtypes are also diverse in term of immune contexture, providing a rationale for patient selection to immunotherapy. Methods: By digital microscopy analysis of a muscle-invasive BC (MIBC) cohort, we explored the density and clinical significance of CD66b+ tumor-associated-neutrophils (TAN) and CD3+ T cells. Bioinformatics analysis of UBC datasets and gene expression analysis of UBC cell lines were additionally performed. Results: Basal type BC contained a significantly higher density of CD66b+ TAN compared to the luminal type. This finding was validated on TCGA, GSE32894 and GSE124305 datasets by computing a neutrophil signature. Of note, basal-type MIBC display a significantly higher level of chemokines (CKs) attracting neutrophils. Moreover, pro-inflammatory stimuli significantly up-regulate CXCL1, CXCL2 and CXCL8 in 5637 and RT4 UBC cell lines and induce neutrophil chemotaxis. In term of survival, a high density of T cells and TAN was significantly associated to a better outcome, with TAN density showing a more limited statistical power and following a non-linear predicting model. Conclusions: TAN are recruited in basal type MIBC by pro-inflammatory CKs. This finding establishes a groundwork for a better understanding of the UBC immunity and its relevance.
- Published
- 2020
44. Abstract C121: Long Pentraxin-3 modulates bladder cancer progression
- Author
-
William Vermi, Sara Matarazzo, Federica Maccarinelli, Sara Rezzola, Luca Zammataro, Sara Taranto, Arianna Giacomini, Mattia Bugatti, Roberto Ronca, Laura Melocchi, Marianna Cerasuolo, Marco Presta, and Elisabetta Grillo
- Subjects
Cancer Research ,Innate immune system ,Bladder cancer ,business.industry ,Cell growth ,Cancer ,PTX3 ,medicine.disease ,Fibroblast growth factor ,Immune system ,Oncology ,Cancer research ,Biomarker (medicine) ,Medicine ,business - Abstract
Bladder tumors are a diffuse type of cancer with a relatively benign prognosis when treated at early stage/low-grade stages, but with poor outcome in the muscle invasive (MIBC) form or at recurrence. Long Pentraxin-3 (PTX3) is a component of the innate immunity with pleiotropic functions in the regulation of immune response, tissue remodeling and cancer progression. PTX3 may act as an oncosuppressor in different contexts, functioning as an antagonist of the FGF/FGFR system, rewiring the immune-microenvironment or acting through mechanisms not yet fully clarified. In this study, we report for the first time that PTX3 is differentially expressed in bladder cancer (BC) biopsies and tumor cell lines during the different stages of BC progression. Taking advantage of BC cell lines representative of different tumor grades, we demonstrate that PTX3 production by tumor cells decreases along the progression from low-grade to high grade/MIBC because of promoter methylation. In vitro and in vivo data reveal that PTX3 modulation in BC cells has a significant impact on different biological features of BC growth, including cell proliferation, motility, metabolism, stemness and drug resistance. In conclusion, PTX3 exerts an oncosuppressive effect on BC progression and may represent a potential functional biomarker in BC evolution. Citation Format: Sara Matarazzo, Laura Melocchi, Sara Rezzola, Elisabetta Grillo, Federica Maccarinelli, Arianna Giacomini, Sara Taranto, Luca Zammataro, Marianna Cerasuolo, Mattia Bugatti, William Vermi, Marco Presta, Roberto Ronca. Long Pentraxin-3 modulates bladder cancer progression [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr C121. doi:10.1158/1535-7163.TARG-19-C121
- Published
- 2019
45. slan
- Author
-
William, Vermi, Alessandra, Micheletti, Giulia, Finotti, Cristina, Tecchio, Federica, Calzetti, Sara, Costa, Mattia, Bugatti, Stefano, Calza, Claudio, Agostinelli, Stefano, Pileri, Piera, Balzarini, Alessandra, Tucci, Giuseppe, Rossi, Lara, Furlani, Giuseppe, Todeschini, Alberto, Zamò, Fabio, Facchetti, Luisa, Lorenzi, Silvia, Lonardi, and Marco A, Cassatella
- Subjects
Adult ,Male ,Adolescent ,Biopsy ,Primary Cell Culture ,Monocytes ,Cohort Studies ,Young Adult ,Antineoplastic Agents, Immunological ,Tumor Microenvironment ,Humans ,Child ,Cells, Cultured ,Aged ,Retrospective Studies ,Aged, 80 and over ,Macrophages ,Tumor Suppressor Proteins ,Antibody-Dependent Cell Cytotoxicity ,Middle Aged ,Antigens, CD20 ,Cytophagocytosis ,Child, Preschool ,Leukocytes, Mononuclear ,Female ,Lymph Nodes ,Lymphoma, Large B-Cell, Diffuse ,Rituximab - Abstract
Terminal tissue differentiation and function of slan
- Published
- 2017
46. Correction: slanDCs/M-DC8+ cells constitute a distinct subset of dendritic cells in human tonsils
- Author
-
Stefania Stefini, William Vermi, Elisa Zoratti, Giulia Finotti, Federica Calzetti, Marco A. Cassatella, Mattia Bugatti, Silvia Lonardi, and Alessandra Micheletti
- Subjects
0301 basic medicine ,T-Lymphocytes ,Palatine Tonsil ,CX3C Chemokine Receptor 1 ,Lipopolysaccharide Receptors ,Biology ,Immunophenotyping ,03 medical and health sciences ,Text mining ,stomatognathic system ,Humans ,dendritic cells ,Immune response ,slan/M-DC8+ cells ,Cells, Cultured ,Antigen Presentation ,Follicular dendritic cells ,business.industry ,CD11 Antigens ,Tumor Necrosis Factor-alpha ,Receptors, IgG ,Research Paper: Immunology ,Immunity ,Correction ,HLA-DR Antigens ,differentiation ,Immunohistochemistry ,Cell biology ,CD11c Antigen ,030104 developmental biology ,Oncology ,tonsil ,Immunology and Microbiology Section ,Receptors, Chemokine ,business ,monocytes - Abstract
Human blood dendritic cells (DCs) include three main distinct subsets, namely the CD1c+ and CD141+ myeloid DCs (mDCs) and the CD303+ plasmacytoid DCs (pDCs). More recently, a population of slan/M-DC8+ cells, also known as "slanDCs", has been described in blood and detected even in inflamed secondary lymphoid organs and non-lymphoid tissues. Nevertheless, hallmarks of slan/M-DC8+ cells in tissues are poorly defined. Herein, we report a detailed characterization of the phenotype and function of slan/M-DC8+ cells present in human tonsils. We found that tonsil slan/M-DC8+ cells represent a unique DC cell population, distinct from their circulating counterpart and also from all other tonsil DC and monocyte/macrophage subsets. Phenotypically, slan/M-DC8+ cells in tonsils display a CD11c+HLA-DR+CD14+CD11bdim/negCD16dim/negCX3CR1dim/neg marker repertoire, while functionally they exhibit an efficient antigen presentation capacity and a constitutive secretion of TNFα. Notably, such DC phenotype and functions are substantially reproduced by culturing blood slan/M-DC8+ cells in tonsil-derived conditioned medium (TDCM), further supporting the hypothesis of a full DC-like differentiation program occurring within the tonsil microenvironment. Taken together, our data suggest that blood slan/M-DC8+ cells are immediate precursors of a previously unrecognizedcompetent DC subset in tonsils, and pave the way for further characterization of slan/M-DC8+ cells in other tissues.
- Published
- 2017
47. Early, transient depletion of plasmacytoid dendritic cells ameliorates autoimmunity in a lupus model
- Author
-
William Vermi, Miguel A. Sanjuan, Sarah L. Rowland, Jeffrey M. Riggs, Susan Gilfillan, Emil R. Unanue, Roland Kolbeck, Marco Colonna, and Mattia Bugatti
- Subjects
0303 health sciences ,Lupus erythematosus ,Systemic lupus erythematosus ,Immunology ,Alpha interferon ,hemic and immune systems ,Glomerulonephritis ,Biology ,medicine.disease_cause ,medicine.disease ,3. Good health ,Autoimmunity ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,biology.protein ,Immunology and Allergy ,Antibody ,030304 developmental biology ,030215 immunology - Abstract
Plasmacytoid dendritic cells (pDCs) have long been implicated in the pathogenesis of lupus. However, this conclusion has been largely based on a correlative link between the copious production of IFN-α/β by pDCs and the IFN-α/β “signature” often seen in human lupus patients. The specific contribution of pDCs to disease in vivo has not been investigated in detail. For this reason, we generated a strain of BXSB lupus-prone mice in which pDCs can be selectively depleted in vivo. Early, transient ablation of pDCs before disease initiation resulted in reduced splenomegaly and lymphadenopathy, impaired expansion and activation of T and B cells, reduced antibodies against nuclear autoantigens and improved kidney pathology. Amelioration of pathology coincided with decreased transcription of IFN-α/β–induced genes in tissues. PDC depletion had an immediate impact on the activation of immune cells, and importantly, the beneficial effects on pathology were sustained even though pDCs later recovered, indicating an early pDC contribution to disease. Together, our findings demonstrate a critical function for pDCs during the IFN-α/β–dependent initiation of autoimmune lupus and point to pDCs as an attractive therapeutic target for the treatment of SLE.
- Published
- 2014
48. Abstract A039: FGF/PTX3 crosstalk in bladder cancer: novel prognostic and therapeutic implications
- Author
-
Mattia Bugatti, Marco Presta, Gaia C. Ghedini, Federica Maccarinelli, William Vermi, Sara Matarazzo, Roberto Ronca, and Laura Melocchi
- Subjects
Cancer Research ,Bladder cancer ,business.industry ,Fibroblast growth factor receptor 1 ,Fibroblast growth factor ,medicine.disease ,Oncology ,Fibroblast growth factor receptor ,Cancer cell ,Cancer research ,Medicine ,Urothelium ,business ,Receptor ,Survival rate - Abstract
Bladder cancer is one of the most common cancers in the world. Although most of urothelial carcinomas (UCs) are low-grade papillary tumors, their propensity to recur and to progress to become invasive with a poor survival rate represents a major challenge. Different members of the Fibroblast growth factor (FGF) family are expressed by human bladder cancer cells, and aberrant expression of FGF receptors (FGFRs) is a typical feature of bladder cancer compared to normal urothelium. High expression levels of FGFR1 are associated with poor survival and FGFR3 is frequently dysregulated in UC. Thus, the FGF/FGFR system may represent a promising therapeutic target in invasive and noninvasive bladder cancer. Long-pentraxin 3 (PTX3) acts as a natural FGF trap by binding FGFs and hampering their biologic activity in various tumor models. Preliminary observations have shown that low-grade UCs express PTX3 while high-grade/invasive UCs lose PTX3 expression. Accordingly, bladder cancer cell lines share an overall presence of FGFRs and FGFs but express different levels of PTX3 in vitro and in vivo. Of note, high-grade/invasive cells express very low or undetectable levels of PTX3, whereas low-grade, papilloma-like cells express high levels of PTX3. Indeed, invasive bladder cancer cell xenografts grow faster in nude mice and express much lower levels of PTX3 than low grade-derived lesions, thus confirming an inverse correlation between UC grade and PTX3 expression. This hypothesis is reinforced by a preliminary case study performed on a small cohort of biopsies from UC patients confirming that the presence of PTX3 is a common feature of low-grade samples while PTX3 is poorly expressed or absent in aggressive/invasive cases. These data point to PTX3 as a natural FGF trap that could play a role in modulating bladder cancer progression and invasiveness and indicate that inhibition of the FGF/FGFR system by natural and synthetic FGF traps may represent a promising target for the therapy of UCs. Citation Format: Sara Matarazzo, Federica Maccarinelli, Gaia Cristina Ghedini, Laura Melocchi, Mattia Bugatti, William Vermi, Marco Presta, Roberto Ronca. FGF/PTX3 crosstalk in bladder cancer: novel prognostic and therapeutic implications [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr A039.
- Published
- 2018
49. Natural Killer Cells Control Tumor Growth by Sensing a Growth Factor
- Author
-
Vladimir Trifonov, Melissa A. Edeling, Piyush Goyal, Sarah Cox, Marco Colonna, Christian Schmedt, William Vermi, Alexander D. Barrow, Benjamin Bohl, Albert H. Kim, Laura Melocchi, John R. Walker, Jingqin Luo, Daved H. Fremont, Mary Andahazy, Jennifer K. Bando, Mattia Bugatti, and Marina Cella
- Subjects
Cytotoxicity, Immunologic ,Male ,0301 basic medicine ,medicine.medical_treatment ,immunosurveillance ,Inbred C57BL ,Mice ,0302 clinical medicine ,Neoplasms ,Cricetinae ,Innate ,Killer Cells ,Platelet-Derived Growth Factor ,Cultured ,Brain Neoplasms ,Innate lymphoid cell ,growth factor ,PDGF-D ,Killer Cells, Natural ,030220 oncology & carcinogenesis ,Natural ,MCF-7 Cells ,cell cycle ,Female ,Tumor necrosis factor alpha ,Platelet-derived growth factor receptor ,Stromal cell ,Cells ,innate lymphoid cells ,CHO Cells ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Interferon-gamma ,03 medical and health sciences ,Paracrine signalling ,Cricetulus ,Tumor Expansion ,medicine ,cancer ,Animals ,Humans ,NK cell ,Autocrine signalling ,Natural Cytotoxicity Triggering Receptor 2 ,Tumor Necrosis Factor-alpha ,Growth factor ,Immunity ,cytokines ,NKp44 ,Cells, Cultured ,Glioblastoma ,Immunity, Innate ,Mice, Inbred C57BL ,Cell Cycle Checkpoints ,030104 developmental biology ,biology.protein ,Cancer research - Abstract
Many tumors produce platelet-derived growth factor (PDGF)-DD, which promotes cellular proliferation, epithelial-mesenchymal transition, stromal reaction, and angiogenesis through autocrine and paracrine PDGFRβ signaling. By screening a secretome library, we found that the human immunoreceptor NKp44, encoded by NCR2 and expressed on natural killer (NK) cells and innate lymphoid cells, recognizes PDGF-DD. PDGF-DD engagement of NKp44 triggered NK cell secretion of interferon gamma (IFN)-γ and tumor necrosis factor alpha (TNF-α) that induced tumor cell growth arrest. A distinctive transcriptional signature of PDGF-DD-induced cytokines and the downregulation of tumor cell-cycle genes correlated with NCR2 expression and greater survival in glioblastoma. NKp44 expression in mouse NK cells controlled the dissemination of tumors expressing PDGF-DD more effectively than control mice, an effect enhanced by blockade of the inhibitory receptor CD96 or CpG-oligonucleotide treatment. Thus, while cancer cell production of PDGF-DD supports tumor growth and stromal reaction, it concomitantly activates innate immune responses to tumor expansion.
- Published
- 2018
50. Truncating Prolactin Receptor Mutations Promote Tumor Growth in Murine Estrogen Receptor-Alpha Mammary Carcinomas
- Author
-
Krishna-Latha Kanchi, Lee Trani, Robert D. Schreiber, Alexander P. Miceli, Christopher A. Miller, Jasreet Hundal, Mattia Bugatti, Heather Schmidt, Malachi Griffith, David E. Larson, Cora D. Arthur, Obi L. Griffith, Rick K. Wilson, Daniele Runci, Elaine R. Mardis, Kilannin Krysiak, Szeman Ruby Chan, William Vermi, Julie A. Allen, Zachary L. Skidmore, and Robert S. Fulton
- Subjects
0301 basic medicine ,Genetics and Molecular Biology (all) ,Estrogen receptor ,medicine.disease_cause ,Biochemistry ,Mice ,STAT1 ,STAT5 Transcription Factor ,Receptor ,lcsh:QH301-705.5 ,STAT5 ,Mice, Knockout ,Mutation ,whole genome sequencing ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,STAT1 Transcription Factor ,Female ,Signal Transduction ,STAT3 Transcription Factor ,Receptors, Prolactin ,mouse model ,Breast Neoplasms ,Mammary Neoplasms, Animal ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,breast cancer ,estrogen-receptor positive ,medicine ,luminal ,PRLR ,Biochemistry, Genetics and Molecular Biology (all) ,Animals ,Humans ,Prolactin receptor ,Carcinoma ,Estrogen Receptor alpha ,Cancer ,Fibroblasts ,medicine.disease ,Embryo, Mammalian ,030104 developmental biology ,lcsh:Biology (General) ,Cancer research ,biology.protein ,Carcinogenesis ,Estrogen receptor alpha - Abstract
SummaryEstrogen receptor alpha-positive (ERα+) luminal tumors are the most frequent subtype of breast cancer. Stat1−/− mice develop mammary tumors that closely recapitulate the biological characteristics of this cancer subtype. To identify transforming events that contribute to tumorigenesis, we performed whole genome sequencing of Stat1−/− primary mammary tumors and matched normal tissues. This investigation identified somatic truncating mutations affecting the prolactin receptor (PRLR) in all tumor and no normal samples. Targeted sequencing confirmed the presence of these mutations in precancerous lesions, indicating that this is an early event in tumorigenesis. Functional evaluation of these heterozygous mutations in Stat1−/− mouse embryonic fibroblasts showed that co-expression of truncated and wild-type PRLR led to aberrant STAT3 and STAT5 activation downstream of the receptor, cellular transformation in vitro, and tumor formation in vivo. In conclusion, truncating mutations of PRLR promote tumor growth in a model of human ERα+ breast cancer and warrant further investigation.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.