1,449 results on '"Serafini M"'
Search Results
2. First-hit SETBP1 mutations cause a myeloproliferative disorder with bone marrow fibrosis
- Author
-
Crespiatico, I, Zaghi, M, Mastini, C, D'Aliberti, D, Mauri, M, Mercado, C, Fontana, D, Spinelli, S, Crippa, V, Inzoli, E, Manghisi, B, Civettini, I, Ramazzotti, D, Sangiorgio, V, Gengotti, M, Brambilla, V, Aroldi, A, Banfi, F, Barone, C, Orsenigo, R, Riera, L, Riminucci, M, Corsi, A, Breccia, M, Morotti, A, Cilloni, D, Roccaro, A, Sacco, A, Stagno, F, Serafini, M, Mottadelli, F, Cazzaniga, G, Pagni, F, Chiarle, R, Azzoni, E, Sessa, A, Gambacorti Passerini, C, Elli, E, Mologni, L, Piazza, R, Crespiatico I., Zaghi M., Mastini C., D'Aliberti D., Mauri M., Mercado C. M., Fontana D., Spinelli S., Crippa V., Inzoli E., Manghisi B., Civettini I., Ramazzotti D., Sangiorgio V., Gengotti M., Brambilla V., Aroldi A., Banfi F., Barone C., Orsenigo R., Riera L., Riminucci M., Corsi A., Breccia M., Morotti A., Cilloni D., Roccaro A., Sacco A., Stagno F., Serafini M., Mottadelli F., Cazzaniga G., Pagni F., Chiarle R., Azzoni E., Sessa A., Gambacorti Passerini C., Elli E. M., Mologni L., Piazza R., Crespiatico, I, Zaghi, M, Mastini, C, D'Aliberti, D, Mauri, M, Mercado, C, Fontana, D, Spinelli, S, Crippa, V, Inzoli, E, Manghisi, B, Civettini, I, Ramazzotti, D, Sangiorgio, V, Gengotti, M, Brambilla, V, Aroldi, A, Banfi, F, Barone, C, Orsenigo, R, Riera, L, Riminucci, M, Corsi, A, Breccia, M, Morotti, A, Cilloni, D, Roccaro, A, Sacco, A, Stagno, F, Serafini, M, Mottadelli, F, Cazzaniga, G, Pagni, F, Chiarle, R, Azzoni, E, Sessa, A, Gambacorti Passerini, C, Elli, E, Mologni, L, Piazza, R, Crespiatico I., Zaghi M., Mastini C., D'Aliberti D., Mauri M., Mercado C. M., Fontana D., Spinelli S., Crippa V., Inzoli E., Manghisi B., Civettini I., Ramazzotti D., Sangiorgio V., Gengotti M., Brambilla V., Aroldi A., Banfi F., Barone C., Orsenigo R., Riera L., Riminucci M., Corsi A., Breccia M., Morotti A., Cilloni D., Roccaro A., Sacco A., Stagno F., Serafini M., Mottadelli F., Cazzaniga G., Pagni F., Chiarle R., Azzoni E., Sessa A., Gambacorti Passerini C., Elli E. M., Mologni L., and Piazza R.
- Abstract
SETBP1 mutations are found in various clonal myeloid disorders. However, it is unclear whether they can initiate leukemia, because SETBP1 mutations typically appear as later events during oncogenesis. To answer this question, we generated a mouse model expressing mutated SETBP1 in hematopoietic tissue: this model showed profound alterations in the differentiation program of hematopoietic progenitors and developed a myeloid neoplasm with megakaryocytic dysplasia, splenomegaly, and bone marrow fibrosis, prompting us to investigate SETBP1 mutations in a cohort of 36 triple-negative primary myelofibrosis (TN-PMF) cases. We identified 2 distinct subgroups, one carrying SETBP1 mutations and the other completely devoid of somatic variants. Clinically, a striking difference in disease aggressiveness was noted, with patients with SETBP1 mutation showing a much worse clinical course. In contrast to myelodysplastic/myeloproliferative neoplasms, in which SETBP1 mutations are mostly found as a late clonal event, single-cell clonal hierarchy reconstruction in 3 patients with TN-PMF from our cohort revealed SETBP1 to be a very early event, suggesting that the phenotype of the different SETBP1+ disorders may be shaped by the opposite hierarchy of the same clonal SETBP1 variants.
- Published
- 2024
3. Modeling skeletal dysplasia in Hurler syndrome using patient-derived bone marrow osteoprogenitor cells
- Author
-
Donsante, S, Pievani, A, Palmisano, B, Finamore, M, Fazio, G, Corsi, A, Biondi, A, Tomatsu, S, Piazza, R, Serafini, M, Riminucci, M, Donsante S., Pievani A., Palmisano B., Finamore M., Fazio G., Corsi A., Biondi A., Tomatsu S., Piazza R., Serafini M., Riminucci M., Donsante, S, Pievani, A, Palmisano, B, Finamore, M, Fazio, G, Corsi, A, Biondi, A, Tomatsu, S, Piazza, R, Serafini, M, Riminucci, M, Donsante S., Pievani A., Palmisano B., Finamore M., Fazio G., Corsi A., Biondi A., Tomatsu S., Piazza R., Serafini M., and Riminucci M.
- Abstract
Dysostosis multiplex is a major cause of morbidity in Hurler syndrome (mucopolysaccharidosis type IH [MPS IH], OMIM #607014) because currently available therapies have limited success in its prevention and reversion. Unfortunately, the elucidation of skeletal pathogenesis in MPS IH is limited by difficulties in obtaining bone specimens from pediatric patients and poor reproducibility in animal models. Thus, the application of experimental systems that can be used to dissect cellular and molecular mechanisms underlying the skeletal phenotype of MPS IH patients and to identify effective therapies is highly needed. Here, we adopted in vitro/in vivo systems based on patient-derived bone marrow stromal cells to generate cartilaginous pellets and bone rudiments. Interestingly, we observed that heparan sulphate accumulation compromised the remodeling of MPS IH cartilage into other skeletal tissues and other critical aspects of the endochondral ossification process. We also noticed that MPS IH hypertrophic cartilage was characterized by dysregulation of signaling pathways controlling cartilage hypertrophy and fate, extracellular matrix organization, and glycosaminoglycan metabolism. Our study demonstrates that the cartilaginous pellet–based system is a valuable tool to study MPS IH dysostosis and to develop new therapeutic approaches for this hard-to-treat aspect of the disease. Finally, our approach may be applied for modeling other genetic skeletal disorders.
- Published
- 2024
4. Exonic knockout and knockin gene editing in hematopoietic stem and progenitor cells rescues RAG1 immunodeficiency
- Author
-
Castiello, M, Brandas, C, Ferrari, S, Porcellini, S, Sacchetti, N, Canarutto, D, Draghici, E, Merelli, I, Barcella, M, Pelosi, G, Vavassori, V, Varesi, A, Jacob, A, Scala, S, Basso Ricci, L, Paulis, M, Strina, D, Di Verniere, M, Sergi Sergi, L, Serafini, M, Holland, S, Bergerson, J, De Ravin, S, Malech, H, Pala, F, Bosticardo, M, Brombin, C, Cugnata, F, Calzoni, E, Crooks, G, Notarangelo, L, Genovese, P, Naldini, L, Villa, A, Castiello, Maria Carmina, Brandas, Chiara, Ferrari, Samuele, Porcellini, Simona, Sacchetti, Nicolò, Canarutto, Daniele, Draghici, Elena, Merelli, Ivan, Barcella, Matteo, Pelosi, Gabriele, Vavassori, Valentina, Varesi, Angelica, Jacob, Aurelien, Scala, Serena, Basso Ricci, Luca, Paulis, Marianna, Strina, Dario, Di Verniere, Martina, Sergi Sergi, Lucia, Serafini, Marta, Holland, Steven M., Bergerson, Jenna R. E., De Ravin, Suk See, Malech, Harry L., Pala, Francesca, Bosticardo, Marita, Brombin, Chiara, Cugnata, Federica, Calzoni, Enrica, Crooks, Gay M., Notarangelo, Luigi D., Genovese, Pietro, Naldini, Luigi, Villa, Anna, Castiello, M, Brandas, C, Ferrari, S, Porcellini, S, Sacchetti, N, Canarutto, D, Draghici, E, Merelli, I, Barcella, M, Pelosi, G, Vavassori, V, Varesi, A, Jacob, A, Scala, S, Basso Ricci, L, Paulis, M, Strina, D, Di Verniere, M, Sergi Sergi, L, Serafini, M, Holland, S, Bergerson, J, De Ravin, S, Malech, H, Pala, F, Bosticardo, M, Brombin, C, Cugnata, F, Calzoni, E, Crooks, G, Notarangelo, L, Genovese, P, Naldini, L, Villa, A, Castiello, Maria Carmina, Brandas, Chiara, Ferrari, Samuele, Porcellini, Simona, Sacchetti, Nicolò, Canarutto, Daniele, Draghici, Elena, Merelli, Ivan, Barcella, Matteo, Pelosi, Gabriele, Vavassori, Valentina, Varesi, Angelica, Jacob, Aurelien, Scala, Serena, Basso Ricci, Luca, Paulis, Marianna, Strina, Dario, Di Verniere, Martina, Sergi Sergi, Lucia, Serafini, Marta, Holland, Steven M., Bergerson, Jenna R. E., De Ravin, Suk See, Malech, Harry L., Pala, Francesca, Bosticardo, Marita, Brombin, Chiara, Cugnata, Federica, Calzoni, Enrica, Crooks, Gay M., Notarangelo, Luigi D., Genovese, Pietro, Naldini, Luigi, and Villa, Anna
- Abstract
Recombination activating genes (RAGs) are tightly regulated during lymphoid differentiation, and their mutations cause a spectrum of severe immunological disorders. Hematopoietic stem and progenitor cell (HSPC) transplantation is the treatment of choice but is limited by donor availability and toxicity. To overcome these issues, we developed gene editing strategies targeting a corrective sequence into the human RAG1 gene by homology-directed repair (HDR) and validated them by tailored two-dimensional, three-dimensional, and in vivo xenotransplant platforms to assess rescue of expression and function. Whereas integration into intron 1 of RAG1 achieved suboptimal correction, in-frame insertion into exon 2 drove physiologic human RAG1 expression and activity, allowing disruption of the dominant-negative effects of unrepaired hypomorphic alleles. Enhanced HDR-mediated gene editing enabled the correction of human RAG1 in HSPCs from patients with hypomorphic RAG1 mutations to overcome T and B cell differentiation blocks. Gene correction efficiency exceeded the minimal proportion of functional HSPCs required to rescue immunodeficiency in Rag1(-/- )mice, supporting the clinical translation of HSPC gene editing for the treatment of RAG1 deficiency.
- Published
- 2024
5. CARs are sharpening their weapons
- Author
-
Pievani, A, Biondi, M, Tettamanti, S, Biondi, A, Dotti, G, Serafini, M, Pievani, Alice, Biondi, Marta, Tettamanti, Sarah, Biondi, Andrea, Dotti, Gianpietro, Serafini, Marta, Pievani, A, Biondi, M, Tettamanti, S, Biondi, A, Dotti, G, Serafini, M, Pievani, Alice, Biondi, Marta, Tettamanti, Sarah, Biondi, Andrea, Dotti, Gianpietro, and Serafini, Marta
- Abstract
The tumor microenvironment hinders CAR T-cell access, activation, and persistence at the tumor site, thereby impacting on the therapeutic efficacy. To tackle these obstacles, ongoing efforts are focusing on further engineering CAR T-cells to enhance their homing, fitness, long-term persistence, and antitumor activity. Advances in genetic modification have prompted the development of armored CAR T-cells equipped with a combination of synergistic elements strengthening their function. These include cytokine release, chemokine receptor expression, immune checkpoint inhibition, gene-editing of inhibitory molecules, or metabolic reprogramming, among others. Multiarmored CAR T-cells may allow addressing the unmet clinical needs of patients with solid tumors or hard-to-treat hematological malignancies who do not benefit from conventional CAR T-cell therapy. Accordingly, several clinical trials are currently assessing the safety and efficacy of these novel CAR constructs.
- Published
- 2024
6. Glossary and Tutorial of Xenobiotic Metabolism Terms Used During Small-Molecule Drug Discovery and Development
- Author
-
Erhardt, Paul, primary, Bachmann, Kenneth, additional, Birkett, Donald, additional, Boberg, Michael, additional, Bodor, Nicholas, additional, Gibson, Gordon, additional, Hawkins, David, additional, Hawksworth, Gabrielle, additional, Hinson, Jack, additional, Koehler, Daniel, additional, Kress, Brian, additional, Luniwal, Amarjit, additional, Matsumoto, Hiroshi, additional, Novak, Raymond, additional, Portoghese, Phillip, additional, Sarver, Jeffrey, additional, Serafini, M. Teresa, additional, Trabbic, Christopher, additional, Vermeulen, Nico, additional, and Wrighton, Steven, additional
- Published
- 2021
- Full Text
- View/download PDF
7. dLLME-μSPE extraction coupled to HPLC-ESI-MS/MS for the determination of F2α-IsoPs in human urine
- Author
-
Fanti, F., Vincenti, F., Montesano, C., Serafini, M., Compagnone, D., and Sergi, M.
- Published
- 2020
- Full Text
- View/download PDF
8. An in vivo humanized model to study homing and sequestration of Plasmodium falciparum transmission stages in the bone marrow.
- Author
-
Donsante, S, Siciliano, G, Ciardo, M, Palmisano, B, Messina, V, de Turris, V, Farinacci, G, Serafini, M, Silvestrini, F, Corsi, A, Riminucci, M, Alano, P, Donsante S, Siciliano G, Ciardo M, Palmisano B, Messina V, de Turris V, Farinacci G, Serafini M, Silvestrini F, Corsi A, Riminucci M, Alano P., Donsante, S, Siciliano, G, Ciardo, M, Palmisano, B, Messina, V, de Turris, V, Farinacci, G, Serafini, M, Silvestrini, F, Corsi, A, Riminucci, M, Alano, P, Donsante S, Siciliano G, Ciardo M, Palmisano B, Messina V, de Turris V, Farinacci G, Serafini M, Silvestrini F, Corsi A, Riminucci M, and Alano P.
- Abstract
Introduction: Recent evidence suggests that the bone marrow (BM) plays a key role in the diffusion of P. falciparum malaria by providing a “niche” for the maturation of the parasite gametocytes, responsible for human-to-mosquito transmission. Suitable humanized in vivo models to study the mechanisms of the interplay between the parasite and the human BM components are still missing. Methods: We report a novel experimental system based on the infusion of immature P. falciparum gametocytes into immunocompromised mice carrying chimeric ectopic ossicles whose stromal and bone compartments derive from human osteoprogenitor cells. Results: We demonstrate that immature gametocytes home within minutes to the ossicles and reach the extravascular regions, where they are retained in contact with different human BM stromal cell types. Discussion: Our model represents a powerful tool to study BM function and the interplay essential for parasite transmission in P. falciparum malaria and can be extended to study other infections in which the human BM plays a role.
- Published
- 2023
9. Engineering tandem CD33xCD146 CAR CIK (cytokine-induced killer) cells to target the acute myeloid leukemia niche
- Author
-
Alberti, G, Arsuffi, C, Pievani, A, Salerno, D, Mantegazza, F, Dazzi, F, Biondi, A, Tettamanti, S, Serafini, M, Alberti G., Arsuffi C., Pievani A., Salerno D., Mantegazza F., Dazzi F., Biondi A., Tettamanti S., Serafini M., Alberti, G, Arsuffi, C, Pievani, A, Salerno, D, Mantegazza, F, Dazzi, F, Biondi, A, Tettamanti, S, Serafini, M, Alberti G., Arsuffi C., Pievani A., Salerno D., Mantegazza F., Dazzi F., Biondi A., Tettamanti S., and Serafini M.
- Abstract
In acute myeloid leukemia (AML), malignant stem cells hijack the normal bone marrow niche where they are largely protected from the current therapeutic approaches. Thus, eradicating these progenitors is the ultimate challenge in the treatment of this disease. Specifically, the development of chimeric antigen receptors (CARs) against distinct mesenchymal stromal cell subpopulations involved in the maintenance of leukemic stem cells within the malignant bone marrow microenvironment could represent a new strategy to improve CAR T-cell therapy efficacy, which is still unsuccessful in AML. As a proof of concept, we generated a novel prototype of Tandem CAR, with one specificity directed against the leukemic cell marker CD33 and the other against the mesenchymal stromal cell marker CD146, demonstrating its capability of simultaneously targeting two different cell types in a 2D co-culture system. Interestingly, we could also observe an in vitro inhibition of CAR T cell functionality mediated by stromal cells, particularly in later effector functions, such as reduction of interferon-gamma and interleukin-2 release and impaired proliferation of the CAR+ effector Cytokine-Induced Killer (CIK) cells. Taken together, these data demonstrate the feasibility of a dual targeting model against two molecules, which are expressed on two different target cells, but also highlight the immunomodulatory effect on CAR CIK cells exerted by stromal cells, confirming that the niche could be an obstacle to the efficacy of CAR T cells. This aspect should be considered in the development of novel CAR T cell approaches directed against the AML bone marrow niche.
- Published
- 2023
10. Engineering tandem CD33xCD146 CAR CIK (cytokine-induced killer) cells to target the acute myeloid leukemia niche
- Author
-
Alberti G., Arsuffi C., Pievani A., Salerno D., Mantegazza F., Dazzi F., Biondi A., Tettamanti S., Serafini M., Alberti, G, Arsuffi, C, Pievani, A, Salerno, D, Mantegazza, F, Dazzi, F, Biondi, A, Tettamanti, S, and Serafini, M
- Subjects
tandem CAR ,cytokine-induced killer (CIK cells) ,MSCs (mesenchymal stem cells) ,Immunology ,Immunology and Allergy ,acute myeloid leukemia ,AML niche - Abstract
In acute myeloid leukemia (AML), malignant stem cells hijack the normal bone marrow niche where they are largely protected from the current therapeutic approaches. Thus, eradicating these progenitors is the ultimate challenge in the treatment of this disease. Specifically, the development of chimeric antigen receptors (CARs) against distinct mesenchymal stromal cell subpopulations involved in the maintenance of leukemic stem cells within the malignant bone marrow microenvironment could represent a new strategy to improve CAR T-cell therapy efficacy, which is still unsuccessful in AML. As a proof of concept, we generated a novel prototype of Tandem CAR, with one specificity directed against the leukemic cell marker CD33 and the other against the mesenchymal stromal cell marker CD146, demonstrating its capability of simultaneously targeting two different cell types in a 2D co-culture system. Interestingly, we could also observe an in vitro inhibition of CAR T cell functionality mediated by stromal cells, particularly in later effector functions, such as reduction of interferon-gamma and interleukin-2 release and impaired proliferation of the CAR+ effector Cytokine-Induced Killer (CIK) cells. Taken together, these data demonstrate the feasibility of a dual targeting model against two molecules, which are expressed on two different target cells, but also highlight the immunomodulatory effect on CAR CIK cells exerted by stromal cells, confirming that the niche could be an obstacle to the efficacy of CAR T cells. This aspect should be considered in the development of novel CAR T cell approaches directed against the AML bone marrow niche.
- Published
- 2023
- Full Text
- View/download PDF
11. Is TRIZ an Ecodesign Method?
- Author
-
Russo, D., Serafini, M., Rizzi, C., Howlett, Robert J., Series editor, Jain, Lakhmi C., Series editor, Setchi, Rossi, editor, Liu, Ying, editor, and Theobald, Peter, editor
- Published
- 2016
- Full Text
- View/download PDF
12. Neuron-Glia Interactions Studied with In Vitro Co-Cultures
- Author
-
Mancino, S., primary, Serafini, M. M., additional, and Viviani, Barbara, additional
- Published
- 2019
- Full Text
- View/download PDF
13. Morphological and immunophenotypical changes of human bone marrow adipocytes in marrow metastasis and myelofibrosis.
- Author
-
Dello Spedale Venti, M, Palmisano, B, Donsante, S, Farinacci, G, Adotti, F, Coletta, I, Serafini, M, Corsi, A, Riminucci, M, Dello Spedale Venti M, Palmisano B, Donsante S, Farinacci G, Adotti F, Coletta I, Serafini M, Corsi A, Riminucci M, Dello Spedale Venti, M, Palmisano, B, Donsante, S, Farinacci, G, Adotti, F, Coletta, I, Serafini, M, Corsi, A, Riminucci, M, Dello Spedale Venti M, Palmisano B, Donsante S, Farinacci G, Adotti F, Coletta I, Serafini M, Corsi A, and Riminucci M
- Abstract
The bone marrow adipose tissue constitutes more than two-thirds of the bone marrow volume in adult life and is known to have unique metabolic and functional properties. In neoplastic disorders, bone marrow adipocytes (BMAds) contribute to create a favorable microenvironment to survival and proliferation of cancer cells. Many studies explored the molecular crosstalk between BMAds and neoplastic cells, predominantly in ex-vivo experimental systems or in animal models. However, little is known on the features of BMAds in the human neoplastic marrow. The aim of our study was to analyze the in situ changes in morphology and immunophenotype of BMAds in two different types of neoplastic marrow conditions. We selected a series of archival iliac crest and vertebral bone biopsies from patients with bone marrow metastasis (MET), patients with myeloproliferative neoplasia with grade-3 myelofibrosis (MPN-MF) and age-matched controls (CTR). We observed a significant reduction in the number of BMAds in MET and MPN-MF compared to CTR. Accordingly, in the same groups, we also detected a significant reduction in the mean cell diameter and area. Immunolocalization of different adipocyte markers showed that, compared to CTR, in both MET and MPN-MF the percentages of adiponectin- and phosphorylated hormone sensitive lipase-positive BMAds were significantly reduced and increased respectively. No statistically significant difference was found between MET and MPN-MF. Interestingly, in one MET sample, “remodeled” BMAds containing a large lipid vacuole and multiple, smaller and polarized lipid droplets were identified. In conclusion, our data show that in different types of marrow cancers, BMAds undergo significant quantitative and qualitative changes, which need to be further investigated in future studies.
- Published
- 2022
14. MPSI Manifestations and Treatment Outcome: Skeletal Focus
- Author
-
De Ponti, G, Donsante, S, Frigeni, M, Pievani, A, Corsi, A, Bernardo, M, Riminucci, M, Serafini, M, De Ponti G, Donsante S, Frigeni M, Pievani A, Corsi A, Bernardo ME, Riminucci M, Serafini M, De Ponti, G, Donsante, S, Frigeni, M, Pievani, A, Corsi, A, Bernardo, M, Riminucci, M, Serafini, M, De Ponti G, Donsante S, Frigeni M, Pievani A, Corsi A, Bernardo ME, Riminucci M, and Serafini M
- Abstract
Mucopolysaccharidosis type I (MPSI) (OMIM #252800) is an autosomal recessive disorder caused by pathogenic variants in the IDUA gene encoding for the lysosomal alpha-L-iduronidase enzyme. The deficiency of this enzyme causes systemic accumulation of glycosaminoglycans (GAGs). Although disease manifestations are typically not apparent at birth, they can present early in life, are progressive, and include a wide spectrum of phenotypic findings. Among these, the storage of GAGs within the lysosomes disrupts cell function and metabolism in the cartilage, thus impairing normal bone development and ossification. Skeletal manifestations of MPSI are often refractory to treatment and severely affect patients’ quality of life. This review discusses the pathological and molecular processes leading to impaired endochondral ossification in MPSI patients and the limitations of current therapeutic approaches. Understanding the underlying mechanisms responsible for the skeletal phenotype in MPSI patients is crucial, as it could lead to the development of new therapeutic strategies targeting the skeletal abnormalities of MPSI in the early stages of the disease.
- Published
- 2022
15. CD14 positive cells accelerate hematopoietic stem cell engraftment
- Author
-
Pievani, A, Granata, V, Desantis, G, Antolini, L, Ornaghi, S, Galleu, A, Biondi, A, Gentner, B, Dazzi, F, Serafini, M, Pievani A., Granata V., Desantis G., Antolini L., Ornaghi S., Galleu A., Biondi A., Gentner B., Dazzi F., Serafini M., Pievani, A, Granata, V, Desantis, G, Antolini, L, Ornaghi, S, Galleu, A, Biondi, A, Gentner, B, Dazzi, F, Serafini, M, Pievani A., Granata V., Desantis G., Antolini L., Ornaghi S., Galleu A., Biondi A., Gentner B., Dazzi F., and Serafini M.
- Abstract
The improvement of hematopoietic stem and progenitor cell (HSPC) engraftment remains a high-priority goal when limited cell doses are available, such as in cord blood (CB) transplantation and HSC gene therapy. We observed that monocytes are highly effective at improving the engraftment of both CB-CD34+ and lentivirus-transfected CD34+ cells in a xenogeneic model of HSC transplantation. Moreover, monocytes, in particular the CD14+CD16− classical subset, in co-culture systems increase survival and stemness of CB-CD34+ cells. Both soluble factors and direct-cell contact interactions, such as JAG/NOTCH and COX-2/PGE2 pathways, are critically involved in the HSC-monocyte crosstalk. Our results indicate that the infusion of monocytes improves engraftment when cell dose is a limiting factor.
- Published
- 2022
16. Catch me if you can: how AML and its niche escape immunotherapy
- Author
-
Tettamanti, S, Pievani, A, Biondi, A, Dotti, G, Serafini, M, Tettamanti S., Pievani A., Biondi A., Dotti G., Serafini M., Tettamanti, S, Pievani, A, Biondi, A, Dotti, G, Serafini, M, Tettamanti S., Pievani A., Biondi A., Dotti G., and Serafini M.
- Abstract
In spite of the remarkable progress in basic and preclinical studies of acute myeloid leukemia (AML), the five-year survival rate of AML patients remains poor, highlighting the urgent need for novel and synergistic therapies. Over the past decade, increased attention has been focused on identifying suitable immunotherapeutic strategies for AML, and in particular on targeting leukemic cells and their progenitors. However, recent studies have also underlined the important contribution of the leukemic microenvironment in facilitating tumor escape mechanisms leading to disease recurrence. Here, we describe the immunological features of the AML niche, with particular attention to the crosstalk between the AML blasts and the cellular components of the altered tumor microenvironment (TME) and the mechanisms of immune escape that hamper the therapeutic effects of the most advanced treatments. Considering the AML complexity, immunotherapy approaches may benefit from a rational combination of complementary strategies aimed at preventing escape mechanisms without increasing toxicity.
- Published
- 2022
17. Effects of social and sensory deprivation in newborns: A lesson from the Covid-19 experience
- Author
-
Purpura, G, Fumagalli, S, Nacinovich, R, Riva, A, Ornaghi, S, Serafini, M, Nespoli, A, Purpura, Giulia, Fumagalli, Simona, Nacinovich, Renata, Riva, Anna, Ornaghi, Sara, Serafini, Marzia, Nespoli, Antonella, Purpura, G, Fumagalli, S, Nacinovich, R, Riva, A, Ornaghi, S, Serafini, M, Nespoli, A, Purpura, Giulia, Fumagalli, Simona, Nacinovich, Renata, Riva, Anna, Ornaghi, Sara, Serafini, Marzia, and Nespoli, Antonella
- Abstract
Background: Infancy is a complex period of human life, in which environmental experiences have a fundamental role for neurodevelopment. Although conditions of social and sensory deprivation are uncommon in high income countries, the Covid-19 pandemic abruptly modified this condition, by depriving people of their social stimuli of daily life. Aim: To understand the impact of this deprivation on infants' behaviour, we investigated the short-term effects of isolation and use of individual protective systems by mothers during the first two weeks of life. Methods: The study included 11 mother-infant dyads with mothers tested positive to SARS-CoV-2 at the time of delivery (Covid group) and 11 dyads with a SARS-CoV-2 negative mother as controls. Neurobehavioral, visual, and sensory processing assessments were performed from birth to 3 months of age. Results: Findings showed the effect of deprivation on some neurobehavioral abilities of infants in the Covid group; in addition, differences in sensory maturation trends were observed, although they tended to gradually decrease until disappearance at 3 months of age. Conclusion: These findings suggest the significant effects of early sensory and social deprivation during the first two weeks of life, but also provide several insights on the ability of the brain to restore its aptitudes by deleting or reducing the effects of early deprivation before the critical periods' closure.
- Published
- 2023
18. Acute Myeloid Leukemia (AML) immunotherapy: development of TIM3-CD33 targeted Dual CAR CIK cells for eradication of LSCs in the bone marrow niche
- Author
-
Arsuffi, C, Biondi, A, Tettamanti, S, Serafini, M, Arsuffi, C, Biondi, A, Tettamanti, S, and Serafini, M
- Published
- 2023
19. Selective homing of CAR-CIK cells to the bone marrow niche enhances control of the Acute Myeloid Leukemia burden
- Author
-
Biondi, M, Tettamanti, S, Galimberti, S, Cerina, B, Tomasoni, C, Piazza, R, Donsante, S, Bido, S, Maria Perriello, V, Broccoli, V, Doni, A, Dazzi, F, Mantovani, A, Dotti, G, Biondi, A, Pievani, A, Serafini, M, Marta Biondi, Sarah Tettamanti, Stefania Galimberti, Beatrice Cerina, Chiara Tomasoni, Rocco Piazza, Samantha Donsante, Simone Bido, Vincenzo Maria Perriello, Vania Broccoli, Andrea Doni, Francesco Dazzi, Alberto Mantovani, Gianpietro Dotti, Andrea Biondi, Alice Pievani, Marta Serafini, Biondi, M, Tettamanti, S, Galimberti, S, Cerina, B, Tomasoni, C, Piazza, R, Donsante, S, Bido, S, Maria Perriello, V, Broccoli, V, Doni, A, Dazzi, F, Mantovani, A, Dotti, G, Biondi, A, Pievani, A, Serafini, M, Marta Biondi, Sarah Tettamanti, Stefania Galimberti, Beatrice Cerina, Chiara Tomasoni, Rocco Piazza, Samantha Donsante, Simone Bido, Vincenzo Maria Perriello, Vania Broccoli, Andrea Doni, Francesco Dazzi, Alberto Mantovani, Gianpietro Dotti, Andrea Biondi, Alice Pievani, and Marta Serafini
- Abstract
Acute myeloid leukemia (AML) is a hematological malignancy derived from neoplastic myeloid progenitor cells characterized by abnormal clonal proliferation and differentiation. Although novel therapeutic strategies have recently been introduced, the prognosis of AML remains unsatisfactory. So far, the efficacy of chimeric antigen receptor (CAR)-T therapy in AML has been hampered by several factors including the poor accumulation of the blood-injected cells in the leukemia bone marrow (BM) niche, where chemotherapy-resistant leukemic stem cells reside. Thus, we hypothesized that overexpression of CXCR4, whose ligand CXCL12 is highly expressed by BM stromal cells within the niche, could improve T cell homing to the BM and consequently enhance their intimate contact with BM-resident AML cells facilitating disease eradication. Specifically, we engineered conventional CD33.CAR-cytokine induced killer cells (CIKs) with the wild-type CXCR4 and the variant CXCR4R334X, responsible for leukocyte sequestration in the BM of WHIM syndrome patients. Overexpression of both CXCR4wt and CXCR4mut in CD33.CAR-CIKs resulted in significant improvement of chemotaxis toward recombinant CXCL12 or BM stromal cell conditioned medium with no observed impairment of cytotoxic potential in vitro. Moreover, CXCR4-overexpressing CD33.CAR-CIKs showed enhanced in vivo BM homing, associated with a prolonged retention for the CXCR4R334X variant. However, only CD33.CAR-CIKs co-expressing CXCR4wt but not CXCR4mut exerted a more sustained in vivo antileukemic activity and extended animal survival, suggesting a non-canonical role for CXCR4 in modulating CAR-CIK functions independent of BM homing. Taken together, these data suggest that arming CAR-CIKs with CXCR4 may represent a promising strategy for increasing their therapeutic potential for AML.
- Published
- 2023
20. A Question of Frame: The Role of the Bone Marrow Stromal Niche in Myeloid Malignancies
- Author
-
Tomasoni, C, Pievani, A, Rambaldi, B, Biondi, A, Serafini, M, Tomasoni, Chiara, Pievani, Alice, Rambaldi, Benedetta, Biondi, Andrea, Serafini, Marta, Tomasoni, C, Pievani, A, Rambaldi, B, Biondi, A, Serafini, M, Tomasoni, Chiara, Pievani, Alice, Rambaldi, Benedetta, Biondi, Andrea, and Serafini, Marta
- Abstract
Until a few years ago, the onset of acute myeloid leukemia (AML) was entirely ascribed to genetic lesions in hematopoietic stem cells. These mutations generate leukemic stem cells, which are known to be the main ones responsible for chemoresistance and relapse. However, in the last years, increasing evidence demonstrated that dynamic interplay between leukemic cells and bone marrow (BM) niche is of paramount relevance in the pathogenesis of myeloid malignancies, including AML. Specifically, BM stromal niche components, such as mesenchymal stromal cells (MSCs) and their osteoblastic cell derivatives, play a key role not only in supporting normal hematopoiesis but also in the manifestation and progression of myeloid malignancies. Here, we reviewed recent clinical and experimental findings about how genetic and functional alterations in MSCs and osteolineage progeny can contribute to leukemogenesis and how leukemic cells in turn generate a corrupted niche able to support myeloid neoplasms. Moreover, we discussed how the newest single-cell technologies may help dissect the interactions between BM stromal cells and malignant hematopoiesis. The deep comprehension of the tangled relationship between stroma and AML blasts and their modulation during disease progression may have a valuable impact on the development of new microenvironment-directed therapeutic strategies, potentially useful for a wide cohort of patients.
- Published
- 2023
21. IL3-zetakine combined with a CD33 costimulatory receptor as a Dual CAR approach for safer and selective targeting of AML
- Author
-
Perriello, V, Rotiroti, M, Pisani, I, Galimberti, S, Alberti, G, Pianigiani, G, Ciaurro, V, Marra, A, Sabino, M, Tini, V, Spinozzi, G, Mezzasoma, F, Morena, F, Martino, S, Salerno, D, Ashby, J, Wingham, B, Serafini, M, Martelli, M, Falini, B, Biondi, A, Tettamanti, S, Perriello, Vincenzo Maria, Rotiroti, Maria Caterina, Pisani, Ilaria, Galimberti, Stefania, Alberti, Gaia, Pianigiani, Giulia, Ciaurro, Valerio, Marra, Andrea, Sabino, Marcella, Tini, Valentina, Spinozzi, Giulio, Mezzasoma, Federica, Morena, Francesco, Martino, Sabata, Salerno, Domenico, Ashby, Julian François, Wingham, Brittany, Serafini, Marta, Martelli, Maria Paola, Falini, Brunangelo, Biondi, Andrea, Tettamanti, Sarah, Perriello, V, Rotiroti, M, Pisani, I, Galimberti, S, Alberti, G, Pianigiani, G, Ciaurro, V, Marra, A, Sabino, M, Tini, V, Spinozzi, G, Mezzasoma, F, Morena, F, Martino, S, Salerno, D, Ashby, J, Wingham, B, Serafini, M, Martelli, M, Falini, B, Biondi, A, Tettamanti, S, Perriello, Vincenzo Maria, Rotiroti, Maria Caterina, Pisani, Ilaria, Galimberti, Stefania, Alberti, Gaia, Pianigiani, Giulia, Ciaurro, Valerio, Marra, Andrea, Sabino, Marcella, Tini, Valentina, Spinozzi, Giulio, Mezzasoma, Federica, Morena, Francesco, Martino, Sabata, Salerno, Domenico, Ashby, Julian François, Wingham, Brittany, Serafini, Marta, Martelli, Maria Paola, Falini, Brunangelo, Biondi, Andrea, and Tettamanti, Sarah
- Abstract
Acute myeloid leukemia (AML) still represents an unmet clinical need for adult and pediatric patients. Adoptive cell therapy by chimeric antigen receptor (CAR)-engineered T cells demonstrated a high therapeutic potential, but further development is required to ensure a safe and durable disease remission in AML, especially in elderly patients. To date, translation of CAR T-cell therapy in AML is limited by the absence of an ideal tumor-specific antigen. CD123 and CD33 are the 2 most widely overexpressed leukemic stem cell biomarkers but their shared expression with endothelial and hematopoietic stem and progenitor cells increases the risk of undesired vascular and hematologic toxicities. To counteract this issue, we established a balanced dual-CAR strategy aimed at reducing off-target toxicities while retaining full functionality against AML. Cytokine-induced killer (CIK) cells, coexpressing a first-generation low affinity anti-CD123 interleukin-3–zetakine (IL-3z) and an anti-CD33 as costimulatory receptor without activation signaling domains (CD33.CCR), demonstrated a powerful antitumor efficacy against AML targets without any relevant toxicity on hematopoietic stem and progenitor cells and endothelial cells. The proposed optimized dual-CAR cytokine-induced killer cell strategy could offer the opportunity to unleash the potential of specifically targeting CD123+/CD33+ leukemic cells while minimizing toxicity against healthy cells.
- Published
- 2023
22. Overexpression of CXCR4 receptor on CD33.CAR-CIK cells enhances the control of the Acute Myeloid Leukemia burden
- Author
-
Biondi, M, Tettamanti, S, Galimberti, S, Cerina, B, Tomasoni, C, Dotti, G, Biondi, A, Pievani, A, Serafini, M, M. Biondi, S. Tettamanti, S. Galimberti, B. Cerina, C. Tomasoni, G. Dotti, A. Biondi, A. Pievani, M. Serafini, Biondi, M, Tettamanti, S, Galimberti, S, Cerina, B, Tomasoni, C, Dotti, G, Biondi, A, Pievani, A, Serafini, M, M. Biondi, S. Tettamanti, S. Galimberti, B. Cerina, C. Tomasoni, G. Dotti, A. Biondi, A. Pievani, and M. Serafini
- Published
- 2023
23. Isolation of single circulating trophoblasts from maternal circulation for noninvasive fetal copy number variant profiling
- Author
-
Doffini, A, Forcato, C, Mangano, C, Lattuada, D, Aversa, R, Maranta, C, Giovannone, E, Buson, G, Bolognesi, C, Maiocchi, R, Dori, M, Jamal, L, Ahmad, R, Yeo, G, Yeo, T, Saragozza, S, Silipigni, R, Serafini, M, Biondi, A, Perego, S, Vergani, P, Ferrazzi, E, Ricciardi-Castagnoli, P, Musci, T, Grati, F, Doffini, Anna, Forcato, Claudio, Mangano, Chiara, Lattuada, Debora, Aversa, Roberta, Maranta, Chiara, Giovannone, Emilia D, Buson, Genny, Bolognesi, Chiara, Maiocchi, Rebecca, Dori, Martina, Jamal, Liyana, Ahmad, Raidah B, Yeo, George S H, Yeo, Tai Wai, Saragozza, Silvia, Silipigni, Rosamaria, Serafini, Marta, Biondi, Andrea, Perego, Sofia, Vergani, Patrizia, Ferrazzi, Enrico, Ricciardi-Castagnoli, Paola, Musci, Thomas J, Grati, Francesca Romana, Doffini, A, Forcato, C, Mangano, C, Lattuada, D, Aversa, R, Maranta, C, Giovannone, E, Buson, G, Bolognesi, C, Maiocchi, R, Dori, M, Jamal, L, Ahmad, R, Yeo, G, Yeo, T, Saragozza, S, Silipigni, R, Serafini, M, Biondi, A, Perego, S, Vergani, P, Ferrazzi, E, Ricciardi-Castagnoli, P, Musci, T, Grati, F, Doffini, Anna, Forcato, Claudio, Mangano, Chiara, Lattuada, Debora, Aversa, Roberta, Maranta, Chiara, Giovannone, Emilia D, Buson, Genny, Bolognesi, Chiara, Maiocchi, Rebecca, Dori, Martina, Jamal, Liyana, Ahmad, Raidah B, Yeo, George S H, Yeo, Tai Wai, Saragozza, Silvia, Silipigni, Rosamaria, Serafini, Marta, Biondi, Andrea, Perego, Sofia, Vergani, Patrizia, Ferrazzi, Enrico, Ricciardi-Castagnoli, Paola, Musci, Thomas J, and Grati, Francesca Romana
- Abstract
Objective: To develop a multi-step workflow for the isolation of circulating extravillous trophoblasts (cEVTs) by describing the key steps enabling a semi-automated process, including a proprietary algorithm for fetal cell origin genetic confirmation and copy number variant (CNV) detection. Methods: Determination of the limit of detection (LoD) for submicroscopic CNV was performed by serial experiments with genomic DNA and single cells from Coriell cell line biobank with known imbalances of different sizes. A pregnancy population of 372 women was prospectively enrolled and blindly analyzed to evaluate the current workflow. Results: An LoD of 800 Kb was demonstrated with Coriell cell lines. This level of resolution was confirmed in the clinical cohort with the identification of a pathogenic CNV of 800 Kb, also detected by chromosomal microarray. The mean number of recovered cEVTs was 3.5 cells per sample with a significant reverse linear trend between gestational age and cEVT recovery rate and number of recovered cEVTs. In twin pregnanices, evaluation of zygosity, fetal sex and copy number profiling was performed in each individual cell. Conclusion: Our semi-automated methodology for the isolation and single-cell analysis of cEVTS supports the feasibility of a cell-based noninvasive prenatal test for fetal genomic profiling.
- Published
- 2023
24. An in vivo humanized model to study homing and sequestration of Plasmodium falciparum transmission stages in the bone marrow
- Author
-
Donsante, S., Siciliano, G., Ciardo, M., Palmisano, B., Messina, V., de Turris, V., Farinacci, G., Serafini, M., Silvestrini, F., Corsi, A., Riminucci, M., and Alano, P.
- Subjects
Microbiology (medical) ,ectopic ossicles ,bone marrow ,Infectious Diseases ,skeletal stem cells ,plasmodium falciparum ,bone marrow adipocytes ,gametocytes ,Immunology ,malaria ,organoids ,Microbiology - Abstract
IntroductionRecent evidence suggests that the bone marrow (BM) plays a key role in the diffusion of P. falciparum malaria by providing a “niche” for the maturation of the parasite gametocytes, responsible for human-to-mosquito transmission. Suitable humanized in vivo models to study the mechanisms of the interplay between the parasite and the human BM components are still missing.MethodsWe report a novel experimental system based on the infusion of immature P. falciparum gametocytes into immunocompromised mice carrying chimeric ectopic ossicles whose stromal and bone compartments derive from human osteoprogenitor cells.ResultsWe demonstrate that immature gametocytes home within minutes to the ossicles and reach the extravascular regions, where they are retained in contact with different human BM stromal cell types.DiscussionOur model represents a powerful tool to study BM function and the interplay essential for parasite transmission in P. falciparum malaria and can be extended to study other infections in which the human BM plays a role.
- Published
- 2023
- Full Text
- View/download PDF
25. Exploring the Role of CK+/CD45+ (Dual-Positive) Circulating Cells in Patients With Early Breast Cancer (EBC)
- Author
-
Nicolo, E., Munoz-Arcos, L., Serafini, M., Gerratana, L., Davis, A., Liu, H., Gradishar, W., Curigliano, G., Reduzzi, C., and Cristofanilli, M.
- Published
- 2023
- Full Text
- View/download PDF
26. Flavanols, proanthocyanidins and antioxidant activity changes during cocoa (Theobroma cacao L.) roasting as affected by temperature and time of processing
- Author
-
Ioannone, F., Di Mattia, C.D., De Gregorio, M., Sergi, M., Serafini, M., and Sacchetti, G.
- Published
- 2015
- Full Text
- View/download PDF
27. 232MO Transcriptomic and tumor microenvironment landscape of EBV related nasopharyngeal carcinoma in endemic and non-endemic areas
- Author
-
Licitra, L.F., primary, Serafini, M., additional, Pistore, F., additional, Canevari, S., additional, Resteghini, C., additional, Cavalieri, S., additional, Trama, A., additional, and De Cecco, L., additional
- Published
- 2022
- Full Text
- View/download PDF
28. Impact of COVID on Utilization of Ultra-Hypofractionation in Breast Cancer Treatment
- Author
-
McDougall, R.P., Shi, Y., Gabriel, J.R., Serafini, M., Robbins, J.R., Hsu, C.C., Morrison, C., and Jr, L. Wang
- Published
- 2024
- Full Text
- View/download PDF
29. Impact of COVID on Utilization of Hypofractionation in Node-Positive Breast Cancer Treatment
- Author
-
McDougall, R.P., Shi, Y., Gabriel, J.R., Serafini, M., Morrison, C., Robbins, J.R., Hsu, C.C., and Jr, L. Wang
- Published
- 2024
- Full Text
- View/download PDF
30. CXCR4-modified CD33.CAR-CIK with enhanced bone marrow homing in Acute Myeloid Leukemia
- Author
-
Biondi, M, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, Serafini, M, Biondi, M, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, and Serafini, M
- Subjects
CAR-T cell therapy ,Acute leukemia - Abstract
Chimeric Antigen Receptor (CAR) Cytokine-Induced Killer (CIK) cell therapy is an emerging treatment for acute myeloid leukemia (AML) although some implementations are required. Specifically, it appears crucial to improve CAR-CIK infiltration ability into the bone marrow (BM) niche to eradicate leukemia stem cells (LSCs) at their location. CAR-CIK ex vivo manipulation influences the expression of several chemokine receptors and may dampen the capacity of infused cells to migrate to the BM. The chemokine ligand 12 (CXCL12), produced by mesenchymal stromal cells (MSCs) within the niche, and its chemokine receptor 4 (CXCR4) modulate leukocytes trafficking to the BM. In AML, CXCL12 binds CXCR4 overexpressed on blasts, enhancing their homing in the niche. CXCR4 expression is consistently downregulated during the culture of CIKs. Therefore, combining the expression of CD33.CAR and CXCR4 might promote CAR-CIK homing to the BM and subsequent leukemia eradication, specifically of LSCs. Two bicistronic Sleeping Beauty transposon vectors were designed: CXCR4(IRES)CD33.CAR and CD33.CAR(2A)CXCR4. The monocistronic CD33.CAR was used as control. Phenotype and CAR-related in vitro effector functions were compared. Migration in vitro toward rhCXCL12 or MSC supernatants was tested using a transwell migration assay. CAR-CIKs in vivo BM homing ability was evaluated in sub-lethally irradiated NSG mice. CAR-CIKs engraftment was assessed in BM, blood, spleen, and lungs. We noticed that both CD33.CAR(2A)CXCR4-CIKs (n=22, P
- Published
- 2022
31. Human T cells engineered with a leukemia lipid-specific TCR enables donor-unrestricted recognition of CD1c-expressing leukemia
- Author
-
Consonni, M, Garavaglia, C, Grilli, A, de Lalla, C, Mancino, A, Mori, L, De Libero, G, Montagna, D, Casucci, M, Serafini, M, Bonini, C, Häussinger, D, Ciceri, F, Bernardi, M, Mastaglio, S, Bicciato, S, Dellabona, P, Casorati, G, Consonni M, Garavaglia C, Grilli A, de Lalla C, Mancino A, Mori L, De Libero G, Montagna D, Casucci M, Serafini M, Bonini C, Häussinger D, Ciceri F, Bernardi M, Mastaglio S, Bicciato S, Dellabona P, Casorati G, Consonni, M, Garavaglia, C, Grilli, A, de Lalla, C, Mancino, A, Mori, L, De Libero, G, Montagna, D, Casucci, M, Serafini, M, Bonini, C, Häussinger, D, Ciceri, F, Bernardi, M, Mastaglio, S, Bicciato, S, Dellabona, P, Casorati, G, Consonni M, Garavaglia C, Grilli A, de Lalla C, Mancino A, Mori L, De Libero G, Montagna D, Casucci M, Serafini M, Bonini C, Häussinger D, Ciceri F, Bernardi M, Mastaglio S, Bicciato S, Dellabona P, and Casorati G
- Abstract
Acute leukemia relapsing after chemotherapy plus allogeneic hematopoietic stem cell transplantation can be treated with donor-derived T cells, but this is hampered by the need for donor/recipient MHC-matching and often results in graft-versus-host disease, prompting the search for new donor-unrestricted strategies targeting malignant cells. Leukemia blasts express CD1c antigen-presenting molecules, which are identical in all individuals and expressed only by mature leukocytes, and are recognized by T cell clones specific for the CD1c-restricted leukemia-associated methyl-lysophosphatidic acid (mLPA) lipid antigen. Here, we show that human T cells engineered to express an mLPA-specific TCR, target diverse CD1c-expressing leukemia blasts in vitro and significantly delay the progression of three models of leukemia xenograft in NSG mice, an effect that is boosted by mLPA-cellular immunization. These results highlight a strategy to redirect T cells against leukemia via transfer of a lipid-specific TCR that could be used across MHC barriers with reduced risk of graft-versus-host disease.
- Published
- 2021
32. Harnessing Mesenchymal Stromal Cells for the Engineering of Human Hematopoietic Niches
- Author
-
Pievani, A, Savoldelli, R, Poelchen, J, Mattioli, E, Anselmi, G, Girardot, A, Utikal, J, Bourdely, P, Serafini, M, Guermonprez, P, Pievani A, Savoldelli R, Poelchen J, Mattioli E, Anselmi G, Girardot A, Utikal J, Bourdely P, Serafini M, Guermonprez P, Pievani, A, Savoldelli, R, Poelchen, J, Mattioli, E, Anselmi, G, Girardot, A, Utikal, J, Bourdely, P, Serafini, M, Guermonprez, P, Pievani A, Savoldelli R, Poelchen J, Mattioli E, Anselmi G, Girardot A, Utikal J, Bourdely P, Serafini M, and Guermonprez P
- Abstract
Tissue engineering opens multiple opportunities in regenerative medicine, drug testing, and modeling of the hematopoiesis in health and disease. Recapitulating the organization of physiological microenvironments supporting leukocyte development is essential to model faithfully the development of immune cells. Hematopoietic organs are shaped by spatially organized niches defined by multiple cellular contributions. A shared feature of immune niches is the presence of mesenchymal stromal cells endowed with unique roles in organizing niche development, maintenance, and function. Here, we review challenges and opportunities in harnessing stromal cells for the engineering of artificial immune niches and hematopoietic organoids recapitulating leukocyte ontogeny both in vitro and in vivo.
- Published
- 2021
33. From stem cells to bone-forming cells
- Author
-
Donsante, S, Palmisano, B, Serafini, M, Robey, P, Corsi, A, Riminucci, M, Donsante S, Palmisano B, Serafini M, Robey PG, Corsi A, Riminucci M., Donsante, S, Palmisano, B, Serafini, M, Robey, P, Corsi, A, Riminucci, M, Donsante S, Palmisano B, Serafini M, Robey PG, Corsi A, and Riminucci M.
- Abstract
Bone formation starts near the end of the embryonic stage of development and continues throughout life during bone modeling and growth, remodeling, and when needed, regeneration. Bone-forming cells, traditionally termed osteoblasts, produce, assemble, and control the mineralization of the type I collagen-enriched bone matrix while participating in the regulation of other cell processes, such as osteoclastogenesis, and metabolic activities, such as phosphate homeostasis. Osteoblasts are generated by different cohorts of skeletal stem cells that arise from different embryonic specifications, which operate in the pre-natal and/or adult skeleton under the control of multiple regulators. In this review, we briefly define the cellular identity and function of osteoblasts and discuss the main populations of osteoprogenitor cells identified to date. We also provide examples of long-known and recently recognized regulatory pathways and mechanisms involved in the specification of the osteogenic lineage, as assessed by studies on mice models and human genetic skeletal diseases.
- Published
- 2021
34. Heterogeneity of the bone marrow niche in patients with myeloproliferative neoplasms: ActivinA secretion by mesenchymal stromal cells correlates with the degree of marrow fibrosis
- Author
-
Rambaldi, B, Diral, E, Donsante, S, Di Marzo, N, Mottadelli, F, Cardinale, L, Dander, E, Isimbaldi, G, Pioltelli, P, Biondi, A, Riminucci, M, D'Amico, G, Elli, E, Pievani, A, Serafini, M, Rambaldi B., Diral E., Donsante S., Di Marzo N., Mottadelli F., Cardinale L., Dander E., Isimbaldi G., Pioltelli P., Biondi A., Riminucci M., D'Amico G., Elli E. M., Pievani A., Serafini M., Rambaldi, B, Diral, E, Donsante, S, Di Marzo, N, Mottadelli, F, Cardinale, L, Dander, E, Isimbaldi, G, Pioltelli, P, Biondi, A, Riminucci, M, D'Amico, G, Elli, E, Pievani, A, Serafini, M, Rambaldi B., Diral E., Donsante S., Di Marzo N., Mottadelli F., Cardinale L., Dander E., Isimbaldi G., Pioltelli P., Biondi A., Riminucci M., D'Amico G., Elli E. M., Pievani A., and Serafini M.
- Abstract
Mesenchymal stromal cells (MSCs) represent an essential component of the bone marrow (BM) niche and display disease-specific alterations in several myeloid malignancies. The aim of this work was to study possible MSC abnormalities in Philadelphia-negative myeloproliferative neoplasms (MPNs) in relationship to the degree of BM fibrosis. MSCs were isolated from BM of 6 healthy donors (HD) and of 23 MPN patients, classified in 3 groups according to the diagnosis and the grade of BM fibrosis: polycythemia vera and essential thrombocythemia (PV/ET), low fibrosis myelofibrosis (LF-MF), and high fibrosis MF (HF-MF). MSC cultures were established from 21 of 23 MPN patients. MPN-derived MSCs did not exhibit any functional impairment in their adipogenic/osteogenic/chondrogenic differentiation potential and displayed a phenotype similar to HD-derived MSCs but with a decreased expression of CD146. All MPN-MSC lines were negative for the patient-specific hematopoietic clone mutations (JAK2, MPL, CALR). MSCs derived from HF-MF patients displayed a reduced clonogenic potential and a lower growth kinetic compared to MSCs from HD, LF-MF, and PV/ET patients. mRNA levels of hematopoiesis regulatory molecules were unaffected in MSCs from HF-MF compared to HD. Finally, in vitro ActivinA secretion by MSCs was increased in HF-MF compared to LF-MF patients, in association with a lower hemoglobin value. Increased ActivinA immunolabeling on stromal cells and erythroid precursors was also observed in HF-MF BM biopsies. In conclusion, higher grade of BM fibrosis is associated with functional impairment of MSCs and the increased secretion of ActivinA may represent a suitable target for anemia treatment in MF patients.
- Published
- 2021
35. Dietary antioxidants, non-enzymatic antioxidant capacity and the risk of osteoarthritis in the Swedish National March Cohort
- Author
-
Veen, L, Hantikainen, E, Bellocco, R, Ye, W, Serafini, M, Ponzano, M, Grotta, A, Trolle Lagerros, Y, Veen L., Hantikainen E., Bellocco R., Ye W., Serafini M., Ponzano M., Grotta A., Trolle Lagerros Y., Veen, L, Hantikainen, E, Bellocco, R, Ye, W, Serafini, M, Ponzano, M, Grotta, A, Trolle Lagerros, Y, Veen L., Hantikainen E., Bellocco R., Ye W., Serafini M., Ponzano M., Grotta A., and Trolle Lagerros Y.
- Abstract
Purpose: Oxidative stress might play an important role in the development of osteoarthritis, but not much is known about the effect of antioxidants on osteoarthritis risk. We, therefore, aimed to investigate the effect of dietary vitamin C, E, beta-carotene, and non-enzymatic antioxidant capacity (NEAC), which measures overall antioxidant activity from the diet, on the risk of osteoarthritis. Methods: For this study 43,865 men and women from the Swedish National March Cohort (SNMC) were followed for up to 19 years. We computed dietary intake of vitamin C, E and beta-carotene using information from a Food Frequency Questionnaire (FFQ). To estimate dietary NEAC we combined the information from the FFQ with food item-specific antioxidant capacity values from an antioxidant food database. Cases of osteoarthritis were identified through the Swedish National Patient Registers. We categorized all exposure variables into sex-specific quartiles and used multivariable-adjusted Cox proportional hazards regression models to estimate hazard ratios (HRs) with 95% confidence intervals (95% CIs). Results: In total, we observed 5976 cases of OA during 469,148 person-years of follow-up. After adjusting for potential confounders, we did not find any association between vitamin C, beta-carotene and NEAC (p-values for trend > 0.5), but a positive association was found with higher dietary vitamin E intake (HR Q4 vs Q1: 1.11; 95% CI 1.02–1.21; p for trend = 0.01) and the risk of OA. Conclusion: Our findings do not provide evidence for dietary antioxidants to protect from the development of OA, and a higher dietary vitamin E intake might even increase the risk.
- Published
- 2021
36. Fludarabine as a cost-effective adjuvant to enhance engraftment of human normal and malignant hematopoiesis in immunodeficient mice
- Author
-
Pievani, A., Michelozzi, I. M., Rambaldi, B., Granata, V., Corsi, A., Dazzi, F., Biondi, A., and Serafini, M.
- Published
- 2018
- Full Text
- View/download PDF
37. Spatial and temporal adjustments allowing the coexistence among carnivores in Liguria (N-W Italy)
- Author
-
Torretta, E., Serafini, M., Puopolo, F., and Schenone, L.
- Published
- 2016
- Full Text
- View/download PDF
38. Use of bone physicochemical characterization and biochemical analyses in an experimental model: A novel approach to assess bone mass loss for osteoporosis bioassay
- Author
-
Lima, C. M., Serafini, M. R., Santos, G. P., Cardoso, J. C., Figueiredo, R. T., Santos, M. S., Melo, M. G. D., Silva, F. A. R., da Costa, L. P., Santos, A. F. C., Albuquerque-Júnior, R. L. C., Quintans-Júnior, L. J., and Araújo, A. A. S.
- Published
- 2016
- Full Text
- View/download PDF
39. Transposon-Based CAR T Cells in Acute Leukemias: Where are We Going?
- Author
-
Magnani, C, Tettamanti, S, Alberti, G, Pisani, I, Biondi, A, Serafini, M, Gaipa, G, Magnani C. F., Tettamanti S., Alberti G., Pisani I., Biondi A., Serafini M., Gaipa G., Magnani, C, Tettamanti, S, Alberti, G, Pisani, I, Biondi, A, Serafini, M, Gaipa, G, Magnani C. F., Tettamanti S., Alberti G., Pisani I., Biondi A., Serafini M., and Gaipa G.
- Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy has become a new therapeutic reality for refractory and relapsed leukemia patients and is also emerging as a potential therapeutic option in solid tumors. Viral vector-based CAR T-cells initially drove these successful efforts; however, high costs and cumbersome manufacturing processes have limited the widespread clinical implementation of CAR T-cell therapy. Here we will discuss the state of the art of the transposon-based gene transfer and its application in CAR T immunotherapy, specifically focusing on the Sleeping Beauty (SB) transposon system, as a valid cost-effective and safe option as compared to the viral vector-based systems. A general overview of SB transposon system applications will be provided, with an update of major developments, current clinical trials achievements and future perspectives exploiting SB for CAR T-cell engineering. After the first clinical successes achieved in the context of B-cell neoplasms, we are now facing a new era and it is paramount to advance gene transfer technology to fully exploit the potential of CAR T-cells towards next-generation immunotherapy.
- Published
- 2020
40. Neonatal combination therapy improves some of the clinical manifestations in the Mucopolysaccharidosis type I murine model
- Author
-
Santi, L, De Ponti, G, Dina, G, Pievani, A, Corsi, A, Riminucci, M, Khan, S, Sawamoto, K, Antolini, L, Gregori, S, Annoni, A, Biondi, A, Quattrini, A, Tomatsu, S, Serafini, M, Santi L., De Ponti G., Dina G., Pievani A., Corsi A., Riminucci M., Khan S., Sawamoto K., Antolini L., Gregori S., Annoni A., Biondi A., Quattrini A., Tomatsu S., Serafini M., Santi, L, De Ponti, G, Dina, G, Pievani, A, Corsi, A, Riminucci, M, Khan, S, Sawamoto, K, Antolini, L, Gregori, S, Annoni, A, Biondi, A, Quattrini, A, Tomatsu, S, Serafini, M, Santi L., De Ponti G., Dina G., Pievani A., Corsi A., Riminucci M., Khan S., Sawamoto K., Antolini L., Gregori S., Annoni A., Biondi A., Quattrini A., Tomatsu S., and Serafini M.
- Abstract
Mucopolysaccharidosis type I (MPS-I), a lysosomal storage disorder caused by a deficiency of alpha-L-iduronidase enzyme, results in the progressive accumulation of glycosaminoglycans and consequent multiorgan dysfunction. Despite the effectiveness of hematopoietic stem cell transplantation (HSCT) and enzyme replacement therapy (ERT) in correcting clinical manifestations related to visceral organs, complete improvement of musculoskeletal and neurocognitive defects remains an unmet challenge and provides an impact on patients' quality of life. We tested the therapeutic efficacy of combining HSCT and ERT in the neonatal period. Using a mouse model of MPS-I, we demonstrated that the combination therapy improved clinical manifestations in organs usually refractory to current treatment. Moreover, combination with HSCT prevented the production of anti-IDUA antibodies that negatively impact ERT efficacy. The added benefits of combining both treatments also resulted in a reduction of skeletal anomalies and a trend towards decreased neuroinflammation and metabolic abnormalities. As currently there are limited therapeutic options for MPS-I patients, our findings suggest that the combination of HSCT and ERT during the neonatal period may provide a further step forward in the treatment of this rare disease.
- Published
- 2020
41. Overexpression of CXCR4 Enhances the Efficacy of CAR-T Therapy for Acute Myeloid Leukemia
- Author
-
Biondi, M, Galimberti, S, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, Serafini, M, Marta Biondi, Stefania Galimberti, Beatrice Cerina, Chiara Tomasoni, Gianpietro Dotti, Sarah Tettamanti, Andrea Biondi, Alice Pievani, Marta Serafini, Biondi, M, Galimberti, S, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, Serafini, M, Marta Biondi, Stefania Galimberti, Beatrice Cerina, Chiara Tomasoni, Gianpietro Dotti, Sarah Tettamanti, Andrea Biondi, Alice Pievani, and Marta Serafini
- Published
- 2022
42. Improving Bone Marrow Homing and Persistence of CD33.CAR-CIK Cells By Overexpression of a Gain-of-Function Variant of CXCR4 to Increase Efficacy in Acute Myeloid Leukemia
- Author
-
Biondi, M, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, Serafini, M, M. Biondi, B. Cerina, C. Tomasoni, G. Dotti, S. Tettamanti, A. Biondi, A. Pievani, M. Serafini, Biondi, M, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, Serafini, M, M. Biondi, B. Cerina, C. Tomasoni, G. Dotti, S. Tettamanti, A. Biondi, A. Pievani, and M. Serafini
- Published
- 2022
43. Ingegnerizzazione di cellule CD33.CAR-CIK con una variante gain-of-function del recettore CXCR4 per aumentarne migrazione e persistenza nella nicchia midollare
- Author
-
Biondi, M, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, Serafini, M, M. Biondi, B. Cerina, C. Tomasoni, G. Dotti, S. Tettamanti, A. Biondi, A. Pievani, M. Serafini, Biondi, M, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, Serafini, M, M. Biondi, B. Cerina, C. Tomasoni, G. Dotti, S. Tettamanti, A. Biondi, A. Pievani, and M. Serafini
- Published
- 2022
44. Sodium Content in Cereal-Based Products Sold in Italy: How Far Are We from the Global Benchmarks?
- Author
-
Martini, Daniela, Strazzullo, Pasquale, Serafini, M., Porrini, Marisa, Pellegrini, N., Angelino, Donato, Martini, Daniela, Strazzullo, Pasquale, Serafini, M., Porrini, Marisa, Pellegrini, N., and Angelino, Donato
- Abstract
Reformulation of food products is one of the measures needed for reducing salt consumption. Accordingly, the World Health Organization (WHO) recently proposed global sodium benchmarks for different food categories to be used for setting national policies. Therefore, the sodium content of cereal-based products currently sold in Italy was compared with the WHO benchmarks, highlighting those categories primarily needing a reformulation. To this aim, the sodium content and several declarations (i.e., nutrition and health claims, organic or gluten free declaration) were retrieved from 2917 cereal-based products sold on the Italian market. All "minimally processed breakfast cereals" had a sodium content below the benchmark, while "flatbreads" and "leavened bread" had the highest percentage of items above the respective sodium benchmarks. Flatbreads and "crackers/savory biscuits" showed the highest median delta values from the respective benchmarks of 360 and 278 mg/100 g, respectively. Large variability in terms of percentage of products with sodium content above the benchmark was observed within the same categories, as well as among products with different declarations. A large number of food products currently sold on the Italian market have a sodium content above the benchmark. This result suggests the need to reformulate many food products currently on the market to achieve the WHO/United Nations (UN) objective of 30% global reduction in sodium intake by 2025
- Published
- 2022
45. Functional properties of edible insects: a systematic review.
- Author
-
D'Antonio, V., Battista, N., Sacchetti, G., Di Mattia, C., and Serafini, M.
- Subjects
LIPID metabolism ,GLUCOSE metabolism ,REGULATION of body weight ,ONLINE information services ,FUNCTIONAL foods ,NUTRITIONAL value ,INFLAMMATION ,BLOOD platelet aggregation ,SYSTEMATIC reviews ,DIET ,ANTIOXIDANTS ,OXIDATIVE stress ,FOOD ,INSECTS ,MEDLINE - Abstract
Consumption of edible insects has been widely suggested as an environmentally sustainable substitute for meat to reduce greenhouse gas emissions. However, the novel research field for edible insects relies on the content of bioactive ingredients and on the ability to induce a functional effect in humans. The goal of this manuscript is to review the available body of evidence on the properties of edible insects in modulating oxidative and inflammatory stress, platelet aggregation, lipid and glucose metabolism and weight control. A search for literature investigating the functional role of edible insects was carried out in the PubMed database using specific keywords. A total of 55 studies, meeting inclusion criteria after screening, were divided on the basis of the experimental approach: in vitro studies, cellular models/ ex vivo studies or in vivo studies. In the majority of the studies, insects demonstrated the ability to reduce oxidative stress, modulate antioxidant status, restore the impaired activity of antioxidant enzymes and reduce markers of oxidative damage. Edible insects displayed anti-inflammatory activity reducing cytokines and modulating specific transcription factors. Results from animal studies suggest that edible insects can modulate lipid and glucose metabolism. The limited number of studies focused on the assessment of anti-coagulation activity of edible insects makes it difficult to draw conclusions. More evidence from dietary intervention studies in humans is needed to support the promising evidence from in vitro and animal models about the functional role of edible insect consumption. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
46. A Clinically Translatable, Extensively Validated Immune-based Classification of Human Papillomavirus-Associated Head and Neck Cancer With Implications for Treatment Deintensification and Immunotherapy
- Author
-
Zeng, P., primary, Cecchini, M., additional, Barrett, J., additional, Shammas-Toma, M., additional, De Cecco, L., additional, Serafini, M., additional, Cavalieri, S., additional, Licitra, L., additional, Hoebers, F., additional, Brakenhoff, R., additional, Leemans, C., additional, Scheckenbach, K., additional, Poli, T., additional, Wang, X., additional, Liu, X., additional, Laxague, F., additional, Prisman, E., additional, Poh, C., additional, Bose, P., additional, Dort, J., additional, Shaikh, M., additional, Ryan, S., additional, Dawson, A., additional, Khan, M., additional, Howlett, C., additional, Stecho, W., additional, Plantinga, P., additional, da Silva, S., additional, Hier, M., additional, Khan, H., additional, MacNeil, D., additional, Mendez, A., additional, Yoo, J., additional, Fung, K., additional, Lang, P., additional, Winquist, E., additional, Palma, D., additional, Ziai, H., additional, Li, S., additional, Boutros, P., additional, Mymryk, J., additional, and Nichols, A., additional
- Published
- 2022
- Full Text
- View/download PDF
47. Is TRIZ an Ecodesign Method?
- Author
-
Russo, D., primary, Serafini, M., additional, and Rizzi, C., additional
- Published
- 2016
- Full Text
- View/download PDF
48. Development of a model for the prediction of mechanical properties for Al-Si-Mg castings
- Author
-
Ransenigo, C., Tocci, M., Viscardi, C., Serafini, M., and Pola, A.
- Subjects
MODEL ,ALUMINUM ,CASTING ,HEAT TREATMENT ,SIMULATION - Published
- 2022
49. Generation of CIK cells co-expressing CXCR4 and CD33.CAR with improved homing and antitumor activity for AML
- Author
-
Biondi, M, Dotti, G, Biondi, A, Tettamanti, S, Pievani, A, Serafini, M, Biondi, M, Dotti, G, Biondi, A, Tettamanti, S, Pievani, A, and Serafini, M
- Subjects
CAR T-cell therapy - Abstract
Chimeric antigen receptor (CAR) T-cell therapy for acute myeloid leukemia (AML) has been quite elusive due to tumor heterogeneity, lack of ideal target antigens and the role of the tumor microenvironment in leukemia protection and leukemia stem cells (LSCs) maintenance. Therefore, CAR design must be improved for an effective anti-AML activity. Besides selecting a good target antigen, it is crucial to improve CAR T-cells infiltration, to eradicate LSCs at their location within the bone marrow (BM) niche. The chemokine receptor 4 (CXCR4) and its ligand, CXC motif ligand 12 (CXCL12) are two fundamental factors orchestrating leukocytes trafficking to the niche. In AML, the CXCL12-CXCR4 axis is co-opted by leukemic cells. CXCL12, released by mesenchymal stromal cells (MSCs), interacts with CXCR4 overexpressed on blasts, boosting their homing in the BM, where they can find a protecting environment. We hypothesize that CXCR4 overexpression by Cytokine-Induced Killer cells (CIKs) combined with a CD33.CAR may improve their homing to the BM and subsequent leukemia eradication. We designed two bicistronic vectors: CXCR4(IRES)CD33.CAR and CD33.CAR(2A)CXCR4. CIKs were non-virally engineered with the Sleeping Beauty transposon system to express our constructs or the monocistronic CD33.CAR, used as control. We assessed the chemotactic activity of the bicistronic constructs employing the transwell migration assay, toward rhCXCL12 and human MSCs supernatants. Additionally, in vitro effector functions were compared. Considering that CXCR4 expression on CAR-CIKs is downregulated over culture, we induced it artificially. CXCR4(IRES)CD33.CAR-CIKs (n=8) expressed higher levels of CXCR4, but lower CD33.CAR expression compared with CD33.CAR-CIKs, while CD33.CAR(2A)CXCR4-CIKs (n=9) showed a significant co-expression of both proteins, as compared to control (P≤ 0.05). The analysis of T-cell markers and memory phenotype uncovered that CXCR4(IRES)CD33.CAR-CIKs (n=7) are enriched in CD8+ compartment and include a higher fraction of TEMRA contrary to CD33-CAR CIKs (P≤0.05), while CD33.CAR(2A)CXCR4-CIKs (n=8) phenotype is comparable to control. Chemotaxis assays toward rhCXCL12 confirmed that CXCR4(IRES)CD33.CAR-CIKs (n=7, P≤0.05) and CD33.CAR(2A)CXCR4-CIKs (n=7, P≤0.05) achieved a migration advantage over CD33.CAR-CIKs alone (n=11), with a mean percentage of migration of 58.5% and 68.8% respectively, compared to 40.5%. We were also able to observe a specific chemotactic response toward MSCs supernatants, as proved by the use of Plerixafor. Moreover, correlation analysis strengthened our observation, as higher CXCR4 expression resulted in increased migratory activity. Concerning CAR-related effector functions, CXCR4(IRES)CD33.CAR-CIKs and CD33.CAR(2A)CXCR4-CIKs displayed similar killing of the CD33+ KG1 cell line, compared to CD33.CAR-CIKs alone, with a mean lysis of 56.6% (E:T ratio 5:1, n=7) for CXCR4(IRES)CD33.CAR-CIKs, and of 66.9% (n=9) for CD33.CAR(2A)CXCR4-CIKs, compared to a mean lysis of 62.5% (n=12) of CD33.CAR-CIKs. The bicistronic constructs also maintained their capacity to produce IL-2 and IFN-y, and to proliferate after CD33 antigen exposure. CXCR4(IRES)CD33.CAR-CIKs displayed not significant but inferior effector responses as compared to control, due to lower CAR expression; CD33.CAR(2A)CXCR4-CIKs performed similarly to CD33.CAR CIKs alone. Compared to conventional CD33.CAR-CIKs, CXCR4-overexpressing CD33.CAR-CIKs exhibit enhanced migration capacity while retaining functional activity against CD33+ target cells. Considering these results, we believe that CD33.CAR(2A)CXCR4 is the best candidate for further in vivo homing and anti-leukemic tests.
- Published
- 2021
50. Cellule CD33.CAR-CIK ingegnerizzate per co-esprimere il recettore CXCR4 al fine di aumentare la capacità di migrazione all’interno della nicchia midollare nella leucemia mieloide acuta
- Author
-
Biondi, M, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, Serafini, M, Biondi, M, Cerina, B, Tomasoni, C, Dotti, G, Tettamanti, S, Biondi, A, Pievani, A, and Serafini, M
- Subjects
leucemia mieloide acuta - Abstract
La terapia con cellule CAR-CIK per la leucemia mieloide acuta (LMA) è limitata dalla ridotta infiltrazione delle cellule ingegnerizzate all’interno della nicchia midollare, rifugio delle cellule staminali leucemiche (LSC). Queste ultime, over-esprimendo il recettore CXCR4, sfruttano l’interazione con la chemochina CXCL12 rilasciata dalle cellule mesenchimali stromali (MSC) per annidarsi nel microambiente. L’espressione di CXCR4 si riduce drasticamente sulle cellule CIK durante la coltura. Conseguentemente, combinare il CD33.CAR con l’over-espressione di CXCR4 potrebbe promuovere la migrazione delle CAR-CIK nel midollo, contribuendo all’eradicazione della malattia. Abbiamo disegnato il vettore bicistronico non virale CD33.CAR(2A)CXCR4 utilizzando il sistema di trasposoni Sleeping Beauty. Abbiamo osservato che le CD33.CAR(2A)CXCR4-CIK (n=22) presentano un’elevata espressione del CAR e mantengono l’espressione di CXCR4 durante la coltura, mentre le CD33.CAR-CIK lo down-regolano progressivamente (n=22, P
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.