326 results on '"Roncarolo, M G"'
Search Results
102. New evidences of genotype-phenotype correlation in metachromatic leukodystrophy
- Author
-
Fumagalli, F., Biffi, A., Cesani, M., Del Carro, U., Baldoli, C., Gerevini, S., Amadio, S., Falautano, M., Canale, S., Comi, G., Roncarolo, M. G., and maria sessa
103. Gene therapy for primary immunodeficiency: State of the art and future perspective,La terapia genica per le immunodeficienze primitive: Stato dell'arte e prospettive future
- Author
-
Aiuti, A., Chiesa, R., Marktel, S., Anna Villa, and Roncarolo, M. G.
104. Longitudinal assessment of health-related quality of life and healthcare satisfaction after haematopoietic stem cell transplantation in children with thalassaemia
- Author
-
Caocci, G., Efficace, F., Ciotti, F., Chiesa, R., Roncarolo, M. G., Vacca, A., Piras, E., Raji, M. S. D., Gary Collins, Mandelli, F., Marktel, S., and La Nasa, G.
105. Phenotypic and functional analysis of T-cell precursors in the human fetal liver and thymus: CD7 expression in the early stages of T- and myeloid-cell development
- Author
-
Bárcena, A., Marcus Muench, Galy, A. H. M., Cupp, J., Roncarolo, M. G., Phillips, J. H., Spits, H., and Other departments
- Subjects
hemic and lymphatic diseases - Abstract
It has been proposed that the CD7 molecule is the first antigen expressed on the membrane of cells committed to the T-cell lineage during human fetal T-cell ontogeny. To further identify the pre-T cell subpopulation that migrates to the thymus early in ontogeny, we analyzed the phenotypic and functional characteristics of the fetal liver populations separated on the basis of CD7 expression. Three populations expressing different levels of CD7 were observed: CD7bright, CD7dull, and CD7-. A CD7bright population depleted of mature T, B, and myeloid cells (lineage negative, lin-) and mostly composed of CD56+ CD34- natural killer cells did not mature into T cells in a fetal thymic organ culture (FTOC) assay and was devoid of myeloid progenitors in a clonal colony-forming cell assay. In contrast, the CD7-/dull CD34+ lin- populations were capable of differentiating into phenotypically mature T cells after injection into FTOC and contained early myeloid progenitors. Here we phenotypically compared the fetal liver CD7 populations with the most immature fetal thymic subset that differentiated in the FTOC assay, namely the triple negative (TN, CD3-CD4-CD8-) thymocytes. Fetal TN lin- expressed high levels of CD34 marker and were further subdivided by their expression of CD1 antigen, because CD1- TN thymocytes express higher levels of CD34 antigen compared with CD1+ TN cells. CD1- lin -TN thymocytes are characterized by expressing high levels of CD2, CD7, and CD34 markers and dull levels of CD5, CD10, and CD28 molecules. We could not find fetal liver pre-T cells with a phenotype equivalent to that of TN thymocytes. Our data show that CD7 does not necessarily identify T-cell precursors during fetal T-cell development and strongly support the hypothesis that the acquisition of early T-cell markers as CD2, CD28, and CD5 molecules on the cell surface of T-cell progenitors takes place intrathymically
106. Human organ rejection in SCID-hu mice
- Author
-
Matthieu Rouleau, Antonenko, S., Carballidoperrig, N., Namikawa, R., and Roncarolo, M. G.
107. New evidences of genotype-phenotype correlations in metachromatic leukodystrophy
- Author
-
Biffi, A., Cesani, M., Del Carro, U., Fumagalli, F., Baldoli, C., Gerevini, S., Talautano, M., Canale, S., Comi, G., Roncarolo, M. G., and maria sessa
108. Clonal tracking of engineered human hematopoiesis through integration sites analysis
- Author
-
Scala, S., Pellin, D., Basso-Ricci, L., Dionisio, F., Baricordi, C., Leonardelli, L., Scaramuzza, S., Ferrua, F., Cicalese, M. P., Giannelli, S., Neduva, V., Dow, D. J., Roncarolo, M. G., Ciceri, F., Vicard, P., Wit, E., Di Serio, C., Bonini, C., Naldini, L., Aiuti, A., Biasco, L., and Stochastic Studies and Statistics
109. Phase I/II clinical trial of haematopoietic stem cell gene therapy for the treatment of metachromatic leukodystrophy
- Author
-
maria sessa, Biffi, A., Fumagalli, F., Lorioli, L., Plati, T., Baldoli, C., Del Carro, U., Gerevini, S., Rovelli, A., Comi, G., Roncarolo, M. G., and Naldini, L.
110. Cooperation between major histocompatibility complex mismatched mononuclear cells from a human chimera in the production of antigen-specific antibody.
- Author
-
Roncarolo, M G, primary, Touraine, J L, additional, and Banchereau, J, additional
- Published
- 1986
- Full Text
- View/download PDF
111. Percorso assistenziale nel bambino ADA SCID sottoposto a terapia genica con cellule staminali ematopoietiche: il ruolo dell'infermiere di ricerca.
- Author
-
Soliman, C., Casiraghi, M., Antonioli, G., Ciceri, F., Roncarolo, M. G., Callegaro, L., and Aiuti, A.
- Published
- 2013
112. T lymphocytes from human chimeras do recognize antigen in the context of allogeneic determinants of the major histocompatibility complex
- Author
-
Touraine, J.-L., Roncarolo, M.-G., Plotnicky, H., and Bachetta, R.
- Published
- 1993
- Full Text
- View/download PDF
113. The X-linked lymphoproliferative-disease gene product SAP regulates signals induced through the co-receptor SLAM.
- Author
-
Sayos, J., Wu, C., Morra, M., Wang, N., Zhang, X., Allen, D., van Schaik, S., Notarangelo, L., Geha, R., Roncarolo, M. G., Oettgen, H., De Vries, J. E., Aversa, G., and Terhorst, C.
- Subjects
- *
LYMPHOPROLIFERATIVE disorders , *LYMPHOCYTES , *T cells , *CELL proliferation , *X chromosome - Abstract
In addition to triggering the activation of B- or T-cell antigen receptors, the binding of a llgand to Its receptor at the cell surface can sometimes determine the physiological outcome of Interactions between antigen-presenting cells, Tand B lymphocytes. The protein SIAIW (also known as CDw150), which Is present on the surface of B and Tcells, forms such a receptor-ligand pair as it is a self-ligand. We now show that a T-cell-specific, SLAIW-associated protein (SAP), which contains an SH2 domain and a short tall, acts as an Inhibitor by blocking recruitment of the SH2-domaln-contalnlng signal-transduction molecule SHP-2 to a docking site in the SLAM cytoplasmic region. The gene encoding SAP maps to the same area of the X chromosome as the locus for X-llnked lymphoprollferatlve disease (XLP) and we found mutations in the SAP gene in three XLP patients. Absence of the inhibitor SAP in XLP patients affects T/B-cell interactions Induced by SIAIW, leading to an Inability to control B-cell proliferation caused by Epstein-Barr virus Infections. [ABSTRACT FROM AUTHOR]
- Published
- 2017
114. Gene therapy for Wiskott-Aldrich syndrome: History, new vectors, future directions
- Author
-
Francesca Ferrua, Francesco Marangoni, Alessandro Aiuti, Maria Grazia Roncarolo, Ferrua, F., Marangoni, F., Aiuti, A., and Roncarolo, M. G.
- Subjects
0301 basic medicine ,Male ,Transplantation Conditioning ,Allergy ,Wiskott–Aldrich syndrome ,Genetic enhancement ,Genetic Vectors ,Immunology ,Article ,Viral vector ,03 medical and health sciences ,0302 clinical medicine ,Gene therapy ,Immunology and Allergy ,Medicine ,Humans ,(GT) ,business.industry ,(LV) ,Wiskott-Aldrich syndrome ,lentiviral vector ,Genetic Therapy ,medicine.disease ,Hematopoietic Stem Cells ,Virology ,Wiskott-Aldrich Syndrome ,030104 developmental biology ,(RIC) ,Hematopoietic Stem/Progenitor Cells ,030220 oncology & carcinogenesis ,Reduced Intensity Conditioning ,(WAS) ,hematopoietic stem/progenitor cell ,(HSPC) ,business ,reduced-intensity conditioning - Abstract
Author(s): Ferrua, Francesca; Marangoni, Francesco; Aiuti, Alessandro; Roncarolo, Maria Grazia
- Published
- 2020
115. Ectopic FOXP3 Expression Preserves Primitive Features Of Human Hematopoietic Stem Cells While Impairing Functional T Cell Differentiation
- Author
-
Maria Grazia Roncarolo, Rosa Bacchetta, Laura Passerini, Fabio Russo, Luigi Naldini, F. R. Santoni de Sio, M. M. Valente, Santoni De Sio, F. R., Passerini, L., Valente, M. M., Russo, F., Naldini, L., Roncarolo, M. G., and Bacchetta, R.
- Subjects
0301 basic medicine ,Regulatory T cell ,Cellular differentiation ,T-Lymphocytes ,lcsh:Medicine ,Biology ,Article ,Immune tolerance ,03 medical and health sciences ,Mice ,medicine ,Animals ,Humans ,lcsh:Science ,Cells, Cultured ,Regulation of gene expression ,Multidisciplinary ,Animal ,lcsh:R ,FOXP3 ,Forkhead Transcription Factors ,Hematopoietic Stem Cell ,Cell Differentiation ,hemic and immune systems ,Anatomy ,Forkhead Transcription Factor ,IPEX syndrome ,medicine.disease ,Hematopoietic Stem Cells ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,T-Lymphocyte ,lcsh:Q ,Stem cell ,Human - Abstract
FOXP3 is the transcription factor ruling regulatory T cell function and maintenance of peripheral immune tolerance, and mutations in its coding gene causes IPEX autoimmune syndrome. FOXP3 is also a cell-cycle inhibitor and onco-suppressor in different cell types. In this work, we investigate the effect of ectopic FOXP3 expression on HSC differentiation and we challenged this approach as a possible HSC-based gene therapy for IPEX. FOXP3-expressing HSC showed reduced proliferation ability and increased maintenance of primitive markers in vitro in both liquid and OP9-ΔL1 co-cultures. When transplanted into immunodeficient mice, FOXP3-expressing HSC showed significantly enhanced engraftment ability. This was due to a pronounced increase in the frequency of repopulating cells, as assessed by extreme limiting dilution assay. Likely underlying the increased repopulating ability, FOXP3 expressing HSC showed significantly enhanced expression of genes controlling stemness features. However, peripheral T cells developed in the FOXP3-humanized mice were quantitatively reduced and hyporesponsive to cytokine and polyclonal stimulation. Our findings reveal unpredicted effects of FOXP3 in the biology of HSC and may provide new tools to manipulate primitive features in HSC for clinical applications. Moreover, they formally prove the need of preserving endogenous FOXP3 regulation for an HSC-based gene therapy approach for IPEX syndrome.
- Published
- 2017
116. Gene correction for SCID-X1 in long-term hematopoietic stem cells
- Author
-
Beruh Dejene, Sruthi Mantri, Mara Pavel-Dinu, Harry L. Malech, Ciaran M. Lee, Niraj Punjya, Matthew A. Inlay, Volker Wiebking, Camille Sindhu, Waracharee Srifa, Carmencita E. Nicolas, Kenneth I. Weinberg, Maria Grazia Roncarolo, Gang Bao, Nivedita Saxena, Suk See DeRavin, Eric J. Kildebeck, Matthew H. Porteus, Pavel-Dinu, M., Wiebking, V., Dejene, B. T., Srifa, W., Mantri, S., Nicolas, C. E., Lee, C., Bao, G., Kildebeck, E. J., Punjya, N., Sindhu, C., Inlay, M. A., Saxena, N., Deravin, S. S., Malech, H., Roncarolo, M. G., Weinberg, K. I., and Porteus, M. H.
- Subjects
Genome instability ,Male ,0301 basic medicine ,Time Factors ,medicine.medical_treatment ,CD34 ,Codon, Initiator ,General Physics and Astronomy ,Antigens, CD34 ,02 engineering and technology ,Hematopoietic stem cell transplantation ,X-Linked Combined Immunodeficiency Diseases ,Genome ,Mice ,0302 clinical medicine ,Genome editing ,Parvovirinae ,Transduction, Genetic ,lcsh:Science ,Gene Editing ,0303 health sciences ,education.field_of_study ,Multidisciplinary ,Hematopoietic Stem Cell Transplantation ,Exons ,Fetal Blood ,021001 nanoscience & nanotechnology ,Healthy Volunteers ,3. Good health ,Haematopoiesis ,030220 oncology & carcinogenesis ,Stem cell ,0210 nano-technology ,Interleukin Receptor Common gamma Subunit ,DNA, Complementary ,Science ,Genetic Vectors ,Primary Cell Culture ,Transplantation, Heterologous ,Population ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,medicine ,Animals ,Humans ,education ,Gene ,030304 developmental biology ,Severe combined immunodeficiency ,Transplantation Chimera ,General Chemistry ,medicine.disease ,Hematopoietic Stem Cells ,Transplantation ,030104 developmental biology ,Mutation ,Cancer research ,lcsh:Q ,CRISPR-Cas Systems - Abstract
Gene correction in human long-term hematopoietic stem cells (LT-HSCs) could be an effective therapy for monogenic diseases of the blood and immune system. Here we describe an approach for X-linked sSevere cCombined iImmunodeficiency (SCID-X1) using targeted integration of a cDNA into the endogenous start codon to functionally correct disease-causing mutations throughout the gene. Using a CRISPR-Cas9/AAV6 based strategy, we achieve up to 20% targeted integration frequencies in LT-HSCs. As measures of the lack of toxicity we observe no evidence of abnormal hematopoiesis following transplantation and no evidence of off-target mutations using a high-fidelity Cas9 as a ribonucleoprotein complex. We achieve high levels of targeting frequencies (median 45%) in CD34+ HSPCs from six SCID-X1 patients and demonstrate rescue of lymphopoietic defect in a patient derived HSPC population in vitro and in vivo. In sum, our study provides specificity, toxicity and efficacy data supportive of clinical development of genome editing to treat SCID-Xl., Gene correction in hematopoietic stem cells could be a powerful way to treat monogenic diseases of the blood and immune system. Here the authors develop a strategy using CRISPR-Cas9 and an aAdeno-Associated vVirus(AAV)-delivered IL2RG cDNA to correct X-linked sSevere Ccombined iImmunodeficiency (SCID-X1) with a high success rate.
- Published
- 2019
117. Lentiviral Gene Therapy in HSCs Restores Lineage-Specific Foxp3 Expression and Suppresses Autoimmunity in a Mouse Model of IPEX Syndrome
- Author
-
Jennifer Laborada, Roger P. Hollis, Katelyn E. Masiuk, Maria Grazia Roncarolo, Donald B. Kohn, Masiuk, K. E., Laborada, J., Roncarolo, M. G., Hollis, R. P., and Kohn, D. B.
- Subjects
Diarrhea ,Genetic enhancement ,Autoimmunity ,chemical and pharmacologic phenomena ,Tregs ,Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Viral vector ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genes, Reporter ,FoxP3 ,Genetics ,medicine ,IPEX ,Animals ,Humans ,Cell Lineage ,IL-2 receptor ,030304 developmental biology ,Autoimmune disease ,0303 health sciences ,autoimmunity ,Lentivirus ,FOXP3 ,hemic and immune systems ,Forkhead Transcription Factors ,Genetic Diseases, X-Linked ,Cell Biology ,Genetic Therapy ,IPEX syndrome ,Immune dysregulation ,medicine.disease ,Hematopoietic Stem Cells ,gene therapy ,Disease Models, Animal ,Diabetes Mellitus, Type 1 ,Immune System Diseases ,Cancer research ,Molecular Medicine ,030217 neurology & neurosurgery - Abstract
Summary Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a devastating autoimmune disease caused by mutations in FoxP3, a transcription factor required for the development and function of regulatory T cells (Treg cells). Allogeneic hematopoietic stem cell transplant (HSCT) can be curative, but suitable donors are often unavailable. Here, we demonstrate a strategy for autologous HSCT and gene therapy utilizing a lentiviral vector (LV) to restore FoxP3 expression under the control of endogenous human FOXP3 regulatory elements. Both murine transplant models and humanized mice engrafted with LV-modified HSCs show high levels of LV expression selective for CD4+CD25+FoxP3+ Treg cells. LV transduction of scurfy (FoxP3mut) HSCs restores development of functional FoxP3+ Treg cells that suppress T cell proliferation in vitro and rescue the scurfy autoimmune phenotype in vivo. These findings demonstrate preclinical efficacy for the treatment of IPEX patients by autologous HSC transplant and may provide valuable insights into new cell therapies for autoimmunity.
- Published
- 2019
118. Do human TH1 and TH2 CD4 + clones exist?
- Author
-
de Vries, J.E., de Waal Malefyt, R., Yssel, H., Roncarolo, M.-G., and Spits, H.
- Published
- 1991
- Full Text
- View/download PDF
119. Treatment with rapamycin can restore regulatory T-cell function in IPEX patients
- Author
-
Pier Alberto Testoni, Rosalia Curto, Federica Barzaghi, Olaf Neth, Maria Grazia Roncarolo, Alessandro Aiuti, Francesca Tucci, Claudia Sartirana, Laura Passerini, Rosa Bacchetta, Vito Lampasona, Graziano Barera, Daniele Zama, Maria Pia Cicalese, Elena Bazzigaluppi, Manfred Hoenig, Ansgar Schulz, Alberto Mariani, Sven Olek, Luca Albarello, Ivana Rabbone, Emanuele Bosi, Markus G. Seidel, Passerini, L., Barzaghi, F., Curto, R., Sartirana, C., Barera, G., Tucci, F., Albarello, L., Mariani, A., Testoni, P. A., Bazzigaluppi, E., Bosi, E., Lampasona, V., Neth, O., Zama, D., Hoenig, M., Schulz, A., Seidel, M. G., Rabbone, I., Olek, S., Roncarolo, M. G., Cicalese, M. P., Aiuti, A., Bacchetta, R., Passerini L., Barzaghi F., Curto R., Sartirana C., Barera G., Tucci F., Albarello L., Mariani A., Testoni P.A., Bazzigaluppi E., Bosi E., Lampasona V., Neth O., Zama D., Hoenig M., Schulz A., Seidel M.G., Rabbone I., Olek S., Roncarolo M.G., Cicalese M.P., Aiuti A., and Bacchetta R.
- Subjects
Male ,0301 basic medicine ,regulatory T cell ,medicine.medical_treatment ,Hematopoietic stem cell transplantation ,Lymphocyte Activation ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,regulatory T cells ,Autoimmunity ,Immunosuppressive Agent ,0302 clinical medicine ,Cell Movement ,IPEX ,Immunology and Allergy ,Sirolimu ,Child ,Cells, Cultured ,autoimmunity ,FOXP3 ,Forkhead Transcription Factors ,Genetic Diseases, X-Linked ,hemic and immune systems ,EBI3 ,Regulatory T cells ,suppression ,Minor Histocompatibility Antigen ,medicine.anatomical_structure ,Immune System Diseases ,mTOR ,Immunosuppressive Agents ,Human ,Diarrhea ,Ebi3 ,Immune System Disease ,Regulatory T cell ,Immunology ,chemical and pharmacologic phenomena ,Minor Histocompatibility Antigens ,03 medical and health sciences ,TIGIT ,Glucocorticoid-Induced TNFR-Related Protein ,Immune Tolerance ,medicine ,Humans ,GITR ,Sirolimus ,rapamycin ,business.industry ,Interleukins ,Forkhead Transcription Factor ,Interleukin ,IPEX syndrome ,medicine.disease ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,Gene Expression Regulation ,CTLA-4 ,Mutation ,Cancer research ,business ,030215 immunology - Abstract
[Background] Immune-dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a lethal disease caused by mutations in a transcription factor critical for the function of thymus-derived regulatory T (Treg) cells (ie, FOXP3), resulting in impaired Treg function and autoimmunity. At present, hematopoietic stem cell transplantation is the therapy of choice for patients with IPEX syndrome. If not available, multiple immunosuppressive regimens have been used with poor disease-free survival at long-term follow-up. Rapamycin has been shown to suppress peripheral T cells while sparing Treg cells expressing wild-type FOXP3, thereby proving beneficial in the clinical setting of immune dysregulation. However, the mechanisms of immunosuppression selective to Treg cells in patients with IPEX syndrome are unclear., [Objective] We sought to determine the cellular and molecular basis of the clinical benefit observed under rapamycin treatment in 6 patients with IPEX syndrome with different FOXP3 mutations., [Methods] Phenotype and function of FOXP3-mutated Treg cells from rapamycin-treated patients with IPEX syndrome were tested by flow cytometry and in vitro suppression assays, and the gene expression profile of rapamycin-conditioned Treg cells by droplet-digital PCR., [Results] Clinical and histologic improvements in patients correlated with partially restored Treg function, independent of FOXP3 expression or Treg frequency. Expression of TNF-receptor-superfamily-member 18 (TNFRSF18, glucocorticoid-induced TNF-receptor–related) and EBV-induced-3 (EBI3, an IL-35 subunit) in patients’ Treg cells increased during treatment as compared with that of Treg cells from untreated healthy subjects. Furthermore inhibition of glucocorticoid-induced TNF-receptor–related and Ebi3 partially reverted in vitro suppression by in vivo rapamycin-conditioned Treg cells., [Conclusions] Rapamycin is able to affect Treg suppressive function via a FOXP3-independent mechanism, thus sustaining the clinical improvement observed in patients with IPEX syndrome under rapamycin treatment.
- Published
- 2020
120. Treatment of X-linked severe combined immunodeficiency by in utero transplantation of paternal bone marrow.
- Author
-
Flake, Alan W., Roncarolo, Maria-Grazia, Puck, Jennifer M., Almeida-Porada, Graza, Evans, Mark I., Johnson, Mark P., Abella, Estaban M., Harrison, Duane D., Zanjani, Esmail D., Flake, A W, Roncarolo, M G, Puck, J M, Almeida-Porada, G, Evans, M I, Johnson, M P, Abella, E M, Harrison, D D, and Zanjani, E D
- Subjects
- *
SEVERE combined immunodeficiency , *FETAL abnormalities , *BONE marrow transplantation , *GENETIC disorder treatment , *HEMATOPOIETIC stem cell transplantation , *THERAPEUTICS - Abstract
This article presents a case study concerning the fetus, now an 11-month-old boy child, of a 28-year-old woman known to carry a mutation found in X-linked severe combined immunodeficiency. The authors report successful treatment through the in-utero transplantation of paternal bone marrow enriched with hematopoietic cell progenitors.
- Published
- 1996
- Full Text
- View/download PDF
121. Demethylation analysis of the FOXP3 locus shows quantitative defects of regulatory T cells in IPEX-like syndrome
- Author
-
Yon Ho Choe, Eleonora Gambineri, Maria Grazia Roncarolo, S. Ciullini Mannurita, Tatjana I. Cornu, Federica Barzaghi, V. Discepolo, Rosa Bacchetta, G. Zuin, Massimiliano Cecconi, Sven Olek, Alessandro Ambrosi, Eun Suk Kang, Claudia Sartirana, Laura Passerini, Caterina Cancrini, A Ikinciogullari, J. Schmidtko, Rachele Ciccocioppo, S Corrente, Barzaghi, F, Passerini, L, Gambineri, E, Ciullini Mannurita, S, Cornu, T, Kang E., S, Choe, Yh, Cancrini, C, Corrente, S, Ciccocioppo, R, Cecconi, M, Zuin, G, Discepolo, V, Sartirana, C, Schmidtko, J, Ikinciogullari, A, Ambrosi, Alessandro, Roncarolo, MARIA GRAZIA, Olek, S, Bacchetta, R., Barzaghi, F., Passerini, L., Gambineri, E., Ciullini Mannurita, S., Cornu, T., Kang, E. S., Choe, Y. H., Cancrini, C., Corrente, S., Ciccocioppo, R., Cecconi, M., Zuin, G., Discepolo, V., Sartirana, C., Schmidtko, J., Ikinciogullari, A., Ambrosi, A., Roncarolo, M. G., and Olek, S.
- Subjects
Male ,CD3 Complex ,T-Lymphocytes ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Autoimmunity ,Cohort Studies ,Immunology and Allergy ,IL-2 receptor ,Treg cell-specific-demethylated-region (TSDR) ,Polyendocrinopathies, Autoimmune ,Child ,Immunologic Deficiency Syndrome ,Regulatory T (Treg) cells ,Forkhead box p3 (FOXP3) ,FOXP3 ,Genetic Diseases, X-Linked ,Forkhead Transcription Factors ,Autoimmune enteropathy ,hemic and immune systems ,Syndrome ,Flow Cytometry ,Regulatory ,Settore MED/02 ,Primary immunodeficiency (PID) ,Genetic Diseases ,Child, Preschool ,Female ,IPEX syndrome ,IPEX-like syndrome ,Human ,Adult ,Adolescent ,IPEX syndrome, IPEX-like syndrome, Regulatory T (Treg) cells, Forkhead box p3 (FOXP3), Treg cell-specific-demethylated-region (TSDR), Autoimmune enteropathy, Primary immunodeficiency (PID) ,CD3 ,Immunology ,chemical and pharmacologic phenomena ,Biology ,Article ,Young Adult ,medicine ,Humans ,Preschool ,Regulatory T (Treg) cell ,Immunologic Deficiency Syndromes ,Infant ,Forkhead Transcription Factor ,X-Linked ,DNA Methylation ,Immune dysregulation ,medicine.disease ,Polyendocrinopathies ,Primary immunodeficiency ,biology.protein ,Cohort Studie ,Autoimmune - Abstract
Immune dysregulation, Polyendocrinopathy, Enteropathy X-linked (IPEX) syndrome is a unique example of primary immunodeficiency characterized by autoimmune manifestations due to defective regulatory T (Treg) cells, in the presence of FOXP3 mutations. However, autoimmune symptoms phenotypically resembling IPEX often occur in the absence of detectable FOXP3 mutations. The cause of this “IPEX-like” syndrome presently remains unclear. To investigate whether a defect in Treg cells sustains the immunological dysregulation in IPEX-like patients, we measured the amount of peripheral Treg cells within the CD3+ T cells by analysing demethylation of the Treg cell-Specific-Demethylated-Region (TSDR) in the FOXP3 locus and demethylation of the T cell-Specific-Demethylated-Region (TLSDR) in the CD3 locus, highly specific markers for stable Treg cells and overall T cells, respectively. TSDR demethylation analysis, alone or normalized for the total T cells, showed that the amount of peripheral Treg cells in a cohort of IPEX-like patients was significantly reduced, as compared to both healthy subjects and unrelated disease controls. This reduction could not be displayed by flow cytometric analysis, showing highly variable percentages of FOXP3+ and CD25+FOXP3+ T cells. These data provide evidence that a quantitative defect of Treg cells could be considered a common biological hallmark of IPEX-like syndrome. Since Treg cell suppressive function was not impaired, we propose that this reduction per se could sustain autoimmunity., Highlights ► FOXP3 (TSDR) and CD3 (TLSDR) demethylation assays allow Treg cell quantification. ► TSDR/TLSDR ratio is altered in the majority of patients with immune dysregulation. ► IPEX-like syndromes share a quantitative but not functional defect of Treg cells.
- Published
- 2012
122. Wild-type FOXP3 is selectively active in CD4+CD25hi regulatory T cells of healthy female carriers of different FOXP3 mutations
- Author
-
Erica Valencic, Lucia Perroni, Ivana Türbachova, Rosa Bacchetta, Alberto Tommasini, Massimiliano Cecconi, Laura Passerini, Udo Baron, Maria Grazia Roncarolo, Sven Olek, Megan K. Levings, Giantonio Cazzola, Sara Di Nunzio, Silvia Vignola, Alicia N. McMurchy, Anne K. Junker, Di Nunzio, S, Cecconi, M, Passerini, L, Mcmurchy, An, Baron, U, Turbachova, I, Vignola, S, Valencic, E, Tommasini, A, Junker, A, Cazzola, G, Olek, S, Levings, Mk, Perroni, L, Roncarolo, MARIA GRAZIA, Bacchetta, R., Nunzio, S. D., Cecconi, M., Passerini, L., Mcmurchy, A. N., Baron, U., Turbachova, I., Vignola, S., Valencic, E., Tommasini, A., Junker, A., Cazzola, G., Olek, S., Levings, M. K., Perroni, L., and Roncarolo, M. G.
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,Heterozygote ,Adult, Autoimmune Diseases, CD4-Positive T-Lymphocytes, Case-Control Studies, Cell Differentiation, Female, Forkhead Transcription Factors, Genes ,X-Linked, Genetic Diseases ,X-Linked, Heterozygote, Humans, Male, Mutation, T-Lymphocyte Subsets, T-Lymphocytes ,Regulatory, X Chromosome Inactivation ,T-Lymphocytes ,Cellular differentiation ,Immunology ,chemical and pharmacologic phenomena ,Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Biochemistry ,X-inactivation ,Autoimmune Diseases ,Genes, X-Linked ,T-Lymphocyte Subsets ,X Chromosome Inactivation ,medicine ,Humans ,IL-2 receptor ,Allele ,X-Linked, Heterozygote, Humans, Male, Mutation, T-Lymphocyte Subsets, T-Lymphocyte ,X-Linked, Genetic Disease ,Mutation ,Wild type ,Adult, Autoimmune Diseases, CD4-Positive T-Lymphocytes, Case-Control Studies, Cell Differentiation, Female, Forkhead Transcription Factors, Gene ,FOXP3 ,Genetic Diseases, X-Linked ,Cell Differentiation ,Forkhead Transcription Factors ,hemic and immune systems ,Cell Biology ,Hematology ,T lymphocyte ,X-Linked ,Regulatory ,Genes ,Genetic Diseases ,Case-Control Studies ,Female - Abstract
Forkhead box P3 (FOXP3) is constitutively expressed by CD4+CD25hi regulatory T cells (nTregs). Mutations of FOXP3 cause a severe autoimmune syndrome known as immune dysregulation polyendocrinopathy enteropathy X-linked, in which nTregs are absent or dysfunctional. Whether FOXP3 is essential for both differentiation and function of human nTreg cells remains to be demonstrated. Because FOXP3 is an X-linked gene subject to X-chromosome inactivation (XCI), we studied 9 healthy female carriers of FOXP3 mutations to investigate the role of wild-type (WT) versus mutated FOXP3 in different cell subsets. Analysis of active WT versus mutated (mut)–FOXP3 allele distribution revealed a random pattern of XCI in peripheral blood lymphocytes and in naive and memory CD4+T cells, whereas nTregs expressed only the active WT-FOXP3. These data demonstrate that expression of WT-FOXP3 is indispensable for the presence of a normal nTreg compartment and suggest that FOXP3 is not necessary for effector T-cell differentiation in humans.
- Published
- 2009
123. The immune response to lentiviral-delivered transgene is modulated in vivo by transgene-expressing antigen-presenting cells but not by CD4+CD25+ regulatory T cells
- Author
-
Lucia Sergi-Sergi, Manuela Battaglia, Maria Grazia Roncarolo, Angelo Lombardo, Andrea Annoni, Luigi Naldini, Antonia Follenzi, Annoni, A, Battaglia, MARCO MARIA, Follenzi, A, Lombardo, A, SERGI SERGI, L, Naldini, Luigi, and Roncarolo, M. G.
- Subjects
CD4-Positive T-Lymphocytes ,Adoptive cell transfer ,Transgene ,Genetic Vectors ,Green Fluorescent Proteins ,Immunology ,Antigen-Presenting Cells ,Cytomegalovirus ,Enzyme-Linked Immunosorbent Assay ,Mice, Transgenic ,Mice, SCID ,Biology ,T-Lymphocytes, Regulatory ,Biochemistry ,Viral vector ,Interferon-gamma ,Mice ,Immune system ,Transduction, Genetic ,Animals ,Humans ,Transgenes ,IL-2 receptor ,Antigen-presenting cell ,Lentivirus ,Interleukin-2 Receptor alpha Subunit ,Cell Biology ,Hematology ,T lymphocyte ,Adoptive Transfer ,Cell biology ,Mice, Inbred C57BL ,Spleen ,CD8 - Abstract
Systemic delivery of lentiviral vector (LV) in immunocompetent mice leads to efficient in vivo cell transduction and expression of the encoded protein under the control of the ubiquitous promoter of human cytomegalovirus (CMV). However, antitransgene immune response results in clearance of transduced cells 4 weeks after injection. T regulatory cells (Tregs), which have been demonstrated to control immune responses in vivo, were tested for their ability to suppress antitransgene response leading to stable long-term expression. Adoptive transfer of natural CD4+CD25+ Tregs (nTregs) isolated from wild type (wt) mice or from transgene tolerant transgenic (tg) mice did not suppress the antitransgene immune response after LV delivery. These data demonstrate that neither increasing the endogenous pool of natural Tregs nor transferring nTregs selected in a transgene-expressing thymus can modulate the immune response and mediate sustained transgene expression. Conversely, adoptive transfer of antigen-presenting cells (APCs) isolated from transgene-tolerant tg mice efficiently reduced the immune response leading to stable LV-encoded protein expression in vivo. Reduction of CD8+ effector T cells was observed in LV-treated mice coinjected with transgene-expressing APCs compared with control mice. These data indicate that antitransgene immune response can be modulated by transgene-expressing APCs possibly through deletion of effector T cells.
- Published
- 2007
124. In vivo tracking of T cells in humans unveils decade-long survival and activity of genetically modified T memory stem cells
- Author
-
Biasco, Luca, Scala, Serena, Basso Ricci, Luca, Dionisio, Francesca, Baricordi, Cristina, Calabria, Andrea, Giannelli, Stefania, Cieri, Nicoletta, Barzaghi, Federica, Pajno, Roberta, Al-Mousa, Hamoud, Scarselli, Alessia, Cancrini, Caterina, Bordignon, Claudio, Roncarolo, Maria Grazia, Montini, Eugenio, BONINI, CHIARA, Aiuti, Alessandro, Biasco, Luca, Scala, Serena, Basso Ricci, Luca, Dionisio, Francesca, Baricordi, Cristina, Calabria, Andrea, Giannelli, Stefania, Cieri, Nicoletta, Barzaghi, Federica, Pajno, Roberta, Al-Mousa, Hamoud, Scarselli, Alessia, Cancrini, Caterina, Bordignon, Claudio, Roncarolo, Maria Grazia, Montini, Eugenio, Bonini, Chiara, Aiuti, Alessandro, Biasco, L, Scala, S, Basso, Ricci L, Dionisio, F, Baricordi, C, Calabria, A, Giannelli, S, Cieri, N, Barzaghi, F, Pajno, R, Al-Mousa, H, Scarselli, A, Cancrini, C, Bordignon, C, Roncarolo, M G, Montini, E, Bonini, C, and Aiuti, A
- Subjects
Time Factors ,T-Lymphocytes ,Genetic enhancement ,Cell ,Longitudinal Studie ,Biochemistry ,Longitudinal Studies ,Child ,Medicine (all) ,Hematology ,General Medicine ,Phenotype ,Tissue Donors ,Haematopoiesis ,Settore MED/02 ,medicine.anatomical_structure ,Cell Tracking ,Tetradecanoylphorbol Acetate ,Stem cell ,Genetic Engineering ,Human ,Adult ,Time Factor ,Cell Survival ,T cell ,Immunology ,Tissue Donor ,Biology ,Viral vector ,Clone Cell ,In vivo ,medicine ,Humans ,Severe combined immunodeficiency ,Hematopoietic Stem Cell ,Cell Biology ,Genetic Therapy ,Hematopoietic Stem Cells ,medicine.disease ,Lymphocyte Subsets ,In vitro ,Clone Cells ,T-Lymphocyte ,Lymphocyte Subset ,Cancer research ,Interleukin-2 ,T memory stem cells ,Bone marrow ,Immunologic Memory ,Ex vivo - Abstract
A deeper understanding of T lymphocytes survival and differentiation potential in humans is paramount for the development of effective gene/cell therapies based on T-cell engineering. We here performed a comprehensive study of T-cells dynamics and plasticity in humans by a unique combination of phenotypic/functional studies and high-throughput integration sites (IS) analyses. We analyzed samples from hematopoietic stem cells (HSC) (n=10) or mature lymphocytes (PBL) gene therapy (GT) (n=4) treated ADA (adenosine deaminase) deficient-SCID patients. For comparative analyses, we also collected data from pediatric (n=19) and adult (n=52) healthy donors (HD), and from bone marrow transplanted patients (BMT) with primary immunodeficiencies (n=10, 4 with ADA-SCID). We observed that vector-positive CD62L+/CD45RA+ putative T naïve cells were detectable 12 years after last infusion of gene-corrected lymphocytes in peripheral blood of PBL-GT patients that lack the support of transduced lymphocytes precursors. We then unveiled that the vast majority of these CD62L+/CD45RA+ cells (80.3%) in PBL-GT patients could be actually classified phenotypically (CD95, IL2Rβ and IL7Rα surface expression) and functionally (IFNγ production and aCD3/aCD28 in vitro differentiation) as active long-lasting T memory stem cells (Tscm). The peculiar Tscm frequency found in PBL-GT patients was most likely due to a combinatorial in vitro and in vivo effect. Indeed, by a series of in vitro assays, we showed that Tscm relative enrichment in CD45RA+CD62L+ compartment have occurred during the in vitro manipulation of T cells before infusion. Additionally, we found higher-then-normal Tscm contribution among CD45RA+/CD62L+ cells even in ADA-SCID patients receiving HSC-GT and BMT, suggesting a role of disease background on in vivo Tscm persistence. Analyzing our cohorts of healthy donors and treated individuals we were able to further correlate Tscm contribution in vivo with age, conditioning regimen, disease background, cell source, and long-term T-cell reconstitution. One unique aspect of our study consisted in the opportunity to track Tscm clonal dynamics in vivo in humans since each gene-corrected cell infused in our GT patients is univocally and permanently tagged by a retroviral integration site.To perform in vivo molecular tracing of individual T-cell clones we sorted T naïve, Tscm, central memory and effector memory subtypes. We then collected from these subpopulations, by LAM-PCR+Illumina-Miseq sequencing, 2.584.137 integration sites (IS) sequences mapped to 1.746 unique chromosomal positions, corresponding to 910 integrations from 5 HSC-GT patients in vivo, 79 integrations from 2 PBL-GT samples of transduced cell products prior to infusion and 754 integrations from 4 PBL-GT patients in vivo. Firstly, to establish a relationship between precursors and terminally differentiated T cells we searched for the presence of identical insertion sites detected in multiple T-cell subtypes, applying stringent analytical filters for cross-contaminations. Strikingly, the level of shared integrations in each subtype was directly correlated to its stage of differentiation with Tscm, isolated from PBL-GT patients, showing the highest proportion of integration sites shared with the other T-cell subsets. Importantly, the results of the same analysis performed on HSC-GT patients were outstandingly coherent with the progressive developmental model of memory T-cell differentiation. We then assessed the survival of individual Tscm clones by performing a longitudinal IS analysis of different T-cell subtypes isolated from 3 PBL-GT patients over a 2 to 5 years timeframe up to 12 years after last infusion. We were able to formally prove the persistence of individual Tscm by re-capturing identical IS tagging specific Tscm clones in two independent timepoints in a 5- years window. Importantly, the same IS were also detected in multiple T-cell subtypes, representing the best indirect evidence that these clones were endowed with long-term precursor activity. We also documented, by IS sequencing reads, the long-term polyclonal composition of each subtype and we did not observe enrichment for IS flanking proto-oncogenes. Overall, this study validates, for the first time in humans, the safe and functional decade-long survival of engineered Tscm, paving the way for their future application in clinical settings. Disclosures No relevant conflicts of interest to declare.
- Published
- 2015
125. Forkhead box protein 3 (FOXP3) mutations lead to increased TH17 cell numbers and regulatory T-cell instability
- Author
-
Federica Barzaghi, Sara Di Nunzio, Sven Olek, Mario Amendola, Rosa Bacchetta, Maria G. Roncarolo, Mario Abinun, Alberto Tommasini, Marco Cipolli, Massimiliano Cecconi, Laura Passerini, Luigi Naldini, Silvia Vignola, Sophie Hambleton, Luisa Guidi, Approches génétiques intégrées et nouvelles thérapies pour les maladies rares (INTEGRARE), Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Saclay-Généthon, Passerini, L, Olek, S, Di Nunzio, S, Barzaghi, F, Hambleton, S, Abinun, M, Tommasini, A, Vignola, S, Cipolli, M, Amendola, M, Naldini, L, Guidi, L, Cecconi, M, Roncarolo, M G, Bacchetta, R, Naldini, Luigi, Roncarolo, MARIA GRAZIA, and Bacchetta, R.
- Subjects
Male ,[SDV.BIO]Life Sciences [q-bio]/Biotechnology ,Regulatory T cell ,Immunology ,Cell ,Autoimmune/genetics ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Forkhead Transcription Factors/genetics ,Gene Expression Regulation/genetics ,Genetic Diseases ,X-Linked/genetics ,Humans ,Immunologic Deficiency Syndromes/genetics ,Polyendocrinopathies ,medicine ,Forkhead Box ,Immunology and Allergy ,Polyendocrinopathies, Autoimmune ,ComputingMilieux_MISCELLANEOUS ,Immunologic Deficiency Syndromes ,FOXP3 ,Forkhead Transcription Factors ,Genetic Diseases, X-Linked ,[SDV.MHEP.HEM]Life Sciences [q-bio]/Human health and pathology/Hematology ,Intestinal Diseases ,medicine.anatomical_structure ,Gene Expression Regulation ,Mutation (genetic algorithm) ,Mutation ,Cancer research - Abstract
International audience
- Published
- 2011
126. Manipulating Immune Tolerance with Micro-RNA Regulated Gene Therapy
- Author
-
Kevin Scott Goudy, Andrea eAnnoni, Luigi eNaldini, Maria Grazia eRoncarolo, Goudy, K. S., Annoni, A., Naldini, L., and Roncarolo, M. G.
- Subjects
Microbiology (medical) ,Mini Review ,Transgene ,Genetic enhancement ,lcsh:QR1-502 ,Biology ,Microbiology ,regulatory cells ,lcsh:Microbiology ,Immune tolerance ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,microRNA ,Antigen-presenting cell ,Tropism ,030304 developmental biology ,0303 health sciences ,tolerance ,Effector ,micro-RNA ,transgene ,Gene Therapy ,Cell biology ,Treg ,Liver ,030220 oncology & carcinogenesis ,Immunology ,Hepatocytes - Abstract
"The success of in vivo gene therapy greatly depends on the ability to control the immune response toward the therapeutic transgene. Over the last decade several vector-based and pharmacological approaches have been explored to control the immune-mediated clearance of transgene-expressing cells after viral delivery. One important outcome from these studies is the concept that expression of a transgene in tolerance-promoting organs, such as the liver and tolerogenic antigen-presenting cells, can help safeguard transgene-expressing cells from immune-mediated clearance. Gene therapists are now manipulating vectors to target naturally occurring tolerogenic properties of the body by: (i) incorporating tissue/cell specific promoters for targeted expression, (ii) using viral-capsid engineering to alter tropism and avoid pre-existing immunity, and (iii) regulating cell and activation dependent expression by including micro-RNA (miR) targets into expression cassettes. The combination of these three layers of vector regulation greatly enhances the targeting of tolerogenic cells and limits off-target expression of the transgene, which can lead to the induction of transgene-specific pathogenic effector T cells. In this review, we discuss the application of using miR transgene regulation to generate tolerogenic responses and speculate on possible mechanisms used by the liver to induce the transgene-specific regulatory T cells."
- Published
- 2011
- Full Text
- View/download PDF
127. Unpredictability of intravenous busulfan pharmacokinetics in children undergoing hematopoietic stem cell transplantation for advanced beta thalassemia: limited toxicity with a dose-adjustment policy
- Author
-
Erika Biral, Roberto Crocchiolo, Barbara Cappelli, Alessandro Aiuti, Sarah Marktel, Maria Grazia Roncarolo, Monica Broglia, Marco Fossati, Ilaria Frugnoli, Alessandra Biffi, Valentina Finizio, Costanza Evangelio, Anna Noè, Fabio Ciceri, Antonella Bartoli, Tito Roccia, Robert Chiesa, Chiesa, R, Cappelli, B, Crocchiolo, R, Frugnoli, I, Biral, E, Noe, A, Evangelio, C, Fossati, M, Roccia, T, Biffi, A, Finizio, V, Aiuti, Alessandro, Broglia, M, Bartoli, A, Ciceri, Fabio, RONCAROLO M., G, and AND MARKTEL, S.
- Subjects
Male ,medicine.medical_specialty ,Pediatrics ,Adolescent ,medicine.medical_treatment ,Thalassemia ,Hematopoietic stem cell transplantation ,Dose-Response Relationship ,Middle East ,Pharmacokinetics ,Conditioning regimen ,Recurrence ,Hemoglobinopathies ,Regimen-related toxicity ,Busulfan ,Child ,Child, Preschool ,Dose-Response Relationship, Drug ,Female ,Hematopoietic Stem Cell Transplantation ,Humans ,Immunosuppressive Agents ,Survival Analysis ,Treatment Outcome ,beta-Thalassemia ,medicine ,Preschool ,Intensive care medicine ,Survival analysis ,Transplantation ,business.industry ,Beta thalassemia ,Hematology ,medicine.disease ,Toxicity ,Drug ,business ,medicine.drug - Abstract
beta-thalassemia is a major health problem worldwide, and stem cell transplantation (SCT) is the only curative option. Oral Busulfan (Bu) based conditioning is widely used in this setting. Due to the variability of Bu systemic exposure, intravenous (i.v.) Bu has been proposed as a standard of care, with no need for drug monitoring and dose adjustment. Patients with beta-thalassemia from countries with limited resources might be at higher risk of erratic Bu metabolism because of liver dysfunction, severe iron overload, and specific ethnic/genetic features. We studied Bu pharmacokinetics in 53 children with advanced beta-thalassemia from Middle Eastern countries who underwent a total of 57 matched related donor SCTs. Forty-two percent of the children required dose adjustment because they did not achieve the therapeutic window after the first dose. With a Bu dose-adjustment policy, regimen-related toxicity was limited. At a median follow-up of 564 days, the probabilities of 2-year survival, current thalassemia-free survival, rejection, and treatment-related mortality were 96%, 88%, 21%, and 4%, respectively. Conditioning with i.v. Bu and dose adjustment is feasible and well tolerated, although recurrence of thalassemia remains an unsolved problem in children with advanced disease. beta-thalassemia is a major health problem worldwide, and stem cell transplantation (SCT) is the only curative option. Oral Busulfan (Bu) based conditioning is widely used in this setting. Due to the variability of Bu systemic exposure, intravenous (i.v.) Bu has been proposed as a standard of care, with no need for drug monitoring and dose adjustment. Patients with beta-thalassemia from countries with limited resources might be at higher risk of erratic Bu metabolism because of liver dysfunction, severe iron overload, and specific ethnic/genetic features. We studied Bu pharmacokinetics in 53 children with advanced beta-thalassemia from Middle Eastern countries who underwent a total of 57 matched related donor SCTs. Forty-two percent of the children required dose adjustment because they did not achieve the therapeutic window after the first dose. With a Bu dose-adjustment policy, regimen-related toxicity was limited. At a median follow-up of 564 days, the probabilities of 2-year survival, current thalassemia-free survival, rejection, and treatment-related mortality were 96%, 88%, 21%, and 4%, respectively. Conditioning with i.v. Bu and dose adjustment is feasible and well tolerated, although recurrence of thalassemia remains an unsolved problem in children with advanced disease. Abstract beta-thalassemia is a major health problem worldwide, and stem cell transplantation (SCT) is the only curative option. Oral Busulfan (Bu) based conditioning is widely used in this setting. Due to the variability of Bu systemic exposure, intravenous (i.v.) Bu has been proposed as a standard of care, with no need for drug monitoring and dose adjustment. Patients with beta-thalassemia from countries with limited resources might be at higher risk of erratic Bu metabolism because of liver dysfunction, severe iron overload, and specific ethnic/genetic features. We studied Bu pharmacokinetics in 53 children with advanced beta-thalassemia from Middle Eastern countries who underwent a total of 57 matched related donor SCTs. Forty-two percent of the children required dose adjustment because they did not achieve the therapeutic window after the first dose. With a Bu dose-adjustment policy, regimen-related toxicity was limited. At a median follow-up of 564 days, the probabilities of 2-year survival, current thalassemia-free survival, rejection, and treatment-related mortality were 96%, 88%, 21%, and 4%, respectively. Conditioning with i.v. Bu and dose adjustment is feasible and well tolerated, although recurrence of thalassemia remains an unsolved problem in children with advanced disease.
- Published
- 2009
128. The Quality of Life of Children and Adolescents with X-Linked Agammaglobulinemia
- Author
-
Soresina A., Nacinovich R., Bomba M., Cassani M., Molinaro A., Sciotto A., Martino S., Cardinale F., De Mattia D., Putti C., Dellepiane R.M., Felici L., Parrinello G., Neri F., Plebani A., Pierani P., DeMattia D., Martire B., Armenio L., Dammacco F., Ranieri G., Masi M., Miniaci A., Pession A., Rondelli R., Notarangelo L. D., Cao, Cossu F., Del Giacco S., Manconi P., Evangelista I., Magro S., Morgione S., STRISCIUGLIO, PIETRO, Anastasio E., Schillirò G., Paganelli R., Sticca M., Sperlì D., Carpino L., Bernini G., Azzari C., Maggi E., Romagnani S., Matucci A., Vultaggio A., Castagnola E., Gattorno M., Presta G., Civino A., Gambaretto G., Fasoli S., Salpietro C., Pietrogrande M.C., DellePiane R.M., Panisi C., Cambiaghi G., Pietrogrande M., Roncarolo M.G., Aiuti A., Masera G., Biondi A., Sala A., PIGNATA, CLAUDIO, Poggi V., Menna G., Di Nardo R., D'Apuzzo A., Pelliccia A., Correra A., Marone G., SPADARO, GIUSEPPE, Carli M., Zanesco L., Basso G., Putti M.C., Semenzato G., Agostini C., Amato G.M., Aricò M., Trizzino A., Izzi G., Bertolini P., Locatelli F., Zecca M., Rondini G., Marseglia G.L., Maccario R., Bossi G., Favre C., Consolini R., Vecchi V., Sacchini P., Rinaldi G., Ugazio A.G., Rossi P., Livadiotti S., Cancrini C., Finocchi A., Stabile A., Duse M., Iacobini M., Quinti I., Moschese V., Cecere F., Morgese G., Acquaviva A., De Zan G., Strafella S., Tamaro P., Rabusin M., Tovo P.A., Nespoli L., Marinoni M., Porcellini A., Cazzola G.A., Annarosa, Soresina, Renata, Nacinovich, Monica, Bomba, Morena, Cassani, Molinaro, Anna, Antonella, Sciotto, Silvana, Martino, Fabio, Cardinale, Domenico, Mattia, Caterina, Putti, Rosa Maria, Dellepiane, Leonardo, Felici, Giovanni, Parrinello, Francesca, Neri, Alessandro, Plebani, Soresina, A, Nacinovich, R, Bomba, M, Cassani, M, Molinaro, A, Sciotto, A, Martino, S, Cardinale, F, De Mattia, D, Putti, C, Dellepiane, R, Felici, L, Parrinello, G, Neri, F, Plebani, A, Soresina, A., Nacinovich, R., Bomba, M., Cassani, M., Molinaro, A., Sciotto, A., Martino, S., Cardinale, F., De Mattia, D., Putti, C., Dellepiane, R. M., Felici, L., Parrinello, G., Neri, F., Plebani, A., Pierani, P., Demattia, D., Martire, B., Armenio, L., Dammacco, F., Ranieri, G., Masi, M., Miniaci, A., Pession, A., Rondelli, R., Notarangelo, L. D., Cao, Cossu, F., Del Giacco, S., Manconi, P., Evangelista, I., Magro, S., Morgione, S., Strisciuglio, Pietro, Anastasio, E., Schillirò, G., Paganelli, R., Sticca, M., Sperlì, D., Carpino, L., Bernini, G., Azzari, C., Maggi, E., Romagnani, S., Matucci, A., Vultaggio, A., Castagnola, E., Gattorno, M., Presta, G., Civino, A., Gambaretto, G., Fasoli, S., Salpietro, C., Pietrogrande, M. C., Panisi, C., Cambiaghi, G., Pietrogrande, M., Roncarolo, M. G., Aiuti, A., Masera, G., Biondi, A., Sala, A., Pignata, Claudio, Poggi, V., Menna, G., Di Nardo, R., D'Apuzzo, A., Pelliccia, A., Correra, A., Marone, G., Spadaro, Giuseppe, Carli, M., Zanesco, L., Basso, G., Putti, M. C., Semenzato, G., Agostini, C., Amato, G. M., Aricò, M., Trizzino, A., Izzi, G., Bertolini, P., Locatelli, F., Zecca, M., Rondini, G., Marseglia, G. L., Maccario, R., Bossi, G., Favre, C., Consolini, R., Vecchi, V., Sacchini, P., Rinaldi, G., Ugazio, A. G., Rossi, P., Livadiotti, S., Cancrini, C., Finocchi, A., Stabile, A., Duse, M., Iacobini, M., Quinti, I., Moschese, V., Cecere, F., Morgese, G., Acquaviva, A., De Zan, G., Strafella, S., Tamaro, P., Rabusin, M., Tovo, P. A., Nespoli, L., Marinoni, M., Porcellini, A., and Cazzola, G. A.
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,x-linked agammaglobulinemia ,Activities of daily living ,Adolescent ,X-linked agammaglobulinemia ,Health Status ,Immunology ,pedsql 4.0 generic core scale ,Quality of life ,children ,Agammaglobulinemia ,Surveys and Questionnaires ,Activities of Daily Living ,health-related quality of life ,parents ,medicine ,Humans ,Immunology and Allergy ,PedsQL 4.0 Generic Core Scale ,Child ,Settore MED/38 - Pediatria Generale e Specialistica ,Health related quality of life ,quality of live ,business.industry ,Immunoglobulins, Intravenous ,Genetic Diseases, X-Linked ,medicine.disease ,Socioeconomic Factors ,Child, Preschool ,Mutation ,Quality of Life ,Female ,X-linked agammaglobulinemia - children - parents - health-related quality of life - PedsQL 4.0 Generic Core Scale ,business - Abstract
Introduction: The health-related quality of life in X-linked agammaglobulinemia was investigated in 25 children and adolescents patients through the Italian version of Pediatric Quality of Life Inventory 4.0 Generic Core Scale for patients aged less then 18 years, comparing child perception to that of the parents and the physician's evaluation. The data were compared with the ones of 80 healthy controls and the literature data of a group of patients with rheumatic diseases. Discussion: The agammaglobulinemia subjects perceived a lower global quality of life than the healthy subjects, but significantly higher than the rheumatic diseases controls. The clinical relevance of health-related quality of life assessment in X-linked agammaglobulinemia pediatric patients is discussed. © 2008 Springer Science+Business Media, LLC.
- Published
- 2009
129. Efficient gene transfer into primitive hematopoietic progenitors using a bone marrow microenvironment cell line engineered to produce retroviral vectors
- Author
-
Js, Dando, Ficara F, Deola S, Mg, Roncarolo, Bordignon C, Alessandro AIUTI, Dando, J., Ficara, F., Deola, S., Roncarolo, M. G., Bordignon, Claudio, Aiuti, Alessandro, Dando, J, Ficara, F, Deola, S, Roncarolo, MARIA GRAZIA, and Aiuti, A.
- Subjects
Viral Structural Proteins ,Mice ,Retroviridae ,Transduction, Genetic ,Genetic Vectors ,Animals ,Humans ,Bone Marrow Cells ,Stromal Cells ,Fetal Blood ,Hematopoietic Stem Cells ,Cell Line - Abstract
Background and Objectives. Effective gene transfer into human hematopoietic stem/progenitor cells is a compromise between achieving high transduction efficiency and maintaining the desired biological characteristics of the target cell. The aim of our work was to exploit the stromal microenvironment to increase gene transfer and maintenance of hematopoietic progenitors. Design and Methods. The murine bone marrow stromal cell line MS-5, known to support primitive human progenitors, was modified into an amphotropic packaging cell, by the stable introduction of DNA coding for retroviral structural proteins, and a viral vector encoding a marker gene. The gene transfer efficiency of the recombinant virus was evaluated by flow cytometry, in vitro assays for committed (CFC) and primitive (LTC-CFC) progenitors, as well as a clonal assay for B and NK lymphoid progenitors. Results. The new packaging cell line (NEXUS) produced equivalent levels of virus as did the established GP+Am12 system, also under serum-free conditions. On average 30% of human mobilized peripheral blood CD34(+) cells were transduced by a single exposure to NEXUS supernatant, representing a three-fold increase over GP+Am12-based technology. Gene transfer into both committed and primitive progenitors increased on average two-fold using NEXUS retroviral supernatant. Furthermore, CD34(+)CD38(low) early progenitor cells purified from umbilical cord blood were efficiently transduced with NEXUS retroviral vector and gave rise to a high frequency of marked B and NK lymphocytes. Interpretation and Conclusions. Our data show that that an established bone marrow stromal cell can be engineered to enhance the genetic modification of primitive hematopoietic and lymphoid progenitors using a clinically relevant method.
- Published
- 2004
130. Pillars Article: The X-Linked Lymphoproliferative Disease Gene Product SAP Regulates Signals Induced through the Co-Receptor SLAM. Nature . 1998. 395: 462-469.
- Author
-
Sayos J, Wu C, Morra M, Wang N, Zhang X, Allen D, van Schaik S, Notarangelo L, Geha R, Roncarolo MG, Oettgen H, De Vries JE, Aversa G, and Terhorst C
- Subjects
- Adult, Animals, Cloning, Molecular, History, 20th Century, Humans, Jurkat Cells, Lymphocyte Activation, Lymphoproliferative Disorders immunology, Male, Mice, Mice, Inbred C57BL, Sequence Alignment, Signaling Lymphocytic Activation Molecule Associated Protein metabolism, Young Adult, Allergy and Immunology history, Lymphoproliferative Disorders genetics, Mutation genetics, Signaling Lymphocytic Activation Molecule Associated Protein genetics, Signaling Lymphocytic Activation Molecule Family Member 1 metabolism, T-Lymphocytes immunology
- Published
- 2017
131. Abnormalities of acid-base balance and predisposition to metabolic acidosis in Metachromatic Leukodystrophy patients.
- Author
-
Lorioli L, Cicalese MP, Silvani P, Assanelli A, Salvo I, Mandelli A, Fumagalli F, Fiori R, Ciceri F, Aiuti A, Sessa M, Roncarolo MG, Lanzani C, and Biffi A
- Subjects
- Acid-Base Equilibrium, Acid-Base Imbalance, Acidosis blood, Acidosis prevention & control, Acidosis urine, Child, Child, Preschool, Cohort Studies, Follow-Up Studies, Genotype, Humans, Infant, Retrospective Studies, Time Factors, Acidosis complications, Leukodystrophy, Metachromatic complications, Leukodystrophy, Metachromatic metabolism, Monitoring, Physiologic
- Abstract
Metachromatic Leukodystrophy (MLD; MIM# 250100) is a rare inherited lysosomal storage disorder caused by the deficiency of Arylsulfatase A (ARSA). The enzymatic defect results in the accumulation of the ARSA substrate that is particularly relevant in myelin forming cells and leads to progressive dysmyelination and dysfunction of the central and peripheral nervous system. Sulfatide accumulation has also been reported in various visceral organs, although little is known about the potential clinical consequences of such accumulation. Different forms of MLD-associated gallbladder disease have been described, and there is one reported case of an MLD patient presenting with functional consequences of sulfatide accumulation in the kidney. Here we describe a wide cohort of MLD patients in whom a tendency to sub-clinical metabolic acidosis was observed. Furthermore in some of them we report episodes of metabolic acidosis of different grades of severity developed in acute clinical conditions of various origin. Importantly, we finally show how a careful acid-base balance monitoring and prompt correction of imbalances might prevent severe consequences of acidosis., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
132. Demethylation analysis of the FOXP3 locus shows quantitative defects of regulatory T cells in IPEX-like syndrome.
- Author
-
Barzaghi F, Passerini L, Gambineri E, Ciullini Mannurita S, Cornu T, Kang ES, Choe YH, Cancrini C, Corrente S, Ciccocioppo R, Cecconi M, Zuin G, Discepolo V, Sartirana C, Schmidtko J, Ikinciogullari A, Ambrosi A, Roncarolo MG, Olek S, and Bacchetta R
- Subjects
- Adolescent, Adult, CD3 Complex immunology, CD3 Complex metabolism, Child, Child, Preschool, Cohort Studies, Female, Flow Cytometry, Genetic Diseases, X-Linked genetics, Genetic Diseases, X-Linked immunology, Humans, Immunologic Deficiency Syndromes genetics, Immunologic Deficiency Syndromes immunology, Infant, Male, Syndrome, Young Adult, DNA Methylation, Forkhead Transcription Factors genetics, Polyendocrinopathies, Autoimmune genetics, Polyendocrinopathies, Autoimmune immunology, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism
- Abstract
Immune dysregulation, Polyendocrinopathy, Enteropathy X-linked (IPEX) syndrome is a unique example of primary immunodeficiency characterized by autoimmune manifestations due to defective regulatory T (Treg) cells, in the presence of FOXP3 mutations. However, autoimmune symptoms phenotypically resembling IPEX often occur in the absence of detectable FOXP3 mutations. The cause of this "IPEX-like" syndrome presently remains unclear. To investigate whether a defect in Treg cells sustains the immunological dysregulation in IPEX-like patients, we measured the amount of peripheral Treg cells within the CD3(+) T cells by analysing demethylation of the Treg cell-Specific-Demethylated-Region (TSDR) in the FOXP3 locus and demethylation of the T cell-Specific-Demethylated-Region (TLSDR) in the CD3 locus, highly specific markers for stable Treg cells and overall T cells, respectively. TSDR demethylation analysis, alone or normalized for the total T cells, showed that the amount of peripheral Treg cells in a cohort of IPEX-like patients was significantly reduced, as compared to both healthy subjects and unrelated disease controls. This reduction could not be displayed by flow cytometric analysis, showing highly variable percentages of FOXP3(+) and CD25(+)FOXP3(+) T cells. These data provide evidence that a quantitative defect of Treg cells could be considered a common biological hallmark of IPEX-like syndrome. Since Treg cell suppressive function was not impaired, we propose that this reduction per se could sustain autoimmunity., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
133. [Gene therapy in pediatrics].
- Author
-
Aiuti A, Cappelli B, Biffi A, Marktel S, and Roncarolo MG
- Subjects
- Child, Humans, Genetic Therapy, Pediatrics methods
- Published
- 2009
134. Progress and prospects: gene therapy clinical trials (part 2).
- Author
-
Aiuti A, Bachoud-Lévi AC, Blesch A, Brenner MK, Cattaneo F, Chiocca EA, Gao G, High KA, Leen AM, Lemoine NR, McNeish IA, Meneguzzi G, Peschanski M, Roncarolo MG, Strayer DS, Tuszynski MH, Waxman DJ, and Wilson JM
- Subjects
- Clinical Trials as Topic, Gene Transfer Techniques adverse effects, Gene Transfer Techniques trends, Genetic Therapy methods, Genetic Vectors, Humans, Neoplasms therapy, Stem Cell Transplantation adverse effects, Stem Cell Transplantation trends, Genetic Therapy trends
- Abstract
This is the second part of a review summarizing progress and prospects in gene therapy clinical research. Twenty key diseases/strategies are succinctly described and commented on by leaders in the field. This part includes clinical trials for skin diseases, neurological disorders, HIV/AIDS, ornithine transcarbamylase deficiency, alpha(1)-antitrypsin deficiency, haemophilia and cancer.
- Published
- 2007
- Full Text
- View/download PDF
135. Myelin deterioration in Twitcher mice: motor evoked potentials and magnetic resonance imaging as in vivo monitoring tools.
- Author
-
Dolcetta D, Amadio S, Guerrini U, Givogri MI, Perani L, Galbiati F, Sironi L, Del Carro U, Roncarolo MG, and Bongarzone E
- Subjects
- Animals, Axons pathology, Corpus Callosum pathology, Corpus Callosum physiopathology, Disease Models, Animal, Female, Male, Mice, Mice, Inbred C57BL, Mice, Neurologic Mutants, Nerve Degeneration pathology, Nerve Degeneration physiopathology, Sciatic Nerve physiology, Evoked Potentials, Motor, Leukodystrophy, Globoid Cell pathology, Leukodystrophy, Globoid Cell physiopathology, Magnetic Resonance Imaging, Myelin Sheath pathology
- Abstract
We have used magnetic resonance imaging (MRI) and motor evoked potentials (MEPs) for monitoring disease progression within the CNS of the Twitcher mouse, the murine model for globoid cell leukodystrophy (GLD). GLD is a lysosomal storage disorder, resulting from galactocerebrosidase deficiency, causing central and peripheral myelin impairment, leading to death, usually during early infancy. Neuroradiological, electrophysiological, and pathological parameters of myelin maturation were evaluated in Twitcher mice between postnatal days 20 and 45. Healthy controls showed a gradual-appearance MRI T2-weighted hypointensity of the corpus callosum (CC) starting at about P30 and ending at about P37, whereas MRI of age-matched Twitcher mice showed a complete loss of the CC-related MRI signal. MEPs allowed the functional assessment of myelin maturation within corticospinal motor pathways and showed a progressive deterioration of MEPs in Twitcher mice with increased central conduction time (CCT; 5.12 +/- 0.49 msec at P27 to 6.45 +/- 1.96 msec at P32), whereas physiological CCT shortening was found in healthy controls (3.01 +/- 0.81 msec at P27 to 2.5 +/- 0.27 msec at P32). These findings were not paralleled by traditional histological stainings. Optical observation of Bielchowsky and Luxol fast blue-PAS stainings showed mild axonal/myelin deterioration of the Twitcher brain within this time frame. Our results demonstrate that serial MRI and MEP readings are sensitive evaluation tools for in vivo monitoring of dysmyelination in Twitcher mice and underscore their potential use for longitudinal evaluation of the therapeutic impact of gene and cell therapies on these animals., (2005 Wiley-Liss, Inc.)
- Published
- 2005
- Full Text
- View/download PDF
136. Analysis of galactocerebrosidase activity in the mouse brain by a new histological staining method.
- Author
-
Dolcetta D, Perani L, Givogri MI, Galbiati F, Orlacchio A, Martino S, Roncarolo MG, and Bongarzone E
- Subjects
- Animals, Enzyme Activation, Galactosylceramidase deficiency, Histocytochemistry methods, Mice, Mice, Inbred C57BL, Mice, Neurologic Mutants, Neuroglia enzymology, Neurons enzymology, Brain enzymology, Galactosylceramidase analysis, Galactosylceramidase metabolism, Staining and Labeling methods
- Abstract
Gene therapy of galactocerebrosidase (GALC) deficient mice (Twitcher mutants) requires a fast and sensitive assay to detect transduced cells in vitro and in vivo. We have developed a new rapid histochemical method that specifically detects GALC activity in situ in neural cells using 5-Br-3Cl-beta-galactopiranoside (X-Gal) in the presence of taurodeoxycholic and oleic acids to enhance suspension of the substrate at low pH. Using this method, we observed robust X-Gal staining in diverse neuronal populations and interfascicular oligodendrocytes in sections from normal mouse brain. In contrast, sections of Twitcher brain did not show a specific staining pattern in neurons or glial cells. The availability of this new sensitive and rapid in situ detection assay is fundamental for the follow-up of Twitcher mice under gene or cellular therapies to correct central GALC deficiency., (Copyright 2004 Wiley-Liss, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
137. Human insulin production and amelioration of diabetes in mice by electrotransfer-enhanced plasmid DNA gene transfer to the skeletal muscle.
- Author
-
Martinenghi S, Cusella De Angelis G, Biressi S, Amadio S, Bifari F, Roncarolo MG, Bordignon C, and Falqui L
- Subjects
- Animals, Diabetes Mellitus, Experimental metabolism, Injections, Intramuscular, Male, Mice, Mice, SCID, DNA administration & dosage, Diabetes Mellitus, Experimental therapy, Electroporation, Genetic Therapy methods, Insulin genetics, Muscle, Skeletal metabolism
- Abstract
A first-line gene therapy for type 1 diabetes should be based on a safe procedure to engineer an accessible tissue for insulin release. We evaluated the ability of the skeletal muscle to release human insulin after electrotransfer (ET)-enhanced plasmid DNA injection in mice. A furin-cleavable proinsulin cDNA under the CMV or the MFG promoter was electrotransferred to immune-incompetent mice with STZ-induced severe diabetes. At 1 week, mature human insulin was detected in the serum of 17/20 mice. After an initial peak of 68.5 +/- 34.9 microU/ml, insulin was consistently detected at significant levels up to 6 weeks after gene transfer. Importantly, untreated diabetic animals died within 3 weeks after STZ, whereas treated mice survived up to 10 weeks. Fed blood glucose (BG) was reduced in correspondence with the insulin peak. Fasting BG was near-normalized when insulin levels were 12.9 +/- 5.3 (CMV group, 2 weeks) and 7.7 +/- 2.6 microU/ml (MFG group, 4 weeks), without frank hypoglycemia. These data indicate that ET-enhanced DNA injection in muscle leads to the release of biologically active insulin, with restoration of basal insulin levels, and lowering of fasting BG with increased survival in severe diabetes. Therefore the skeletal muscle can be considered as a platform for basal insulin secretion.
- Published
- 2002
- Full Text
- View/download PDF
138. A novel human packaging cell line with hematopoietic supportive capacity increases gene transfer into early hematopoietic progenitors.
- Author
-
Dando JS, Roncarolo MG, Bordignon C, and Aiuti A
- Subjects
- Antigens, CD34 analysis, Cell Line, Hematopoietic Stem Cells immunology, Humans, Hematopoietic Stem Cells cytology, Retroviridae genetics, Transfection, Virus Assembly
- Abstract
The hematopoietic stem/progenitor cell (HSPC) represents the ideal target for gene therapy of disorders of the hematopoietic system, but still faces problems related to ex vivo manipulation and gene transfer efficiency. We demonstrate that soluble factors from the human endothelial-like cell line ECV 304/T24 support the growth of human CD34(+) progenitor cells as primary human bone marrow stroma and increase the rate of gene transfer into progenitor cells up to 5-fold. ECV 304/T24 was used to generate split-function amphotropic packaging cell lines (named APEX) with the purpose of combining, in the same cells, hematopoietic support and gene transfer vehicle functions. The APEX cell lines were negative for the presence of replication-competent retroviruses and produced complement-resistant vector particles. When mobilized peripheral blood or umbilical cord blood CD34(+) cells were exposed once to APEX supernatants, the level of gene transfer was equivalent to that observed with GP + Am12, in spite of the lower titer of the APEX producers. More importantly, APEX supernatants gave rise reproducibly to a 2-fold increase in transduction of early progenitors (long-term culture-initiating cells), reaching on average 50% gene transfer. This novel packaging cell represents a significant advance in HSPC genetic modification technology, combining both a beneficial hematopoietic supportive effect and the gene transfer vector function in a human-based system.
- Published
- 2001
- Full Text
- View/download PDF
139. Type 1 T regulatory cells.
- Author
-
Roncarolo MG, Bacchetta R, Bordignon C, Narula S, and Levings MK
- Subjects
- Animals, Cell Differentiation, Clonal Anergy immunology, Clone Cells cytology, Clone Cells immunology, Clone Cells metabolism, Cytokines immunology, Humans, T-Lymphocyte Subsets cytology, Interleukin-10 immunology, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Transforming Growth Factor beta immunology
- Abstract
Suppression by T regulatory (Tr) cells is essential for induction of tolerance. Many types of Tr cells have been described in a number of systems, and their biology has been the subject of intensive investigation. Although many aspects of the mechanisms by which these cells exert their effects remain to be elucidated, it is well established that Tr cells suppress immune responses via cell-to-cell interactions and/or the production of interleukin (IL)-10 and transforming growth factor (TGF)-beta. Type-1 T regulatory (Tr1) cells are defined by their ability to produce high levels of IL-10 and TGF-beta. Tr1 cells specific for a variety of antigens arise in vivo, but may also differentiate from naive CD4+ T cells in the presence of IL-10 in vitro. Tr1 cells have a low proliferative capacity, which can be overcome by IL-15. Tr1 cells suppress naive and memory T helper type 1 or 2 responses via production of IL-10 and TGF-beta. Further characterisation of Tr1 cells at the molecular level will define their mechanisms of action and clarify their relationship with other subsets of Tr cells. The use of Tr1 cells to identify novel targets for the development of new therapeutic agents, and as a cellular therapy to modulate peripheral tolerance, can be foreseen.
- Published
- 2001
- Full Text
- View/download PDF
140. Human cd25(+)cd4(+) t regulatory cells suppress naive and memory T cell proliferation and can be expanded in vitro without loss of function.
- Author
-
Levings MK, Sangregorio R, and Roncarolo MG
- Subjects
- Abatacept, Antigens, CD, Antigens, Differentiation analysis, CTLA-4 Antigen, Cell Line, Cytokines biosynthesis, Humans, Immunophenotyping, Interleukin-10 biosynthesis, Isoantigens immunology, Transforming Growth Factor beta biosynthesis, CD4 Antigens analysis, Immunoconjugates, Lymphocyte Activation, Receptors, Interleukin-2 analysis, T-Lymphocytes immunology, T-Lymphocytes, Regulatory physiology
- Abstract
Active suppression by T regulatory (Tr) cells plays an important role in the downregulation of T cell responses to foreign and self-antigens. Mouse CD4(+) Tr cells that express CD25 possess remarkable suppressive activity in vitro and in autoimmune disease models in vivo. Thus far, the existence of a similar subset of CD25(+)CD4(+) Tr cells in humans has not been reported. Here we show that human CD25(+)CD4(+) Tr cells isolated from peripheral blood failed to proliferate and displayed reduced expression of CD40 ligand (CD40L), in response to T cell receptor-mediated polyclonal activation, but strongly upregulated cytotoxic T lymphocyte-associated antigen (CTLA)-4. Human CD25(+)CD4(+) Tr cells also did not proliferate in response to allogeneic antigen-presenting cells, but they produced interleukin (IL)-10, transforming growth factor (TGF)-beta, low levels of interferon (IFN)-gamma, and no IL-4 or IL-2. Importantly, CD25(+)CD4(+) Tr cells strongly inhibited the proliferative responses of both naive and memory CD4(+) T cells to alloantigens, but neither IL-10, TGF-beta, nor CTLA-4 seemed to be directly required for their suppressive effects. CD25(+)CD4(+) Tr cells could be expanded in vitro in the presence of IL-2 and allogeneic feeder cells and maintained their suppressive capacities. These findings that CD25(+)CD4(+) Tr cells with immunosuppressive effects can be isolated from peripheral blood and expanded in vitro without loss of function represent a major advance towards the therapeutic use of these cells in T cell-mediated diseases.
- Published
- 2001
- Full Text
- View/download PDF
141. IFN-alpha and IL-10 induce the differentiation of human type 1 T regulatory cells.
- Author
-
Levings MK, Sangregorio R, Galbiati F, Squadrone S, de Waal Malefyt R, and Roncarolo MG
- Subjects
- Animals, CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes immunology, Cell Differentiation immunology, Cells, Cultured, Clone Cells, Coculture Techniques, Drug Combinations, Drug Synergism, Fetal Blood cytology, Fetal Blood immunology, Humans, Interferon-alpha blood, Interleukin-10 blood, Intracellular Fluid immunology, Isoantigens immunology, L Cells, Lymphocyte Activation immunology, Lymphocyte Culture Test, Mixed, Mice, Receptors, Antigen, T-Cell physiology, Signal Transduction immunology, Transforming Growth Factor beta pharmacology, Interferon-alpha pharmacology, Interleukin-10 pharmacology, T-Lymphocyte Subsets cytology, T-Lymphocyte Subsets immunology, T-Lymphocytes, Regulatory cytology, T-Lymphocytes, Regulatory immunology
- Abstract
CD4(+) T regulatory type 1 (Tr1) cells suppress Ag-specific immune responses in vitro and in vivo. Although IL-10 is critical for the differentiation of Tr1 cells, the effects of other cytokines on differentiation of naive T cells into Tr1 cells have not been investigated. Here we demonstrate that endogenous or exogenous IL-10 in combination with IFN-alpha, but not TGF-beta, induces naive CD4(+) T cells derived from cord blood to differentiate into Tr1 cells: IL-10(+)IFN-gamma(+)IL-2(-/low)IL-4(-). Naive CD4(+) T cells derived from peripheral blood require both exogenous IL-10 and IFN-alpha for Tr1 cell differentiation. The proliferative responses of the Tr1-containing lymphocyte populations, following activation with anti-CD3 and anti-CD28 mAbs, were reduced. Similarly, cultures containing Tr1 cells displayed reduced responses to alloantigens via a mechanism that was partially mediated by IL-10 and TGF-beta. More importantly, Tr1-containing populations strongly suppressed responses of naive T cells to alloantigens. Collectively, these results show that IFN-alpha strongly enhances IL-10-induced differentiation of functional Tr1 cells, which represents a first major step in establishing specific culture conditions to generate T regulatory cells for biological and biochemical analysis, and for cellular therapy to induce peripheral tolerance in humans.
- Published
- 2001
- Full Text
- View/download PDF
142. Differentiation of T regulatory cells by immature dendritic cells.
- Author
-
Roncarolo MG, Levings MK, and Traversari C
- Subjects
- Cell Communication, Cell Differentiation, Dendritic Cells classification, Humans, Immune Tolerance, Inflammation immunology, Self Tolerance, Dendritic Cells cytology, Dendritic Cells immunology, T-Lymphocytes cytology, T-Lymphocytes immunology
- Published
- 2001
- Full Text
- View/download PDF
143. Altered T-cell receptor + CD28-mediated signaling and blocked cell cycle progression in interleukin 10 and transforming growth factor-beta-treated alloreactive T cells that do not induce graft-versus-host disease.
- Author
-
Boussiotis VA, Chen ZM, Zeller JC, Murphy WJ, Berezovskaya A, Narula S, Roncarolo MG, and Blazar BR
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Blood Group Incompatibility, CD28 Antigens drug effects, CD28 Antigens immunology, CD4-Positive T-Lymphocytes immunology, Cell Cycle drug effects, Cell Cycle immunology, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclin-Dependent Kinases drug effects, Cyclin-Dependent Kinases metabolism, Drug Synergism, Graft vs Host Disease immunology, Immune Tolerance drug effects, Interleukin-10 immunology, Lymphocyte Culture Test, Mixed, Membrane Proteins drug effects, Membrane Proteins immunology, Membrane Proteins physiology, Mice, Mice, Inbred C57BL, Mitogen-Activated Protein Kinases metabolism, Mitogen-Activated Protein Kinases pharmacology, Models, Animal, Phosphoproteins metabolism, Phosphorylation drug effects, Receptors, Antigen, T-Cell drug effects, Receptors, Antigen, T-Cell immunology, Receptors, Antigen, T-Cell physiology, Signal Transduction drug effects, Signal Transduction immunology, Transforming Growth Factor beta immunology, Tyrosine metabolism, CD28 Antigens physiology, CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes drug effects, Graft vs Host Disease prevention & control, Interleukin-10 pharmacology, Transforming Growth Factor beta pharmacology
- Abstract
The induction of anergy in T cells, although widely accepted as critical for the maintenance of tolerance, is still poorly understood at the molecular level. Recent evidence demonstrates that in addition to blockade of costimulation using monoclonal antibodies (mAbs) directed against cell surface determinants, treatment of mixed lymphocyte reaction (MLR) cultures with interleukin 10 (IL-10) and transforming growth factor-beta (TGF-beta) results in induction of tolerance, rendering alloreactive murine CD4(+) T cells incapable of inducing graft-versus-host disease (GVHD) after in vivo transfer to histoincompatible recipients. The present study, using these cells prior to adoptive transfer, determined that IL-10 + TGF-beta-tolerant CD4(+) T cells exhibit an altered pattern of T-cell receptor (TCR) + CD28-mediated signaling and are incapable of progressing out of the G(1) phase of the cell cycle during stimulation with HLA class II disparate antigen-presenting cells. TGFbeta + IL-10-tolerant cells were incapable of phosphorylating TCR-zeta, or activating ZAP-70, Ras, and MAPK, similarly to T-cell tolerized by blockade of B7/CD28 and CD40/CD40L pathways. Moreover, these cells were incapable of clonal expansion due to defective synthesis of cyclin D3 and cyclin A, and defective activation of cyclin-dependent kinase (cdk)4, cdk6, and cdk2. These cells also exhibited defective down-regulation of p27(kip1) cdk inhibitor and lack of cyclin D2-cdk4 activation, Rb hyperphosphorylation, and progression to the S phase of the cell cycle. These data link anergy-specific proximal biochemical alterations and the downstream nuclear pathways that control T-cell expansion and provide a biochemical profile of IL-10 + TGF-beta-tolerant alloreactive T cells that do not induce GVHD when transferred into MHC class II disparate recipients in vivo.
- Published
- 2001
- Full Text
- View/download PDF
144. The role of different subsets of T regulatory cells in controlling autoimmunity.
- Author
-
Roncarolo MG and Levings MK
- Subjects
- Animals, CD4-Positive T-Lymphocytes cytology, Cell Differentiation, Clonal Anergy immunology, Cytokines immunology, Humans, Leukocyte Common Antigens immunology, Receptors, Interleukin-2 immunology, T-Lymphocytes, Helper-Inducer immunology, Autoimmunity immunology, CD4-Positive T-Lymphocytes immunology, T-Lymphocyte Subsets immunology
- Abstract
T regulatory cells-in addition to clonal deletion and anergy-are essential for the downregulation of T cell responses to both foreign and self antigens, and for the prevention of autoimmunity. Recent progress has been made in characterising the different subsets of T regulatory cells, the factors that drive their differentiation, and their mode of action. The resolution of these mechanisms will make it possible to use T regulatory cells therapeutically in human autoimmune diseases.
- Published
- 2000
- Full Text
- View/download PDF
145. T cell receptor-dependent activation of human lymphocytes through cell surface ganglioside GT1b: implications for innate immunity.
- Author
-
Bukowski JF, Roncarolo MG, Spits H, Krangel MS, Morita CT, Brenner MB, and Band H
- Subjects
- Cell Line, Transformed, Humans, Gangliosides immunology, Lymphocyte Activation immunology, Receptors, Antigen, T-Cell, gamma-delta immunology, Signal Transduction immunology, T-Lymphocytes immunology
- Abstract
Gangliosides form a component of the glycosphingolipid-rich membrane microdomains recently shown to play an important role in receptor signal transduction. Specific gangliosides also serve as receptors for binding and internalization of bacterial toxins. In the course of characterizing the basis of the native tetanus toxin (TTx) reactivity of a human gamma delta T cell clone, we observed that transfer of the TCR was required to impart TTx reactivity on a TCR-negative recipient T cell. However, the reconstitution of toxin reactivity could be achieved regardless of the antigen specificity of the TCR chains. Further analysis showed that the T cell recognition of native TTx was dependent on the presence of its ganglioside receptor, GT1b, on the T cell surface. Incorporation of exogenous GT1b into plasma membrane conferred TTx reactivity on otherwise non-reactive T cells provided these cells expressed the TCR. Finally, reconstitution of TCR-negative Jurkat T cells with a CD8-CD3zeta chain chimera demonstrated that the cytoplasmic region of the CD3zeta chain was sufficient to couple ganglioside-mediated TTx binding to T cell activation. These data reveal a novel mode of TCR-dependent reactivity to a bacterial toxin that could mobilize a large subset of T cells, thus representing a form of innate immunity. Given the possibility that endogenous ligands may bind to cell surface gangliosides, regulation of their levels and topology on the cell surface may constitute an immunoregulatory mechanism.
- Published
- 2000
- Full Text
- View/download PDF
146. T-regulatory 1 cells: a novel subset of CD4 T cells with immunoregulatory properties.
- Author
-
Levings MK and Roncarolo MG
- Subjects
- Antibody Formation drug effects, CD4-Positive T-Lymphocytes metabolism, Cytokines biosynthesis, Down-Regulation drug effects, Humans, Immune Tolerance, Interleukin-10 biosynthesis, Interleukin-10 immunology, Interleukin-10 physiology, Transforming Growth Factor beta biosynthesis, CD4-Positive T-Lymphocytes classification, CD4-Positive T-Lymphocytes immunology
- Abstract
Clonal deletion and clonal anergy are well established mechanisms of peripheral tolerance. A role has also been described for clonal suppression by regulatory cells in the induction of peripheral tolerance to a variety of antigens. However, it has been difficult to isolate regulatory cells and to define their mechanism of action. We have recently reported the in vitro generation and characterization of a novel subset of CD4(+) T cells that have regulatory properties and are able to suppress antigen-specific immune responses in vitro and in vivo. These T-regulatory 1 (Tr1) cells are defined by their unique profile of cytokine production and make high levels of IL-10 and TGF-beta, but no IL-4 or IL-2. The IL-10 and TGF-beta produced by these cells mediate the inhibition of primary naive T cells in vitro. There is also evidence that Tr1 cells exist in vivo, and we have documented the presence of high IL-10-producing CD4(+) T cells in patients with severe combined immunodeficiency who have received allogeneic stem-cell transplants. These findings support the notion that Tr1 cells are involved in the regulation of peripheral tolerance and that they could potentially be used as a cellular therapy to modulate immune responses in vivo.
- Published
- 2000
- Full Text
- View/download PDF
147. Generation of primary antigen-specific human T- and B-cell responses in immunocompetent SCID-hu mice.
- Author
-
Carballido JM, Namikawa R, Carballido-Perrig N, Antonenko S, Roncarolo MG, and de Vries JE
- Subjects
- Animals, Antibodies, Heterophile blood, Antibody Formation, Antigens, CD analysis, Antigens, CD genetics, Bone Transplantation immunology, Bone Transplantation pathology, Fetal Tissue Transplantation immunology, Fetal Tissue Transplantation pathology, Humans, Immunity, Cellular, Immunoglobulin G blood, Immunoglobulin M blood, Immunohistochemistry, In Situ Hybridization, Mice, Mice, SCID, Skin Transplantation pathology, Thymus Gland immunology, Transplantation, Isogeneic, Antigens, Heterophile immunology, B-Lymphocytes immunology, Skin Transplantation immunology, T-Lymphocytes immunology, Transplantation, Heterologous immunology
- Published
- 2000
- Full Text
- View/download PDF
148. Induction of CD4+ T cell alloantigen-specific hyporesponsiveness by IL-10 and TGF-beta.
- Author
-
Zeller JC, Panoskaltsis-Mortari A, Murphy WJ, Ruscetti FW, Narula S, Roncarolo MG, and Blazar BR
- Subjects
- Amino Acid Sequence, Animals, Antigens immunology, Cells, Cultured, Drug Combinations, Epitopes, T-Lymphocyte immunology, Graft vs Host Disease etiology, Graft vs Host Disease mortality, Immunosuppressive Agents pharmacology, Interleukin-10 pharmacology, Lymphocyte Culture Test, Mixed methods, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Molecular Sequence Data, Ovalbumin immunology, Transforming Growth Factor beta pharmacology, CD4-Positive T-Lymphocytes immunology, Immune Tolerance immunology, Interleukin-10 physiology, Isoantigens immunology, Transforming Growth Factor beta physiology
- Abstract
Induction and maintenance of Ag-specific tolerance are pivotal for immune homeostasis, prevention of autoimmune disorders, and the goal of transplantation. Recent studies suggest that certain cytokines, notably IL-10 and TGF-beta, may play a role in down-regulating immune functions. To further examine the role of cytokines in Ag-specific hyporesponsiveness, murine CD4+ T cells were exposed ex vivo to alloantigen-bearing stimulators in the presence of exogenous IL-10 and/or TGF-beta. Primary but not secondary alloantigen proliferative responses were inhibited by IL-10 alone. However, the combined addition of IL-10 + TGF-beta markedly induced alloantigen hyporesponsiveness in both primary and secondary MLR cultures. Alloantigen-specific hyporesponsiveness was observed also under conditions in which nominal Ag responses were intact. In adoptive transfer experiments, IL-10 + TGF-beta-treated CD4+ T cells, but not T cells treated with either cytokine alone, were markedly impaired in inducing graft-vs-host disease alloresponses to MHC class II disparate recipients. These data provide the first formal evidence that IL-10 and TGF-beta have at least an additive effect in inducing alloantigen-specific tolerance, and that in vitro cytokines can be exploited to suppress CD4+ T cell-mediated Ag-specific responses in vivo.
- Published
- 1999
149. IL-10 transgenic mice present a defect in T cell development reminiscent of SCID patients.
- Author
-
Rouleau M, Cottrez F, Bigler M, Antonenko S, Carballido JM, Zlotnik A, Roncarolo MG, and Groux H
- Subjects
- Animals, Animals, Newborn genetics, Animals, Newborn immunology, CD4-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes cytology, Cell Differentiation genetics, Cell Differentiation immunology, Dendritic Cells cytology, Fetus, Humans, Interleukin-10 biosynthesis, Killer Cells, Natural cytology, Killer Cells, Natural metabolism, Male, Mice, Mice, Inbred BALB C, Organ Culture Techniques, Receptors, Antigen, T-Cell, gamma-delta genetics, Severe Combined Immunodeficiency genetics, Stem Cells immunology, Stem Cells pathology, Stromal Cells immunology, Stromal Cells metabolism, T-Lymphocytes immunology, Thymus Gland immunology, Thymus Gland pathology, Interleukin-10 genetics, Mice, Transgenic immunology, Severe Combined Immunodeficiency immunology, Severe Combined Immunodeficiency pathology, T-Lymphocytes pathology
- Abstract
To analyze the effect of IL-10 overexpressed by APCs as observed in some SCID patients, we have expressed the human IL-10 cDNA under the control of the murine MHC class II promoter in transgenic mice. Similar to SCID patients, these mice presented a defect in T cell maturation characterized by a rapid thymic aplasia that started after birth. The blockage in T cell maturation was strictly restricted to TCR-alpha beta T cells as the absolute number of thymic dendritic, TCR-gamma delta and NK1.1 T cells were equivalent to control littermates. Crossing IL-10 transgenic mice with TCR transgenic mice or treatment with staphylococcal enterotoxin B showed that the defect was not related to the impairment of positive or negative selection. However, repopulating of IL-10 transgenic mouse-fetal thymic organ culture with different stages of triple negative T cells isolated from control mice showed that the blockage occurred specifically at the pre-T cell stage and was reverted by treatment with blocking anti-IL-10 mAbs. These results demonstrate that IL-10 regulates T cell maturation and that dysregulation of IL-10 expression can lead to severe T cell immunodeficiency.
- Published
- 1999
150. Administration of Flk2/Flt3 ligand induces expansion of human high-proliferative potential colony-forming cells in the SCID-hu mouse.
- Author
-
Namikawa R, Muench MO, Firpo MT, Humeau L, Xu Y, Menon S, and Roncarolo MG
- Subjects
- Animals, Antigens, CD analysis, Antigens, Differentiation, Myelomonocytic analysis, Bone Transplantation, Bone and Bones embryology, Flow Cytometry, Hematopoietic Stem Cells immunology, Humans, Lewis X Antigen analysis, Mice, Mice, SCID, Recombinant Proteins pharmacology, Sialic Acid Binding Ig-like Lectin 3, Transplantation, Heterologous, fms-Like Tyrosine Kinase 3, Cell Division, Hematopoietic Stem Cells cytology, Proto-Oncogene Proteins pharmacology, Receptor Protein-Tyrosine Kinases pharmacology
- Abstract
The effects of Flk2/Flt3 ligand (FL) administration on human hematopoiesis were investigated using SCID-hu mice transplanted with human fetal bone fragments. Treatment with recombinant human FL induced significant increases in the frequencies of the high-proliferative potential colony-forming cells and low-proliferative potential colony-forming cells in steady-state human bone marrow. FL also promoted the expansion of high-proliferative potential colony-forming cells and low-proliferative potential colony-forming cells in the human bone marrow during the recovery phase after irradiation, which was evident in increases in the frequencies as well as in the absolute numbers of colony-forming cells. Furthermore, higher percentages of CD33+ CD15- cells were found in the marrows treated with FL as compared to that of controls, indicating that FL hastened the recovery of at least some aspect of myelopoiesis after irradiation. These results indicate that FL induces the expansion of primitive hematopoietic progenitor cells in vivo and, therefore, may be useful in treating patients to promote an early hematopoietic recovery after cytoablative therapies.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.