176 results on '"Rosa Bacchetta"'
Search Results
2. Case report: Refractory Evans syndrome in two patients with spondyloenchondrodysplasia with immune dysregulation treated successfully with JAK1/JAK2 inhibition
- Author
-
Yael Gernez, Mansi Narula, Alma-Martina Cepika, Juanita Valdes Camacho, Elisabeth G. Hoyte, Kirsten Mouradian, Bertil Glader, Deepika Singh, Bindu Sathi, Latha Rao, Ana L. Tolin, Kenneth I. Weinberg, David B. Lewis, Rosa Bacchetta, and Katja G. Weinacht
- Subjects
spondyloenchondrodysplasia ,ACP5 ,tartrate-resistant acid phosphatase ,autoimmunity ,interferonopathy ,JAK inhibitor ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Biallelic mutations in the ACP5 gene cause spondyloenchondrodysplasia with immune dysregulation (SPENCDI). SPENCDI is characterized by the phenotypic triad of skeletal dysplasia, innate and adaptive immune dysfunction, and variable neurologic findings ranging from asymptomatic brain calcifications to severe developmental delay with spasticity. Immune dysregulation in SPENCDI is often refractory to standard immunosuppressive treatments. Here, we present the cases of two patients with SPENCDI and recalcitrant autoimmune cytopenias who demonstrated a favorable clinical response to targeted JAK inhibition over a period of more than 3 years. One of the patients exhibited steadily rising IgG levels and a bone marrow biopsy revealed smoldering multiple myeloma. A review of the literature uncovered that approximately half of the SPENCDI patients reported to date exhibited increased IgG levels. Screening for multiple myeloma in SPENCDI patients with rising IgG levels should therefore be considered.
- Published
- 2024
- Full Text
- View/download PDF
3. A novel FOXP3 knockout-humanized mouse model for pre-clinical safety and efficacy evaluation of Treg-like cell products
- Author
-
Yohei Sato, Abinaya Nathan, Suzette Shipp, John Fraser Wright, Keri Marie Tate, Prachi Wani, Maria-Grazia Roncarolo, and Rosa Bacchetta
- Subjects
CRISPR-Cas9 ,FOXP3 ,Humanized mouse model ,IPEX syndrome ,Regulatory T cells ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Forkhead box P3 (FOXP3) is an essential transcription factor for regulatory T cell (Treg) function. Defects in Tregs mediate many immune diseases including the monogenic autoimmune disease immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX), which is caused by FOXP3 mutations. Treg cell products are a promising modality to induce allograft tolerance or reduce the use of immunosuppressive drugs to prevent rejection, as well as in the treatment of acquired autoimmune diseases. We have recently opened a phase I clinical trial for IPEX patients using autologous engineered Treg-like cells, CD4LVFOXP3. To facilitate the pre-clinical studies, a novel humanized-mouse (hu-mouse) model was developed whereby immune-deficient mice were transplanted with human hematopoietic stem progenitor cells (HSPCs) in which the FOXP3 gene was knocked out (FOXP3KO) using CRISPR-Cas9. Mice transplanted with FOXP3KO HSPCs had impaired survival, developed lymphoproliferation 10–12 weeks post-transplant and T cell infiltration of the gut, resembling human IPEX. Strikingly, injection of CD4LVFOXP3 into the FOXP3KO hu-mice restored in vivo regulatory functions, including control of lymphoproliferation and inhibition of T cell infiltration in the colon. This hu-mouse disease model can be reproducibly established and constitutes an ideal model to assess pre-clinical efficacy of human Treg cell investigational products.
- Published
- 2023
- Full Text
- View/download PDF
4. Analyses of thymocyte commitment to regulatory T cell lineage in thymus of healthy subjects and patients with 22q11.2 deletion syndrome
- Author
-
Simon Borna, Beruh Dejene, Uma Lakshmanan, Janika Schulze, Kenneth Weinberg, and Rosa Bacchetta
- Subjects
Treg – regulatory T cell ,22q11.2DS ,TSDR ,Treg progenitors ,FOXP3 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The Chromosome 22q11.2 deletion syndrome (22q11.2DS) results in an inborn error of immunity due to defective thymic organogenesis. Immunological abnormalities in 22q11.2DS patients are thymic hypoplasia, reduced output of T lymphocytes by the thymus, immunodeficiency and increased incidence of autoimmunity. While the precise mechanism responsible for increased incidence of autoimmunity is not completely understood, a previous study suggested a defect in regulatory T cells (Treg) cell lineage commitment during T cell development in thymus. Here, we aimed to analyze this defect in more detail. Since Treg development in human is still ill-defined, we first analyzed where Treg lineage commitment occurs. We performed systematic epigenetic analyses of the Treg specific demethylation region (TSDR) of the FOXP3 gene in sorted thymocytes at different developmental stages. We defined CD3+CD4+CD8+ FOXP3+CD25+ as the T cell developmental stage in human where TSDR demethylation first occurs. Using this knowledge, we analyzed the intrathymic defect in Treg development in 22q11.2DS patients by combination of TSDR, CD3, CD4, CD8 locus epigenetics and multicolor flow cytometry. Our data showed no significant differences in Treg cell frequencies nor in their basic phenotype. Collectively, these data suggest that although 22q11.2DS patients present with reduced thymic size and T cell output, the frequencies and the phenotype of Treg cell at each developmental stage are surprisingly well preserved.
- Published
- 2023
- Full Text
- View/download PDF
5. Rare immune diseases paving the road for genome editing-based precision medicine
- Author
-
Mara Pavel-Dinu, Simon Borna, and Rosa Bacchetta
- Subjects
CRISPR/Cas9 ,adeno-associated virus 6 ,base editing ,prime and twin prime editing ,hematopoietic stem and progenitor cells ,RAG2 deficiency ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) genome editing platform heralds a new era of gene therapy. Innovative treatments for life-threatening monogenic diseases of the blood and immune system are transitioning from semi-random gene addition to precise modification of defective genes. As these therapies enter first-in-human clinical trials, their long-term safety and efficacy will inform the future generation of genome editing-based medicine. Here we discuss the significance of Inborn Errors of Immunity as disease prototypes for establishing and advancing precision medicine. We will review the feasibility of clustered regularly interspaced short palindromic repeats-based genome editing platforms to modify the DNA sequence of primary cells and describe two emerging genome editing approaches to treat RAG2 deficiency, a primary immunodeficiency, and FOXP3 deficiency, a primary immune regulatory disorder.
- Published
- 2023
- Full Text
- View/download PDF
6. Autoantibody discovery across monogenic, acquired, and COVID-19-associated autoimmunity with scalable PhIP-seq
- Author
-
Sara E Vazquez, Sabrina A Mann, Aaron Bodansky, Andrew F Kung, Zoe Quandt, Elise MN Ferré, Nils Landegren, Daniel Eriksson, Paul Bastard, Shen-Ying Zhang, Jamin Liu, Anthea Mitchell, Irina Proekt, David Yu, Caleigh Mandel-Brehm, Chung-Yu Wang, Brenda Miao, Gavin Sowa, Kelsey Zorn, Alice Y Chan, Veronica M Tagi, Chisato Shimizu, Adriana Tremoulet, Kara Lynch, Michael R Wilson, Olle Kämpe, Kerry Dobbs, Ottavia M Delmonte, Rosa Bacchetta, Luigi D Notarangelo, Jane C Burns, Jean-Laurent Casanova, Michail S Lionakis, Troy R Torgerson, Mark S Anderson, and Joseph L DeRisi
- Subjects
PhIP-seq ,autoantibody ,autoantigen ,COVID-19 ,APS1 ,IPEX ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Phage immunoprecipitation sequencing (PhIP-seq) allows for unbiased, proteome-wide autoantibody discovery across a variety of disease settings, with identification of disease-specific autoantigens providing new insight into previously poorly understood forms of immune dysregulation. Despite several successful implementations of PhIP-seq for autoantigen discovery, including our previous work (Vazquez et al., 2020), current protocols are inherently difficult to scale to accommodate large cohorts of cases and importantly, healthy controls. Here, we develop and validate a high throughput extension of PhIP-seq in various etiologies of autoimmune and inflammatory diseases, including APS1, IPEX, RAG1/2 deficiency, Kawasaki disease (KD), multisystem inflammatory syndrome in children (MIS-C), and finally, mild and severe forms of COVID-19. We demonstrate that these scaled datasets enable machine-learning approaches that result in robust prediction of disease status, as well as the ability to detect both known and novel autoantigens, such as prodynorphin (PDYN) in APS1 patients, and intestinally expressed proteins BEST4 and BTNL8 in IPEX patients. Remarkably, BEST4 antibodies were also found in two patients with RAG1/2 deficiency, one of whom had very early onset IBD. Scaled PhIP-seq examination of both MIS-C and KD demonstrated rare, overlapping antigens, including CGNL1, as well as several strongly enriched putative pneumonia-associated antigens in severe COVID-19, including the endosomal protein EEA1. Together, scaled PhIP-seq provides a valuable tool for broadly assessing both rare and common autoantigen overlap between autoimmune diseases of varying origins and etiologies.
- Published
- 2022
- Full Text
- View/download PDF
7. Editorial: IPEX 2020: An Expanding Disease Spectrum and Novel Precision Therapies
- Author
-
Rosa Bacchetta and Talal Chatila
- Subjects
FOXP3 ,regulatory T cells ,IPEX ,autoimmune enteropathy ,TSDR ,Pediatrics ,RJ1-570 - Published
- 2022
- Full Text
- View/download PDF
8. Co-Expression of FOXP3FL and FOXP3Δ2 Isoforms Is Required for Optimal Treg-Like Cell Phenotypes and Suppressive Function
- Author
-
Yohei Sato, Jessica Liu, Esmond Lee, Rhonda Perriman, Maria Grazia Roncarolo, and Rosa Bacchetta
- Subjects
Foxp3 ,alternative splicing ,gene editing (CRISPR/Cas9) ,gene therapy ,suppressive function ,regulatory T cell (Treg) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
FOXP3 is the master transcription factor in both murine and human FOXP3+ regulatory T cells (Tregs), a T-cell subset with a central role in controlling immune responses. Loss of the functional Foxp3 protein in scurfy mice leads to acute early-onset lethal lymphoproliferation. Similarly, pathogenic FOXP3 mutations in humans lead to immunodysregulation, polyendocrinopathy, enteropathy, and X-linked (IPEX) syndrome, which are characterized by systemic autoimmunity that typically begins in the first year of life. However, although pathogenic FOXP3 mutations lead to overlapping phenotypic consequences in both systems, FOXP3 in human Tregs, but not mouse, is expressed as two predominant isoforms, the full length (FOXP3FL) and the alternatively spliced isoform, delta 2 (FOXP3Δ2). Here, using CRISPR/Cas9 to generate FOXP3 knockout CD4+ T cells (FOXP3KOGFP CD4+ T cells), we restore the expression of each isoform by lentiviral gene transfer to delineate their functional roles in human Tregs. When compared to FOXP3FL or FOXP3Δ2 alone, or double transduction of the same isoform, co-expression of FOXP3FL and FOXP3Δ2 induced the highest overall FOXP3 protein expression in FOXP3KOGFP CD4+ T cells. This condition, in turn, led to optimal acquisition of Treg-like cell phenotypes including downregulation of cytokines, such as IL-17, and increased suppressive function. Our data confirm that co-expression of FOXP3FL and FOXP3Δ2 leads to optimal Treg-like cell function and supports the need to maintain the expression of both when engineering therapeutics designed to restore FOXP3 function in otherwise deficient cells.
- Published
- 2021
- Full Text
- View/download PDF
9. BHLHE40 Regulates IL-10 and IFN-γ Production in T Cells but Does Not Interfere With Human Type 1 Regulatory T Cell Differentiation
- Author
-
Molly Javier Uyeda, Robert A. Freeborn, Brandon Cieniewicz, Rosa Romano, Ping (Pauline) Chen, Jeffrey Mao-Hwa Liu, Benjamin Thomas, Esmond Lee, Alma-Martina Cepika, Rosa Bacchetta, and Maria Grazia Roncarolo
- Subjects
T cell ,regulatory ,cytokine ,transcription factor ,transcriptome ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Type 1 regulatory T (Tr1) cells are subset of peripherally induced antigen-specific regulatory T cells. IL-10 signaling has been shown to be indispensable for polarization and function of Tr1 cells. However, the transcriptional machinery underlying human Tr1 cell differentiation and function is not yet elucidated. To this end, we performed RNA sequencing on ex vivo human CD49b+LAG3+ Tr1 cells. We identified the transcription factor, BHLHE40, to be highly expressed in Tr1 cells. Even though Tr1 cells characteristically produce high levels of IL-10, we found that BHLHE40 represses IL-10 and increases IFN-γ secretion in naïve CD4+ T cells. Through CRISPR/Cas9-mediated knockout, we determined that IL10 significantly increased in the sgBHLHE40-edited cells and BHLHE40 is dispensable for naïve CD4+ T cells to differentiate into Tr1 cells in vitro. Interestingly, BHLHE40 overexpression induces the surface expression of CD49b and LAG3, co-expressed surface molecules attributed to Tr1 cells, but promotes IFN-γ production. Our findings uncover a novel mechanism whereby BHLHE40 acts as a regulator of IL-10 and IFN-γ in human CD4+ T cells.
- Published
- 2021
- Full Text
- View/download PDF
10. 146 Alloantigen-specific Tr1 cells designed to prevent GvHD have a distinct molecular identity and suppress through CTLA-4 and PD-1
- Author
-
Mark M Davis, Alice Bertaina, Alma-Martina Cepika, Pauline P Chen, Molly Uyeda, Brandon Cieniewicz, Mansi Narula, Laura Amaya, David M Louis, Liwen Xu, Xuhuai Ji, Rajni Agarwal-Hashmi, Everett Meyer, Rosa Bacchetta, and Maria Grazia Roncarolo
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
11. Engineered type 1 regulatory T cells designed for clinical use kill primary pediatric acute myeloid leukemia cells
- Author
-
Brandon Cieniewicz, Molly Javier Uyeda, Ping (Pauline) Chen, Ece Canan Sayitoglu, Jeffrey Mao-Hwa Liu, Grazia Andolfi, Katharine Greenthal, Alice Bertaina, Silvia Gregori, Rosa Bacchetta, Norman James Lacayo, Alma-Martina Cepika, and Maria Grazia Roncarolo
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Type 1 regulatory (Tr1) T cells induced by enforced expression of IL-10 (LV-10) are being developed as a novel treatment for chemotherapy-resistant myeloid leukemias. In vivo, LV-10 cells do not cause graft vs host disease while mediating graft vs leukemia (GvL) effect against adult acute myeloid leukemia (AML). Since pediatric AML (pAML) and adult AML are different on a genetic and epigenetic level, we investigate herein whether LV-10 cells also efficiently kill pAML cells. We show that the majority of primary pAML are killed by LV-10 cells, with different levels of sensitivity to killing. Transcriptionally, pAML sensitive to LV-10 killing expressed a myeloid maturation signature. Overlaying the signatures of sensitive and resistant pAML onto the public NCI TARGET pAML dataset revealed that sensitive pAML clustered with M5 monocytic pAML and pAML with MLL rearrangement. Resistant pAML clustered with myelomonocytic leukemias and those bearing the core binding factor translocations inv(16) or t(8;21)(RUNX1-RUNX1T1). Furthermore, resistant pAML upregulated the membrane glycoprotein CD200, which binds to the inhibitory receptor CD200R1 on LV-10 cells. To examine if CD200 expression on target cells can impair LV-10 cell function, we overexpressed CD200 in myeloid leukemia cell lines ordinarily sensitive to LV-10 killing. Indeed, LV-10 cells degranulated less and killed fewer CD200-overexpressing cells compared to controls, indicating that pAML can utilize CD200 expression for immune evasion. Altogether, the majority of pAML are killed by LV-10 cells in vitro, supporting further LV-10 cell development as an innovative cell therapy for pAML.
- Published
- 2020
- Full Text
- View/download PDF
12. Hematopoietic Cell Transplantation in Patients With Primary Immune Regulatory Disorders (PIRD): A Primary Immune Deficiency Treatment Consortium (PIDTC) Survey
- Author
-
Alice Y. Chan, Jennifer W. Leiding, Xuerong Liu, Brent R. Logan, Lauri M. Burroughs, Eric J. Allenspach, Suzanne Skoda-Smith, Gulbu Uzel, Luigi D. Notarangelo, Mary Slatter, Andrew R. Gennery, Angela R. Smith, Sung-Yun Pai, Michael B. Jordan, Rebecca A. Marsh, Morton J. Cowan, Christopher C. Dvorak, John A. Craddock, Susan E. Prockop, Shanmuganathan Chandrakasan, Neena Kapoor, Rebecca H. Buckley, Suhag Parikh, Deepak Chellapandian, Benjamin R. Oshrine, Jeffrey J. Bednarski, Megan A. Cooper, Shalini Shenoy, Blachy J. Davila Saldana, Lisa R. Forbes, Caridad Martinez, Elie Haddad, David C. Shyr, Karin Chen, Kathleen E. Sullivan, Jennifer Heimall, Nicola Wright, Monica Bhatia, Geoffrey D. E. Cuvelier, Frederick D. Goldman, Isabelle Meyts, Holly K. Miller, Markus G. Seidel, Mark T. Vander Lugt, Rosa Bacchetta, Katja G. Weinacht, Jeffrey R. Andolina, Emi Caywood, Hey Chong, Maria Teresa de la Morena, Victor M. Aquino, Evan Shereck, Jolan E. Walter, Morna J. Dorsey, Christine M. Seroogy, Linda M. Griffith, Donald B. Kohn, Jennifer M. Puck, Michael A. Pulsipher, and Troy R. Torgerson
- Subjects
primary immune deficiencies ,autoimmunity ,immune dysregulation ,hematopoietic cell transplant ,genetics ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Primary Immune Regulatory Disorders (PIRD) are an expanding group of diseases caused by gene defects in several different immune pathways, such as regulatory T cell function. Patients with PIRD develop clinical manifestations associated with diminished and exaggerated immune responses. Management of these patients is complicated; oftentimes immunosuppressive therapies are insufficient, and patients may require hematopoietic cell transplant (HCT) for treatment. Analysis of HCT data in PIRD patients have previously focused on a single gene defect. This study surveyed transplanted patients with a phenotypic clinical picture consistent with PIRD treated in 33 Primary Immune Deficiency Treatment Consortium centers and European centers. Our data showed that PIRD patients often had immunodeficient and autoimmune features affecting multiple organ systems. Transplantation resulted in resolution of disease manifestations in more than half of the patients with an overall 5-years survival of 67%. This study, the first to encompass disorders across the PIRD spectrum, highlights the need for further research in PIRD management.
- Published
- 2020
- Full Text
- View/download PDF
13. Case Study: Mechanism for Increased Follicular Helper T Cell Development in Activated PI3K Delta Syndrome
- Author
-
Timothy J. Thauland, Laurence Pellerin, Robert S. Ohgami, Rosa Bacchetta, and Manish J. Butte
- Subjects
primary immunodeficiency ,activated PI3K delta syndrome (APDS) ,PI3K ,T follicular helper cells ,osteopontin ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Gain-of-function variants in p110δ, the catalytic subunit of phosphatidylinositol 3-kinase (PI3K) expressed in lymphocytes, cause activated PI3-kinase δ syndrome (APDS), a primary immunodeficiency that is characterized by recurrent infections, viremia, lymphadenopathy, and autoimmunity. The mechanism of autoimmunity in APDS has not been well-understood. Here, we show the profound skewing of peripheral CD4+ T cells to a T follicular helper (TFH) phenotype in a patient with APDS bearing a novel p110δ variant, Y524S. We also saw a diminishment of transient Foxp3 expression in activated T cells. Mechanistic studies revealed that both the new variant and a previously described, pathogenic variant (E81K) enhanced an interaction between intracellular Osteopontin and p85α. This interaction had been shown in mice to promote TFH differentiation. Our results demonstrate a new influence of PI3K on human T cell differentiation that is unrelated to its lipid-kinase activity and suggest that TFH should be monitored in APDS patients.
- Published
- 2019
- Full Text
- View/download PDF
14. APVO210: A Bispecific Anti-CD86-IL-10 Fusion Protein (ADAPTIR™) to Induce Antigen-Specific T Regulatory Type 1 Cells
- Author
-
Laurence Pellerin, Ping Chen, Silvia Gregori, Gabriela Hernandez-Hoyos, Rosa Bacchetta, and Maria Grazia Roncarolo
- Subjects
IL-10 ,CD86 ,T regulatory type 1 cells ,tolerogenic dendritic cells ,anergy ,immunomodulation ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IL-10 is a potent immunosuppressive cytokine that promotes the differentiation of tolerogenic dendritic cells (DC-10), and the subsequent induction of antigen-specific T regulatory type 1 (Tr1) cells, which suppress immune responses. However, IL-10 acts on multiple cell types and its effects are not solely inhibitory, therefore, limiting its use as immunomodulant. APVO210 is a bispecific fusion protein composed of an anti-CD86 antibody fused with monomeric IL-10 (ADAPTIR™ from Aptevo Therapeutics). APVO210 specifically induces IL-10R signaling in CD86+ antigen-presenting cells, but not in T and B cells. In this study, we tested whether APVO210 promotes the differentiation of tolerogenic DC-10 and the differentiation of antigen-specific CD4+ Tr1 cells in vitro. We compared the effect of APVO210 with that of recombinant human (rh) IL-10 on the in vitro differentiation of DC-10, induction of alloantigen-specific anergic CD4+ T cells, enrichment in CD49b+LAG3+ Tr1 cells mediating antigen-specific suppression, and stability upon exposure to inflammatory cytokines. APVO210 induced the differentiation of tolerogenic DC (DC-A210) that produced high levels of IL-10, expressed CD86, HLA-G, and intermediate levels of CD14 and CD16. These DC-A210 induced alloantigen-specific anergic T-cell cultures (T-alloA210) that were enriched in CD49b+ LAG3+ Tr1 cells, produced high levels of IL-10, and had suppressive properties. The phenotype and high IL-10 production by DC-A210, and the alloantigen-specific anergy of T-alloA210 were preserved upon exposure to the inflammatory cytokines IL-1β, IL-6, and TNF-α. The effects of APVO210 were comparable to that of dimeric rh IL-10. In conclusion, our data demonstrate that APVO210 drives the differentiation of tolerogenic DC and functional alloantigen-specific Tr1 cells in vitro. Since APVO210 specifically targets CD86+ cells, we hypothesize that it will specifically target CD86+ DC to induce Tr1 cells in vivo, and mediate antigen-specific immunological tolerance by induction of tolerogenic DC and Tr1 cells.
- Published
- 2018
- Full Text
- View/download PDF
15. Neutralizing Anti-Cytokine Autoantibodies Against Interferon-α in Immunodysregulation Polyendocrinopathy Enteropathy X-Linked
- Author
-
Jacob M. Rosenberg, Maria E. Maccari, Federica Barzaghi, Eric J. Allenspach, Claudio Pignata, Giovanna Weber, Troy R. Torgerson, Paul J. Utz, and Rosa Bacchetta
- Subjects
anti-cytokine autoantibodies ,interferon-alpha ,immunodysregulation polyendocrinopathy enteropathy X-linked ,autoimmune polyendocrine syndrome type I ,protein microarrays ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Anti-cytokine autoantibodies (ACAAs) have been described in a growing number of primary immunodeficiencies with autoimmune features, including autoimmune polyendocrine syndrome type I (APS-1), a prototypical disease of defective T cell-mediated central tolerance. Whether defects in peripheral tolerance lead to similar ACAAs is unknown. Immunodysregulation polyendocrinopathy enteropathy X-linked (IPEX) is caused by mutations in FOXP3, a master regulator of T regulatory cells (Treg), and consequently results in defective T cell-mediated peripheral tolerance. Unique autoantibodies have previously been described in IPEX. To test the hypothesis that ACAAs are present in IPEX, we designed and fabricated antigen microarrays. We discovered elevated levels of IgG ACAAs against interferon-α (IFN-α) in a cohort of IPEX patients. Serum from IPEX patients blocked IFN-α signaling in vitro and blocking activity was tightly correlated with ACAA titer. To show that blocking activity was mediated by IgG and not other serum factors, we purified IgG and showed that blocking activity was contained entirely in the immunoglobulin fraction. We also screened for ACAAs against IFN-α in a second geographically distinct cohort. In these samples, ACAAs against IFN-α were elevated in a post hoc analysis. In summary, we report the discovery of ACAAs against IFN-α in IPEX, an experiment of nature demonstrating the important role of peripheral T cell tolerance.
- Published
- 2018
- Full Text
- View/download PDF
16. Minimum Information about T Regulatory Cells: A Step toward Reproducibility and Standardization
- Author
-
Anke Fuchs, Mateusz Gliwiński, Nathali Grageda, Rachel Spiering, Abul K. Abbas, Silke Appel, Rosa Bacchetta, Manuela Battaglia, David Berglund, Bruce Blazar, Jeffrey A. Bluestone, Martin Bornhäuser, Anja ten Brinke, Todd M. Brusko, Nathalie Cools, Maria Cristina Cuturi, Edward Geissler, Nick Giannoukakis, Karolina Gołab, David A. Hafler, S. Marieke van Ham, Joanna Hester, Keli Hippen, Mauro Di Ianni, Natasa Ilic, John Isaacs, Fadi Issa, Dorota Iwaszkiewicz-Grześ, Elmar Jaeckel, Irma Joosten, David Klatzmann, Hans Koenen, Cees van Kooten, Olle Korsgren, Karsten Kretschmer, Megan Levings, Natalia Maria Marek-Trzonkowska, Marc Martinez-Llordella, Djordje Miljkovic, Kingston H.G. Mills, Joana P. Miranda, Ciriaco A. Piccirillo, Amy L. Putnam, Thomas Ritter, Maria Grazia Roncarolo, Shimon Sakaguchi, Silvia Sánchez-Ramón, Birgit Sawitzki, Ljiljana Sofronic-Milosavljevic, Megan Sykes, Qizhi Tang, Marta Vives-Pi, Herman Waldmann, Piotr Witkowski, Kathryn J. Wood, Silvia Gregori, Catharien M. U. Hilkens, Giovanna Lombardi, Phillip Lord, Eva M. Martinez-Caceres, and Piotr Trzonkowski
- Subjects
minimum information model ,T regulatory cells ,immunotherapy ,good manufacturing practice ,cell therapy ,immune tolerance ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Cellular therapies with CD4+ T regulatory cells (Tregs) hold promise of efficacious treatment for the variety of autoimmune and allergic diseases as well as posttransplant complications. Nevertheless, current manufacturing of Tregs as a cellular medicinal product varies between different laboratories, which in turn hampers precise comparisons of the results between the studies performed. While the number of clinical trials testing Tregs is already substantial, it seems to be crucial to provide some standardized characteristics of Treg products in order to minimize the problem. We have previously developed reporting guidelines called minimum information about tolerogenic antigen-presenting cells, which allows the comparison between different preparations of tolerance-inducing antigen-presenting cells. Having this experience, here we describe another minimum information about Tregs (MITREG). It is important to note that MITREG does not dictate how investigators should generate or characterize Tregs, but it does require investigators to report their Treg data in a consistent and transparent manner. We hope this will, therefore, be a useful tool facilitating standardized reporting on the manufacturing of Tregs, either for research purposes or for clinical application. This way MITREG might also be an important step toward more standardized and reproducible testing of the Tregs preparations in clinical applications.
- Published
- 2018
- Full Text
- View/download PDF
17. Forkhead-Box-P3 Gene Transfer in Human CD4+ T Conventional Cells for the Generation of Stable and Efficient Regulatory T Cells, Suitable for Immune Modulatory Therapy
- Author
-
Laura Passerini and Rosa Bacchetta
- Subjects
regulatory T cells ,forkhead box P3 ,tolerance ,regulatory T cell-based cell therapy ,gene transfer ,antigen specificity ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The development of novel approaches to control immune responses to self- and allogenic tissues/organs represents an ambitious goal for the management of autoimmune diseases and in transplantation. Regulatory T cells (Tregs) are recognized as key players in the maintenance of peripheral tolerance in physiological and pathological conditions, and Treg-based cell therapies to restore tolerance in T cell-mediated disorders have been designed. However, several hurdles, including insufficient number of Tregs, their stability, and their antigen specificity, have challenged Tregs clinical applicability. In the past decade, the ability to engineer T cells has proven a powerful tool to redirect specificity and function of different cell types for specific therapeutic purposes. By using lentivirus-mediated gene transfer of the thymic-derived Treg transcription factor forkhead-box-P3 (FOXP3) in conventional CD4+ T cells, we converted effector T cells into Treg-like cells, endowed with potent in vitro and in vivo suppressive activity. The resulting CD4FOXP3 T-cell population displays stable phenotype and suppressive function. We showed that this strategy restores Treg function in T lymphocytes from patients carrying mutations in FOXP3 [immune-dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX)], in whom CD4FOXP3 T cell could be used as therapeutics to control autoimmunity. Here, we will discuss the potential advantages of using CD4FOXP3 T cells for in vivo application in inflammatory diseases, where tissue inflammation may undermine the function of natural Tregs. These findings pave the way for the use of engineered Tregs not only in IPEX syndrome but also in autoimmune disorders of different origin and in the context of stem cell and organ transplantation.
- Published
- 2017
- Full Text
- View/download PDF
18. Identification of STAT5A and STAT5B target genes in human T cells.
- Author
-
Takahiro Kanai, Scott Seki, Jennifer A Jenks, Arunima Kohli, Trupti Kawli, Dorrelyn Patacsil Martin, Michael Snyder, Rosa Bacchetta, and Kari C Nadeau
- Subjects
Medicine ,Science - Abstract
Signal transducer and activator of transcription (STAT) comprises a family of universal transcription factors that help cells sense and respond to environmental signals. STAT5 refers to two highly related proteins, STAT5A and STAT5B, with critical function: their complete deficiency is lethal in mice; in humans, STAT5B deficiency alone leads to endocrine and immunological problems, while STAT5A deficiency has not been reported. STAT5A and STAT5B show peptide sequence similarities greater than 90%, but subtle structural differences suggest possible non-redundant roles in gene regulation. However, these roles remain unclear in humans. We applied chromatin immunoprecipitation followed by DNA sequencing using human CD4(+) T cells to detect candidate genes regulated by STAT5A and/or STAT5B, and quantitative-PCR in STAT5A or STAT5B knock-down (KD) human CD4(+) T cells to validate the findings. Our data show STAT5A and STAT5B play redundant roles in cell proliferation and apoptosis via SGK1 interaction. Interestingly, we found a novel, unique role for STAT5A in binding to genes involved in neural development and function (NDRG1, DNAJC6, and SSH2), while STAT5B appears to play a distinct role in T cell development and function via DOCK8, SNX9, FOXP3 and IL2RA binding. Our results also suggest that one or more co-activators for STAT5A and/or STAT5B may play important roles in establishing different binding abilities and gene regulation behaviors. The new identification of these genes regulated by STAT5A and/or STAT5B has major implications for understanding the pathophysiology of cancer progression, neural disorders, and immune abnormalities.
- Published
- 2014
- Full Text
- View/download PDF
19. Autoantibodies to harmonin and villin are diagnostic markers in children with IPEX syndrome.
- Author
-
Vito Lampasona, Laura Passerini, Federica Barzaghi, Carlo Lombardoni, Elena Bazzigaluppi, Cristina Brigatti, Rosa Bacchetta, and Emanuele Bosi
- Subjects
Medicine ,Science - Abstract
Autoantibodies to enterocyte antigens harmonin (75 kDa USH1C protein) and villin (actin-binding 95 kDa protein) are associated with the Immune dysregulation, Polyendocrinopathy, Enteropathy, X-linked (IPEX) syndrome. In this study we evaluated the diagnostic value of harmonin and villin autoantibodies in IPEX and IPEX-like syndromes. Harmonin and villin autoantibodies were measured by a novel Luminescent-Immuno-Precipitation-System (LIPS) quantitative assay, in patients with IPEX, IPEX-like syndrome, Primary Immunodeficiencies (PID) with enteropathy, all diagnosed by sequencing of the FOXP3 gene, and in type 1 diabetes (T1D), celiac disease and healthy blood donors as control groups. Harmonin and villin autoantibodies were detected in 12 (92%) and 6 (46%) of 13 IPEX patients, and in none of the IPEX-like, PID, T1D, celiac patients, respectively. All IPEX patients, including one case with late and atypical clinical presentation, had either harmonin and/or villin autoantibodies and tested positive for enterocyte antibodies by indirect immunofluorescence. When measured in IPEX patients in remission after immunosuppressive therapy or hematopoietic stem cell transplantation, harmonin and villin autoantibodies became undetectable or persisted at low titers in all cases but one in whom harmonin autoantibodies remained constantly high. In one patient, a peak of harmonin antibodies paralleled a relapse phase of enteropathy. Our study demonstrates that harmonin and villin autoantibodies, measured by LIPS, are sensitive and specific markers of IPEX, differentiate IPEX, including atypical cases, from other early childhood disorders associated with enteropathy, and are useful for screening and clinical monitoring of affected children.
- Published
- 2013
- Full Text
- View/download PDF
20. Molecular and functional characterization of allogantigen-specific anergic T cells suitable for cell therapy
- Author
-
Rosa Bacchetta, Silvia Gregori, Giorgia Serafini, Claudia Sartirana, Ute Schulz, Elisabetta Zino, Stefan Tomiuk, Uwe Jansen, Maurilio Ponzoni, Carlo Terenzio Paties, Katharina Fleischhauer, and Maria Grazia Roncarolo
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background CD4+ regulatory T cells are a specialized subset of T cells that actively control immune responses. Several experimental protocols have been used to expand natural regulatory T cells and to generate adaptive type 1 regulatory T cells for regulatory T-cell-based therapies.Design and Methods The ability of exogenous recombinant human interleukin-10 to induce alloantigen-specific anergy in T cells was investigated and compared to that of interleukin-10 derived from tolerogenic dendritic cells, in mixed lymphocyte cultures. A detailed characterization of the effector functions of the resulting anergized T cells is reported.Results Interleukin-10, whether exogenous or derived from tolerogenic dendritic cells, induces a population of alloantigen-specific T cells (interleukin-10-anergized T cells) containing type 1 regulatory T cells, which are anergic and actively suppress alloantigen-specific effector T cells present within the mixed population. Interleukin-10-induced anergy is transforming growth factor-β independent, and is associated with a decreased frequency of alloantigen-specific cytotoxic T lymphocyte precursors, but interleukin-10-anergized T cells are still responsive to third-party, bacterial, and viral antigens. Tolerogenic dendritic cells are more powerful than exogenous interleukin-10 in generating type 1 regulatory T-cell precursors, and are also effective in the context of HLA-matched donors.Conclusions Based on these studies, we have developed an efficient and reproducible in vitro method to generate antigen-specific type 1 regulatory T-cell precursors starting from total peripheral blood cells with minimal cell manipulation and suitable for generating type 1 regulatory T cells for regulatory T-cell-based therapies.
- Published
- 2010
- Full Text
- View/download PDF
21. Type 1 regulatory T cells are associated with persistent split erythroid/lymphoid chimerism after allogeneic hematopoietic stem cell transplantation for thalassemia
- Author
-
Giorgia Serafini, Marco Andreani, Manuela Testi, MariaRosa Battarra, Andrea Bontadini, Eika Biral, Katharina Fleischhauer, Sarah Marktel, Guido Lucarelli, Maria Grazia Roncarolo, and Rosa Bacchetta
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background Thalassemia major can be cured with allogeneic hematopoietic stem cell transplantation. Persistent mixed chimerism develops in around 10% of transplanted thalassemic patients, but the biological mechanisms underlying this phenomenon are poorly understood.Design and Methods The presence of interleukin-10-producing T cells in the peripheral blood of eight patients with persistent mixed chimerism and five with full donor chimerism was investigated. A detailed characterization was then performed, by T-cell cloning, of the effector and regulatory T-cell repertoire of one patient with persistent mixed chimerism, who developed stable split erythroid/lymphoid chimerism after a hematopoietic stem cell transplant from an HLA-matched unrelated donor.Results Higher levels of interleukin-10 were produced by peripheral blood mononuclear cells from patients with persistent mixed chimerism than by the same cells from patients with complete donor chimerism or normal donors. T-cell clones of both host and donor origin could be isolated from the peripheral blood of one, selected patient with persistent mixed chimerism. Together with effector T-cell clones reactive against host or donor alloantigens, regulatory T-cell clones with a cytokine secretion profile typical of type 1 regulatory cells were identified at high frequencies. Type 1 regulatory cell clones, of both donor and host origin, were able to inhibit the function of effector T cells of either donor or host origin in vitro.Conclusions Overall these results suggest that interleukin-10 and type 1 regulatory cells are associated with persistent mixed chimerism and may play an important role in sustaining long-term tolerance in vivo. These data provide new insights into the mechanisms of peripheral tolerance in chimeric patients and support the use of cellular therapy with regulatory T cells following hematopoietic stem cell transplantation.
- Published
- 2009
- Full Text
- View/download PDF
22. FOXP3 TSDR Measurement Could Assist Variant Classification and Diagnosis of IPEX Syndrome
- Author
-
Rebecca C. Wyatt, Sven Olek, Elisa De Franco, Bjoern Samans, Kashyap Patel, Jayne Houghton, Steffi Walter, Janika Schulze, Rosa Bacchetta, Andrew T. Hattersley, Sarah E. Flanagan, and Matthew B. Johnson
- Subjects
Immunology ,Immunology and Allergy - Abstract
Pathogenic FOXP3 variants cause immune dysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome, a progressive autoimmune disease resulting from disruption of the regulatory T cell (Treg) compartment. Assigning pathogenicity to novel variants in FOXP3 is challenging due to the heterogeneous phenotype and variable immunological abnormalities. The number of cells with demethylation at the Treg cell-specific demethylated region (TSDR) is an independent biomarker of IPEX. We aimed to investigate if diagnosing IPEX at presentation with isolated diabetes could allow for effective monitoring of disease progression and assess whether TSDR analysis can aid FOXP3 variant classification and predict disease course. We describe a large genetically diagnosed IPEX cohort (n = 65) and 13 individuals with other monogenic autoimmunity subtypes in whom we quantified the proportion of cells with FOXP3 TSDR demethylation, normalized to the number with CD4 demethylation (%TSDR/CD4) and compare them to 29 unaffected controls. IPEX patients presenting with isolated diabetes (50/65, 77%) often later developed enteropathy (20/50, 40%) with a median interval of 23.5 weeks. %TSDR/CD4 was a good discriminator of IPEX vs. unaffected controls (ROC-AUC 0.81, median 13.6% vs. 8.5%, p p = 1.0). Identifying increased %TSDR/CD4 in patients with novel FOXP3 mutations presenting with isolated diabetes facilitates diagnosis and could offer an opportunity to monitor patients and begin immune modulatory treatment before onset of severe enteropathy.
- Published
- 2023
23. Autoantibody discovery across monogenic, acquired, and COVID-19-associated autoimmunity with scalable PhIP-seq
- Author
-
Aaron Bodansky, Sabrina A Mann, Sara E Vazquez, Andrew F Kung, Zoe Quandt, Elise MN Ferré, Nils Landegren, Daniel Eriksson, Paul Bastard, Shen-Ying Zhang, Jamin Liu, Anthea Mitchell, Irina Proekt, David Yu, Caleigh Mandel-Brehm, Chung-Yu Wang, Brenda Miao, Gavin Sowa, Kelsey Zorn, Alice Y Chan, Veronica M Tagi, Chisato Shimizu, Adriana Tremoulet, Kara Lynch, Michael R Wilson, Olle Kämpe, Kerry Dobbs, Ottavia M Delmonte, Rosa Bacchetta, Luigi D Notarangelo, Jane C Burns, Jean-Laurent Casanova, Michail S Lionakis, Troy R Torgerson, Mark S Anderson, and Joseph L DeRisi
- Subjects
Proteome ,APS1 ,Autoimmunity ,Autoantigens ,Autoimmune Disease ,Article ,General Biochemistry, Genetics and Molecular Biology ,Autoimmune Diseases ,immunology ,IPEX ,Humans ,Immunoprecipitation ,2.1 Biological and endogenous factors ,Bacteriophages ,human ,Aetiology ,Autoantibodies ,Homeodomain Proteins ,General Immunology and Microbiology ,Inflammatory and immune system ,General Neuroscience ,Immunology in the medical area ,COVID-19 ,General Medicine ,autoantigen ,inflammation ,Immunologi inom det medicinska området ,Biochemistry and Cell Biology ,PhIP-seq ,autoantibody - Abstract
Phage Immunoprecipitation-Sequencing (PhIP-Seq) allows for unbiased, proteome-wide autoantibody discovery across a variety of disease settings, with identification of disease-specific autoantigens providing new insight into previously poorly understood forms of immune dysregulation. Despite several successful implementations of PhIP-Seq for autoantigen discovery, including our previous work (Vazquez et al. 2020), current protocols are inherently difficult to scale to accommodate large cohorts of cases and importantly, healthy controls. Here, we develop and validate a high throughput extension of PhIP-seq in various etiologies of autoimmune and inflammatory diseases, including APS1, IPEX, RAG1/2 deficiency, Kawasaki Disease (KD), Multisystem Inflammatory Syndrome in Children (MIS-C), and finally, mild and severe forms of COVID19. We demonstrate that these scaled datasets enable machine-learning approaches that result in robust prediction of disease status, as well as the ability to detect both known and novel autoantigens, such as PDYN in APS1 patients, and intestinally expressed proteins BEST4 and BTNL8 in IPEX patients. Remarkably, BEST4 antibodies were also found in 2 patients with RAG1/2 deficiency, one of whom had very early onset IBD. Scaled PhIP-Seq examination of both MIS-C and KD demonstrated rare, overlapping antigens, including CGNL1, as well as several strongly enriched putative pneumonia-associated antigens in severe COVID19, including the endosomal protein EEA1. Together, scaled PhIP-Seq provides a valuable tool for broadly assessing both rare and common autoantigen overlap between autoimmune diseases of varying origins and etiologies.
- Published
- 2022
24. Author response: Autoantibody discovery across monogenic, acquired, and COVID-19-associated autoimmunity with scalable PhIP-seq
- Author
-
Aaron Bodansky, Sabrina A Mann, Sara E Vazquez, Andrew F Kung, Zoe Quandt, Elise MN Ferré, Nils Landegren, Daniel Eriksson, Paul Bastard, Shen-Ying Zhang, Jamin Liu, Anthea Mitchell, Irina Proekt, David Yu, Caleigh Mandel-Brehm, Chung-Yu Wang, Brenda Miao, Gavin Sowa, Kelsey Zorn, Alice Y Chan, Veronica M Tagi, Chisato Shimizu, Adriana Tremoulet, Kara Lynch, Michael R Wilson, Olle Kämpe, Kerry Dobbs, Ottavia M Delmonte, Rosa Bacchetta, Luigi D Notarangelo, Jane C Burns, Jean-Laurent Casanova, Michail S Lionakis, Troy R Torgerson, Mark S Anderson, and Joseph L DeRisi
- Published
- 2022
25. T-allo10 Infusion after αβ depleted-HSCT in Children and Young Adults with Hematologic Malignancies: Improved Immune Reconstitution in the Absence of Severe GvHD
- Author
-
Alice Bertaina, Rosa Bacchetta, David C. Shyr, Gopin Saini, Jennifer Lee, Karen Kristovich, Rajni Agarwal-Hashmi, Dr. Orly R. Klein, Kathryn Melsop, Keri Tate, Giulia Barbarito, Linda Oppizzi, Pauline Chen, Alma-Martina Cepika, and Maria Grazia Roncarolo
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2023
26. Loss of FOXP3 function causes expansion of two pools of autoreactive T cells in patients with IPEX syndrome
- Author
-
Šimon Borna, Esmond Lee, Uma Lakshmanan, Melissa Mavers, Mansi Narula, Akshaya Ramachandran, Jeanette Baker, Janika Schulze, Sven Olek, Louis Marois, Yael Gernez, Monica Bhatia, Alice Bertaina, Maria Grazia Roncarolo, Eric Meffre, and Rosa Bacchetta
- Abstract
The monogenic autoimmune disease Immunedysregulation polyendocrynopathy entheropathy X-linked syndrome (IPEX) has elucidated the essential function of the transcription factor FOXP3 and of thymic-derived regulatory T (Treg) cells in controlling autoimmunity. However, the presence of autoreactive T cells in IPEX remains undetermined, thus representing a crucial gap in understanding the origin of autoimmunity in a FOXP3 deficient immune system. Combining epigenetic analysis as a lineage marker of Treg identity and TCR sequencing to assess the self-reactive clones, we showed that IPEX patients have two pools of expanded autoreactive T cells. The first originates from the expansion of autoreactive effector T cells (Teff), likely due to loss of Treg suppressive function since it is absent in carrier mothers, in whom Treg cells are functional. The second pool originates, unexpectedly, from Treg cells which lose their phenotypic markers, including CD25 and FOXP3. We call these loss of identity Treg cells and show that they are i) suppressed by healthy donor Treg in a patient post hematopoietic transplantation despite low donor chimerism, and ii) not detectable in patients with Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy syndrome (APECED), a monogenic autoimmune disease of thymic origin. Moreover, we demonstrate that FOXP3 knock-out in Treg cells leads to increased Treg expansion and production of Th17 and Th2 cytokines, known to be increased in IPEX patients. These results suggest that the loss of identity Treg cells could directly contribute to immune dysregulation in IPEX. Collectively, we provide a better understanding of autoimmunity and novel ways to monitor the effects of Treg cell therapies in IPEX disease or other autoimmune diseases.One Sentence SummaryMutations of FOXP3 gene in humans cause expansion of autoreactive T cells originating from both effector T cells and regulatory T cells which gain effector function.
- Published
- 2022
27. Epigenetic and immunological indicators of IPEX disease in subjects with FOXP3 gene mutation
- Author
-
Mansi Narula, Uma Lakshmanan, Simon Borna, Janika J. Schulze, Tyson H. Holmes, Nicholas Harre, Matthew Kirkey, Akshaya Ramachandran, Veronica Maria Tagi, Federica Barzaghi, Eyal Grunebaum, Julia E.M. Upton, Vy Hong-Diep Kim, Christian Wysocki, Victoria R. Dimitriades, Kenneth Weinberg, Katja G. Weinacht, Yael Gernez, Bindu K. Sathi, Magdalena Schelotto, Matthew Johnson, Sven Olek, Christoph Sachsenmaier, Maria-Grazia Roncarolo, and Rosa Bacchetta
- Subjects
Immunology ,Immunology and Allergy - Abstract
Forkhead box protein 3 (FOXP3) is the master transcription factor in CD4We sought to study the type and extent of immunologic abnormalities that remain ill-defined in IPEX, across genetic and clinical heterogeneity.We performed Treg-cell-specific epigenetic quantification and immunologic characterization of severe "typical" (n = 6) and "atypical" or asymptomatic (n = 9) patients with IPEX.Increased number of cells with Treg-cell-Specific Demethylated Region demethylation in FOXP3 is a consistent feature in patients with IPEX, with (1) highest values in those with typical IPEX, (2) increased values in subjects with pathogenic FOXP3 but still no symptoms, and (3) gradual increase over the course of disease progression. Large-scale profiling using Luminex identified plasma inflammatory signature of macrophage activation and TElevated TSDR-demethylated cells, combined with elevation of plasmatic and cellular markers of a polarized type 2 inflammatory immune response, extends our understanding of IPEX diagnosis and heterogeneity.
- Published
- 2022
28. Engineered type 1 regulatory T cells designed for clinical use kill primary pediatric acute myeloid leukemia cells
- Author
-
Alice Bertaina, Silvia Gregori, Katharine Greenthal, Ece Canan Sayitoglu, Jeffrey Liu, Brandon Cieniewicz, Maria Grazia Roncarolo, Ping Pauline Chen, Alma-Martina Cepika, Norman J. Lacayo, Grazia Andolfi, Molly Javier Uyeda, and Rosa Bacchetta
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Myeloid ,Graft-vs-Leukemia Effect ,Myeloid leukemia ,Adult Acute Myeloid Leukemia ,Graft vs Leukemia Effect ,Hematology ,Biology ,medicine.disease ,T-Lymphocytes, Regulatory ,Article ,Translocation, Genetic ,Cell therapy ,Leukemia ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Immune system ,Cell culture ,hemic and lymphatic diseases ,medicine ,Cancer research ,Humans ,Child - Abstract
Type 1 regulatory (Tr1) T cells induced by enforced expression of interleukin-10 (LV-10) are being developed as a novel treatment for chemotherapy-resistant myeloid leukemias. In vivo, LV-10 cells do not cause graft-versus-host disease while mediating graft-versus-leukemia effect against adult acute myeloid leukemia (AML). Since pediatric AML (pAML) and adult AML are different on a genetic and epigenetic level, we investigate herein whether LV-10 cells also efficiently kill pAML cells. We show that the majority of primary pAML are killed by LV-10 cells, with different levels of sensitivity to killing. Transcriptionally, pAML sensitive to LV-10 killing expressed a myeloid maturation signature. Overlaying the signatures of sensitive and resistant pAML onto the public NCI TARGET pAML dataset revealed that sensitive pAML clustered with M5 monocytic pAML and pAML with MLL rearrangement. Resistant pAML clustered with myelomonocytic leukemias and those bearing the core binding factor translocations inv(16) or t(8;21)(RUNX1- RUNX1T1). Furthermore, resistant pAML upregulated the membrane glycoprotein CD200, which binds to the inhibitory receptor CD200R1 on LV-10 cells. In order to examine if CD200 expression on target cells can impair LV-10 cell function, we overexpressed CD200 in myeloid leukemia cell lines ordinarily sensitive to LV-10 killing. Indeed, LV-10 cells degranulated less and killed fewer CD200-overexpressing cells compared to controls, indicating that pAML can utilize CD200 expression for immune evasion. Altogether, the majority of pAML are killed by LV-10 cells in vitro, supporting further LV-10 cell development as an innovative cell therapy for pAML.
- Published
- 2020
29. Towards gene therapy for IPEX syndrome
- Author
-
Simon Borna, Esmond Lee, Yohei Sato, and Rosa Bacchetta
- Subjects
Diarrhea ,Diabetes Mellitus, Type 1 ,Immune System Diseases ,Immunology ,Mutation ,Immunology and Allergy ,Humans ,Forkhead Transcription Factors ,Genetic Diseases, X-Linked ,Genetic Therapy ,T-Lymphocytes, Regulatory - Abstract
Immune dysregulation polyendocrinopathy enteropathy X linked (IPEX) syndrome is an uncurable disease of the immune system, with immune dysregulation that is caused by mutations in FOXP3. Current treatment options, such as pharmacological immune suppression and allogeneic hematopoietic stem cell transplantation, have been beneficial but present limitations, and their life-long consequences are ill-defined. Other similar blood monogenic diseases have been successfully treated using gene transfer in autologous patient cells, thus providing an effective and less invasive therapeutic. Development of gene therapy for patients with IPEX is particularly challenging because successful strategies must restore the complex expression profile of the transcription factor FOXP3, ensuring it is tightly regulated and its cell subset-specific roles are maintained. This review summarizes current efforts toward achieving gene therapy to treat immune dysregulation in IPEX patients.
- Published
- 2022
30. Design of experiments as a decision tool for cell therapy manufacturing
- Author
-
Esmond Lee, Devin Shah, Matthew Porteus, J. Fraser Wright, and Rosa Bacchetta
- Subjects
Gene Editing ,Cancer Research ,Transplantation ,Oncology ,Immunology ,Cell- and Tissue-Based Therapy ,Immunology and Allergy ,Humans ,Cell Biology ,Prospective Studies ,CRISPR-Cas Systems ,Genetics (clinical) ,RNA, Guide, Kinetoplastida - Abstract
Cell therapies are costlier to manufacture than small molecules and protein therapeutics because they require multiple manipulations and are often produced in an autologous manner. Strategies to lower the cost of goods to produce a cell therapy could make a significant impact on its total cost.Borrowing from the field of bioprocess development, the authors took a design of experiments (DoE)-based approach to understanding the manufacture of a cell therapy product in pre-clinical development, analyzing main cost factors in the production process. The cells used for these studies were autologous CD4Using gene editing efficiency as the response variable, an initial screen was conducted for other variables that could influence the editing frequency. The multiplicity of infection (MOI) of AAV and amount of single guide RNA (sgRNA) were the significant factors used for the optimization step to generate a response contour plot. Cost analysis was done for multiple points in the design space to find cost drivers that could be reduced. For the range of values tested (50 000-750 000 vg/cell AAV and 0.8-4 μg sgRNA), editing with the highest MOI and sgRNA yielded the best gene editing frequency. However, cost analysis showed the optimal solution was gene editing at 193 000 vg/cell AAV and 1.78 μg sgRNA.The authors used DoE to define key factors affecting the gene editing process for a potential investigational therapeutic, providing a novel and faster data-based approach to understanding factors driving complex biological processes. This approach could be applied in process development and aid in achieving more robust strategies for the manufacture of cellular therapeutics.
- Published
- 2021
31. Phase 1/1b Study of T-allo10 Infusion after HLA-Partially Matched αβ depleted-HSCT in Children and Young Adults with Hematologic Malignancies: Preliminary Results
- Author
-
Alice Bertaina, Rosa Bacchetta, David C. Shyr, Gopin Saini, Karen Kristovich, Rajni Agarwal, Orly Klein, Kathryn Melsop, Keri Tate, Giulia Barbarito, Pauline Chen, Alma-Martina Cepika, and Maria Grazia Roncarolo
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
32. The Biology of T Regulatory Type 1 Cells and Their Therapeutic Application in Immune-Mediated Diseases
- Author
-
Manuela Battaglia, Silvia Gregori, Nicola Gagliani, Maria Grazia Roncarolo, and Rosa Bacchetta
- Subjects
0301 basic medicine ,Immunology ,Cell ,Population ,Cell- and Tissue-Based Therapy ,Receptors, Antigen, T-Cell ,Biology ,T-Lymphocytes, Regulatory ,Autoimmune Diseases ,03 medical and health sciences ,Immune system ,Antigen ,medicine ,Animals ,Humans ,Immunology and Allergy ,Antigens ,education ,education.field_of_study ,Effector ,Models, Immunological ,FOXP3 ,Cell biology ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Cytokines ,Function (biology) - Abstract
Thirty years ago, one of the first types of CD4+ T regulatory cells was discovered and named T regulatory type 1 (Tr1) cells. Tr1 cells represent a distinct population of T cells, which are induced in the periphery upon antigen exposure under tolerogenic conditions. They produce the immunosuppressive cytokines interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β), do not constitutively express FOXP3, and suppress the function of effector immune cells. In this review, the key studies leading to the identification and biological characterization of Tr1 cells are recapitulated. The fundamental role of Tr1 cells in regulating immune responses to pathogenic and non-pathogenic antigens, as well as their use as cell therapeutics, is summarized.
- Published
- 2018
33. Tregopathies: Monogenic diseases resulting in regulatory T-cell deficiency
- Author
-
Alma-Martina Cepika, Molly Javier Uyeda, Yohei Sato, Rosa Bacchetta, Maria Grazia Roncarolo, and Jeffrey Liu
- Subjects
0301 basic medicine ,Regulatory T cell ,Immunology ,chemical and pharmacologic phenomena ,Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,LRBA ,03 medical and health sciences ,Immune system ,medicine ,Animals ,Humans ,Immunology and Allergy ,Receptors, Interleukin-10 ,Immunologic Deficiency Syndromes ,FOXP3 ,Peripheral tolerance ,Forkhead Transcription Factors ,Immune dysregulation ,Acquired immune system ,Interleukin-10 ,030104 developmental biology ,medicine.anatomical_structure ,CTLA-4 - Abstract
Monogenic diseases of the immune system, also known as inborn errors of immunity, are caused by single-gene mutations resulting in immune deficiency and dysregulation. More than 350 diseases have been described to date, and the number is rapidly expanding, with increasing availability of next-generation sequencing facilitating the diagnosis. The spectrum of immune dysregulation is wide, encompassing deficiencies in humoral, cellular, innate, and adaptive immunity; phagocytosis; and the complement system, which lead to autoinflammation and autoimmunity. Multiorgan autoimmunity is a dominant symptom when genetic mutations lead to defects in molecules essential for the development, survival, and/or function of regulatory T (Treg) cells. Studies of "Tregopathies" are providing critical mechanistic information on Treg cell biology, the role of Treg cell-associated molecules, and regulation of peripheral tolerance in human subjects. The pathogenic immune networks underlying these diseases need to be dissected to apply and develop immunomodulatory treatments and design curative treatments using cell and gene therapy. Here we review the pathogenetic mechanisms, clinical presentation, diagnosis, and current and future treatments of major known Tregopathies caused by mutations in FOXP3, CD25, cytotoxic T lymphocyte-associated antigen 4 (CTLA4), LPS-responsive and beige-like anchor protein (LRBA), and BTB domain and CNC homolog 2 (BACH2) and gain-of-function mutations in signal transducer and activator of transcription 3 (STAT3). We also discuss deficiencies in genes encoding STAT5b and IL-10 or IL-10 receptor as potential Tregopathies.
- Published
- 2018
34. Pre-clinical development and molecular characterization of an engineered type 1 regulatory T-cell product suitable for immunotherapy
- Author
-
Jeffrey Liu, Benjamin Craig Thomas, Ece Canan Sayitoglu, Rosa Bacchetta, Ping Chen, Brandon Cieniewicz, Yohei Sato, Alma-Martina Cepika, Maria Grazia Roncarolo, and Molly Javier Uyeda
- Subjects
CD4-Positive T-Lymphocytes ,Cancer Research ,Myeloid ,medicine.medical_treatment ,Immunology ,Graft vs Host Disease ,Graft vs Leukemia Effect ,T-Lymphocytes, Regulatory ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Transplantation ,Type 1 Regulatory T-Cell ,business.industry ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Immunotherapy ,medicine.disease ,3. Good health ,Interleukin 10 ,surgical procedures, operative ,Graft-versus-host disease ,medicine.anatomical_structure ,Oncology ,Cancer research ,business ,CD8 ,030215 immunology - Abstract
Background aims Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a curative therapeutic approach for many hematological disorders. However, allo-HSCT is frequently accompanied by a serious side effect: graft-versus-host disease (GVHD). The clinical use of allo-HSCT is limited by the inability of current immunosuppressive regimens to adequately control GvHD without impairing the graft-versus-leukemia effect (GvL) conferred by transplanted healthy immune cells. To address this, the authors have developed an engineered type 1 regulatory T-cell product called CD4IL-10 cells. CD4IL-10 cells are obtained through lentiviral transduction, which delivers the human IL10 gene into purified polyclonal CD4+ T cells. CD4IL-10 cells may provide an advantage over standard-of-care immunosuppressants because of the ability to suppress GvHD through continuous secretion of IL-10 and enhance the GvL effect in myeloid malignancies through targeted killing of malignant myeloid cells. Methods Here the authors established a production process aimed at current Good Manufacturing Practice (cGMP) production for CD4IL-10 cells. Results The authors demonstrated that the CD4IL-10 cell product maintains the suppressive and cytotoxic functions of previously described CD4IL-10 cells. In addition, RNA sequencing analysis of CD4IL-10 identified novel transcriptome changes, indicating that CD4IL-10 cells primarily upregulate cytotoxicity-related genes. These include four molecules with described roles in CD8+ T and natural killer cell-mediated cytotoxicity: CD244, KLRD1, KLRC1 and FASLG. Finally, it was shown that CD4IL-10 cells upregulate IL-22, which mediates wound healing and tissue repair, particularly in the gut. Conclusions Collectively, these results pave the way toward clinical translation of the cGMP-optimized CD4IL-10 cell product and uncover new molecules that have a role in the clinical application of CD4IL-10 cells.
- Published
- 2020
35. 146 Alloantigen-specific Tr1 cells designed to prevent GvHD have a distinct molecular identity and suppress through CTLA-4 and PD-1
- Author
-
Alice Bertaina, Laura Amaya, Maria Grazia Roncarolo, Alma-Martina Cepika, Rajni Agarwal-Hashmi, Mark M. Davis, David M. Louis, Rosa Bacchetta, Mansi Narula, Everett Meyer, Molly Javier Uyeda, Liwen Xu, Pauline P. Chen, Brandon Cieniewicz, and Xuhuai Ji
- Subjects
LAG3 ,medicine.medical_treatment ,T cell ,FOXP3 ,Biology ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,CD49b ,Cell therapy ,Cytokine ,medicine.anatomical_structure ,Antigen ,CTLA-4 ,Cancer research ,medicine - Abstract
Background Graft-vs-host-disease (GvHD) is a life-threatening complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT), limiting the use of this potentially curative treatment for hematological malignancies. To address this, we have developed T-allo10 cell therapy, which is enriched with type 1 regulatory T (Tr1) cells. Tr1 cells are peripherally inducible, CD49b+LAG3+IL-10+FOXP3- regulatory T cells that can confer alloantigen-specific tolerance, making them an attractive alternative to existing GvHD therapies, which non-discriminately impair both GvHD and protective immunity. T-allo10 cells are currently being evaluated in a phase I clinical trial in patients with hematological malignancies undergoing allo-HSCT (NCT03198234). Herein, we aimed to confirm that Tr1 cells are the active ingredient responsible for the T-allo10 suppressive function, and reveal the underlying molecular signatures to elucidate the mechanisms of Tr1 cell-mediated suppression. Methods T-allo10 cells were generated in a co-culture of healthy host or patient tolerogenic dendritic cells (DC-10) with allogeneic healthy donor CD4+ T cells, then tested for Tr1 phenotype, anergy, suppression and cytokine production. Sorted T-allo10-derived Tr1 cells and non-Tr1 cells, as well as control effector T cells (Teff) and parental CD4+ T cells, were analyzed by TCR- and RNA-seq. Protein expression for key differentially expressed genes were validated, and the functional roles for IL-10, CTLA-4 and PD-1 in T-allo10-mediated suppression were confirmed in a suppression assay. Results We show that the T-allo10 cell product is: i) enriched for Tr1 cells, ii) anergic in response to alloantigen re-challenge, but not to non-specific stimuli or 3rd party antigens, and iii) suppresses host-reactive T cells, but not T cell responses to other antigens. Furthermore, T-allo10-derived, isolated Tr1 cells had a restricted TCR repertoire, suggesting they clonally expand in response to alloantigens. T-allo10-derived Tr1 cells have a distinct signature compared to non-Tr1 cells, and, in addition to IL-10, express high levels of CTLA-4 and PD-1 (but not FOXP3). Notably, blockade of CTLA-4 and the PD-1 pathway completely abolishes T-allo10-mediated suppression of T cell responses. Conclusions Our data shows that Tr1 cells are the active, suppressive, and antigen-specific ingredient of T-allo10 cells. Furthermore, while the role of IL-10 in Tr1 cell-mediated suppression is well known, we uncover that Tr1 suppress in addition through CTLA-4 and PD-1. Collectively, these intriguing findings underscore the importance of CTLA-4 and PD-1 pathways in conferring cell-mediated immunological tolerance. Further, they define the key characteristics and modes of action of antigen-specific Tr1 cells, providing crucial information for the ongoing T-all10 trial and future design of novel Tr1 cell-based therapies. Ethics Approval The patient study was approved by Administrative Panels on Human Subjects in Medical Research, Stanford University, Tallo10 eProtocol # 38734. Healthy donor samples were purchased as deidentified human blood products from the Stanford Blood Center, and are thus exempt.
- Published
- 2020
36. Thymic origins of autoimmunity-lessons from inborn errors of immunity
- Author
-
Rosa Bacchetta and Kenneth I. Weinberg
- Subjects
Regulatory T cell ,Autoimmune regulator (AIRE) ,Immunology ,Receptors, Antigen, T-Cell ,Autoimmunity ,Review ,Primary immune deficiencies (PID) ,Biology ,Major histocompatibility complex ,T-Lymphocytes, Regulatory ,Clonal deletion ,Mice ,Immune system ,Forkhead box P3 (FOXP3) ,medicine ,Immunology and Allergy ,Animals ,Humans ,Transcription factor ,Effector ,T-cell receptor ,FOXP3 ,Epithelial Cells ,Forkhead Transcription Factors ,Cell biology ,medicine.anatomical_structure ,Primary immune regulatory diseases (PIRD) ,biology.protein ,Medullary thymic epithelial cell (mTEC) ,Thymic Regulatory T cells (tTreg) - Abstract
During their intrathymic development, nascent T cells are empowered to protect against pathogens and to be operative for a life-long acceptance of self. While autoreactive effector T (Teff) cell progenitors are eliminated by clonal deletion, the intrathymic mechanisms by which thymic regulatory T cell (tTreg) progenitors maintain specificity for self-antigens but escape deletion to exert their regulatory functions are less well understood. Both tTreg and Teff development and selection result from finely coordinated interactions between their clonotypic T cell receptors (TCR) and peptide/MHC complexes expressed by antigen-presenting cells, such as thymic epithelial cells and thymic dendritic cells. tTreg function is dependent on expression of the FOXP3 transcription factor, and induction of FOXP3 gene expression by tTreg occurs during their thymic development, particularly within the thymic medulla. While initial expression of FOXP3 is downstream of TCR activation, constitutive expression is fixed by interactions with various transcription factors that are regulated by other extracellular signals like TCR and cytokines, leading to epigenetic modification of the FOXP3 gene. Most of the understanding of the molecular events underlying tTreg generation is based on studies of murine models, whereas gaining similar insight in the human system has been very challenging. In this review, we will elucidate how inborn errors of immunity illuminate the critical non-redundant roles of certain molecules during tTreg development, shedding light on how their abnormal development and function cause well-defined diseases that manifest with autoimmunity alone or are associated with states of immune deficiency and autoinflammation.
- Published
- 2020
37. Human inborn errors of immunity: An expanding universe
- Author
-
Luigi D. Notarangelo, Jean-Laurent Casanova, Rosa Bacchetta, and Helen C. Su
- Subjects
0301 basic medicine ,Extramural ,Immunology ,Genetic Diseases, Inborn ,General Medicine ,Biology ,Immunity, Innate ,Article ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immunity ,030220 oncology & carcinogenesis ,Animals ,Humans ,Neuroscience - Abstract
Molecular, cellular, and clinical studies of human inborn errors of immunity have revolutionized our understanding of their pathogenesis, considerably broadened their spectrum of immunological and clinical phenotypes, and enabled successful targeted therapeutic interventions. These studies have also been of great scientific merit, challenging a number of immunological notions initially established in inbred mice, while revealing previously unrecognized mechanisms of host defense by leukocytes and other cells, and of both innate and adaptive tolerance to self.
- Published
- 2020
38. CRISPR-based gene editing enables FOXP3 gene repair in IPEX patient cells
- Author
-
Ciaran M. Lee, Gang Bao, C. S. Bauer, M. Garcia-Lloret, Ayal Hendel, Adam Sheikali, Rosa Bacchetta, Ami J. Shah, Federica Barzaghi, Alice Bertaina, S. Shipp, Maria Grazia Roncarolo, L. Froessl, Mara Pavel-Dinu, Matt Porteus, U. Lakshmanan, Mansi Narula, Esmond Lee, Holly K. Miller, Manish J. Butte, Marianne Goodwin, and Laura Passerini
- Subjects
T-Lymphocytes ,Regenerative Medicine ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,0302 clinical medicine ,Genome editing ,2.1 Biological and endogenous factors ,CRISPR ,Aetiology ,Child ,Research Articles ,Pediatric ,Gene Editing ,0303 health sciences ,Multidisciplinary ,Effector ,Gene correction ,SciAdv r-articles ,FOXP3 ,Forkhead Transcription Factors ,Genetic Diseases, X-Linked ,Regulatory ,Haematopoiesis ,Phenotype ,Genetic Diseases ,030220 oncology & carcinogenesis ,IPEX syndrome ,Development of treatments and therapeutic interventions ,Biotechnology ,Research Article ,Immune regulation ,Immunology ,Biology ,Autoimmune Disease ,03 medical and health sciences ,Rare Diseases ,Genetics ,medicine ,Humans ,Gene ,030304 developmental biology ,5.2 Cellular and gene therapies ,Inflammatory and immune system ,X-Linked ,Immune dysregulation ,Stem Cell Research ,medicine.disease ,Mutation ,Cancer research ,Forkhead box protein 3 gene - Abstract
Gene editing of FOXP3 ensures regulated expression and restored function in T cells, supporting clinical applicability., The prototypical genetic autoimmune disease is immune dysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome, a severe pediatric disease with limited treatment options. IPEX syndrome is caused by mutations in the forkhead box protein 3 (FOXP3) gene, which plays a critical role in immune regulation. As a monogenic disease, IPEX is an ideal candidate for a therapeutic approach in which autologous hematopoietic stem and progenitor (HSPC) cells or T cells are gene edited ex vivo and reinfused. Here, we describe a CRISPR-based gene correction permitting regulated expression of FOXP3 protein. We demonstrate that gene editing preserves HSPC differentiation potential, and that edited regulatory and effector T cells maintain their in vitro phenotype and function. Additionally, we show that this strategy is suitable for IPEX patient cells with diverse mutations. These results demonstrate the feasibility of gene correction, which will be instrumental for the development of therapeutic approaches for other genetic autoimmune diseases.
- Published
- 2020
39. Human‐engineered Treg‐like cells suppress FOXP3‐deficient T cells but preserve adaptive immune responses in vivo
- Author
-
Maria Grazia Roncarolo, Michael Snyder, Alice Bertaina, Silvia Gregori, Yohei Sato, Laura Passerini, Rosa Bacchetta, Molly Javier Uyeda, Marianne Goodwin, and Brian D. Piening
- Subjects
0301 basic medicine ,FOXP3 ,Genetic enhancement ,Immunology ,chemical and pharmacologic phenomena ,Biology ,medicine.disease_cause ,regulatory T cells ,Flow cytometry ,Viral vector ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,medicine ,Immunology and Allergy ,CRISPR/Cas9 ,General Nursing ,medicine.diagnostic_test ,lentiviral vector ,Original Articles ,IPEX syndrome ,Immune dysregulation ,medicine.disease ,gene therapy ,030104 developmental biology ,030220 oncology & carcinogenesis ,Original Article - Abstract
Objectives Genetic or acquired defects in FOXP3+ regulatory T cells (Tregs) play a key role in many immune‐mediated diseases including immune dysregulation polyendocrinopathy, enteropathy, X‐linked (IPEX) syndrome. Previously, we demonstrated CD4+ T cells from healthy donors and IPEX patients can be converted into functional Treg‐like cells by lentiviral transfer of FOXP3 (CD4LVFOXP3). These CD4LVFOXP3 cells have potent regulatory function, suggesting their potential as an innovative therapeutic. Here, we present molecular and preclinical in vivo data supporting CD4LVFOXP3 cell clinical progression. Methods The molecular characterisation of CD4LVFOXP3 cells included flow cytometry, qPCR, RNA‐seq and TCR‐seq. The in vivo suppressive function of CD4LVFOXP3 cells was assessed in xenograft‐versus‐host disease (xeno‐GvHD) and FOXP3‐deficient IPEX‐like humanised mouse models. The safety of CD4LVFOXP3 cells was evaluated using peripheral blood (PB) humanised (hu)‐ mice testing their impact on immune response against pathogens, and immune surveillance against tumor antigens. Results We demonstrate that the conversion of CD4+ T cells to CD4LVFOXP3 cells leads to specific transcriptional changes as compared to CD4+ T‐cell transduction in the absence of FOXP3, including upregulation of Treg‐related genes. Furthermore, we observe specific preservation of a polyclonal TCR repertoire during in vitro cell production. Both allogeneic and autologous CD4LVFOXP3 cells protect from xeno‐GvHD after two sequential infusions of effector T cells. CD4LVFOXP3 cells prevent hyper‐proliferation of CD4+ memory T cells in the FOXP3‐deficient IPEX‐like hu‐mice. CD4LVFOXP3 cells do not impede in vivo expansion of antigen‐primed T cells or tumor clearance in the PB hu‐mice. Conclusion These data support the clinical readiness of CD4LVFOXP3 cells to treat IPEX syndrome and other immune‐mediated diseases caused by insufficient or dysfunctional FOXP3+ Tregs., In this study, we present novel molecular and preclinical in vivo data that support CD4LVFOXP3 clinical progression. These data support the clinical readiness of CD4LVFOXP3 to treat immune‐mediated diseases caused by insufficient or dysfunctional FOXP3+ Tregs.
- Published
- 2020
40. Neutralizing Anti-Cytokine Autoantibodies Against Interferon-α in IPEX
- Author
-
Jacob Rosenberg, Maria E. Maccari, Federica Barzaghi, Eric J. Allenspach, Claudio Pignata, Giovanna Weber, Troy R. Torgerson, Paul J. Utz and Rosa Bacchetta., Rosenberg, Jacob, Maccari, Maria E., Barzaghi, Federica, Allenspach, Eric J., Pignata, Claudio, Weber, Giovanna, Torgerson, Troy R., and J. Utz and Rosa Bacchetta., Paul
- Subjects
anti-cytokine autoantibodies, Interferon-alpha, IPEX, APS-1, Protein Microarrays - Abstract
Anti-cytokine autoantibodies (ACAAs) have been described in a growing number of primary immunodeficiencies with autoimmune features including Autoimmune Polyendocrine Syndrome Type I (APS-1), a prototypical disease of defective T cell central tolerance. Whether defects in peripheral tolerance lead to similar ACAAs is unknown. Immunodysregulation Polyendocrinopathy Enteropathy X-linked (IPEX) is caused by mutations in FOXP3, a master regulator of T regulatory cells (Treg), and consequently results in defective T cell peripheral tolerance. Unique autoantibodies have previously been described in IPEX. To test the hypothesis that ACAAs are present in IPEX, we designed and fabricated antigen microarrays. We discovered elevated levels of IgG ACAAs against interferon-alpha (IFN-alpha) in a cohort of IPEX patients. Serum from IPEX patients blocked IFN-alpha signaling in vitro, and blocking activity was tightly correlated with ACAA titer. To show that blocking activity was mediated by IgG and not other serum factors, we purified IgG and showed that blocking activity was contained entirely in the immunoglobulin fraction. We also screened for ACAAs against IFN-alpha in a second geographically distinct cohort. In these samples, ACAAs against IFN-alpha were elevated in a post-hoc analysis. In summary, we report the discovery of ACAAs against IFN-alpha in IPEX, an experiment of nature demonstrating the important role of peripheral T cell tolerance in preventing the development of IFN-alpha ACAAs.
- Published
- 2018
41. 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
42. Severe Toxoplasma gondii infection in a member of a NFKB2-deficient family with T and B cell dysfunction
- Author
-
Jacob M. Rosenberg, Marco Cappa, Annalisa Deodati, Maria Chiriaco, F. Angelini, Paul J. Utz, Andrea Angius, Silvia Di Cesare, Giovanna Stefania Colafati, S Corrente, Matteo Floris, Caterina Cancrini, Maria Elena Maccari, Alessandra Fierabracci, Paolo Rossi, Alessandro Aiuti, Alessia Scarselli, Rita Carsetti, Rosa Bacchetta, Paola Cambiaso, Maccari, Maria-Elena, Scarselli, Alessia, Di Cesare, Silvia, Floris, Matteo, Angius, Andrea, Deodati, Annalisa, Chiriaco, Maria, Cambiaso, Paola, Corrente, Stefania, Colafati, Giovanna Stefania, Utz, Paul J., Angelini, Federica, Fierabracci, Alessandra, Aiuti, Alessandro, Carsetti, Rita, Rosenberg, Jacob M., Cappa, Marco, Rossi, Paolo, Bacchetta, Rosa, and Cancrini, Caterina
- Subjects
0301 basic medicine ,B cell defect ,Immunology ,Immune-dysregulation ,NFkB2 ,STAT5A ,T cell defect ,Toxoplasmosis ,medicine.disease_cause ,Toxoplasmosi ,03 medical and health sciences ,medicine ,Immunology and Allergy ,B cell ,Settore MED/38 - Pediatria Generale e Specialistica ,biology ,business.industry ,Toxoplasma gondii ,Immune dysregulation ,biology.organism_classification ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,business - Published
- 2017
43. Regulatory Type 1 T Cell Infusion in Mismatched Related or Unrelated Hematopoietic Stem Cell Transplantation (HSCT) for Hematologic Malignancies
- Author
-
Julia Chu, Neehar Bhatia, Alice Bertaina, Rajni Agarwal, Maria Grazia Roncarolo, Rosa Bacchetta, Gopin Saini, and Pauline Chen
- Subjects
Transplantation ,LAG3 ,Regulatory T cell ,business.industry ,medicine.medical_treatment ,T cell ,T-cell receptor ,Hematopoietic stem cell ,Hematopoietic stem cell transplantation ,Hematology ,Calcineurin ,medicine.anatomical_structure ,Sirolimus ,Immunology ,medicine ,business ,medicine.drug - Abstract
A major complication of mismatched unmanipulated hematopoietic stem cell transplant (HSCT) is Graft-versus-Host Disease (GvHD), which results in significant morbidity and increased non-relapse mortality. Novel strategies to reduce GvHD and improve long-term tolerance between mismatched donor-host pairs include regulatory T cell therapy. We are investigating a new cell product named T-allo10 which contains suppressive anergic cells and is enriched in IL-10 producing regulatory type 1 T cells (Tr1). The main advantage of adoptive immunotherapy with Tr1 compared to other regulatory T cells is the host alloantigen specificity that is established in the donor Tr1 during in vitro culture in the presence of IL-10 and host tolerogenic dendritic cells. Here we report the preliminary results of our phase I trial (IND 17292) on the use of escalating doses of T-allo10 cells for patients aged 3-45 years affected by hematological malignancies. At present, we completed the first cohort of the phase I portion of the study (Table 1). The donors were class I mismatched for patient 1 and class II HLA-mismatched for patients 2 and 3, respectively. The GvHD prophylaxis consisted of Sirolimus and Mycophenolate and was serotherapy and Calcineurin inhibitory-free. Patients received 1 × 106 T-allo10 cells/Kg on Day-1. No adverse events were observed after the T-allo10 cell infusion. All 3 patients met the safety criteria and are alive and disease-free at 2 years, 15 months and 2 months post-HSCT, respectively. Tr1, phenotypically defined as CD4+CD45RA−CD49b+LAG3+ were detectable in the peripheral blood shortly after the infusion at 21% and 26% in patient 2 and 3, respectively. At day 28 post-HSCT, Tr1 represented 12% of circulating memory CD4+ T cells in patient 1, 3% in patient 2 and 2% in patient 3. TCR sequencing of T-allo10 cells prior and after infusion showed a restricted clonotype diversity with persistence of certain clonotypes in vivo, demonstrating Tr1 cell recirculation and survival. These data show that the T-allo10 cell infusion is safe and well tolerated and demonstrate that T-allo10 cells are detectable in the recipients after the infusion and traceable by TCR clonotype analysis.
- Published
- 2020
44. CD4+ T Regulatory Cells and Modulation of Undesired Immune Responses
- Author
-
Maria-Grazia Roncarolo, Megan K. Levings, and Rosa Bacchetta
- Subjects
Immune system ,Modulation ,Chemistry ,Cell biology - Published
- 2019
45. A reprogramming human T cell function and specificity with non-viral genome targeting
- Author
-
Michael Haugwitz, orell M, May Ap, el Gaudio D, Paige Krystofinski, Lee Mr, Amy L. Putnam, Gorka Alkorta-Aranburu, Theodore L. Roth, Li Pj, Eric Meffre, Julia Carnevale, Laurence Pellerin, Greeley Saw, Maria Grazia Roncarolo, David Carmody, David N. Nguyen, Leonetti, Mao Y, Channabasavaiah B. Gurumurthy, Justin Saco, shworth A, Cristina Puig-Saus, Alexander Marson, Cho M, Rolen M. Quadros, Eric Shifrut, Rosa Bacchetta, Kevan C. Herold, Hiatt, Tobin, Jean-Nicolas Schickel, Jonathan S. Weissman, Chen Jw, Yu R, Jonathan H. Esensten, Antoni Ribas, Andrea L. Ferris, Neil Romberg, Matsumoto H, Kathrin Schumann, Gary M. Kupfer, Baz Smith, Hang Li, and Stephen H. Hughes
- Subjects
medicine.anatomical_structure ,T cell ,medicine ,Biology ,Reprogramming ,Genome ,Function (biology) ,Cell biology - Published
- 2019
46. Case Study: Mechanism for Increased Follicular Helper T Cell Development in Activated PI3K Delta Syndrome
- Author
-
Robert S. Ohgami, Rosa Bacchetta, Manish J. Butte, Laurence Pellerin, and Timothy J. Thauland
- Subjects
0301 basic medicine ,osteopontin ,Helper-Inducer ,Protein Conformation ,T-Lymphocytes ,activated PI3K delta syndrome ,Case Report ,medicine.disease_cause ,PI3K ,Autoimmunity ,0302 clinical medicine ,T-Lymphocyte Subsets ,Models ,Immunology and Allergy ,2.1 Biological and endogenous factors ,Osteopontin ,Aetiology ,Child ,B-Lymphocytes ,biology ,Chemistry ,activated PI3K delta syndrome (APDS) ,3. Good health ,Pedigree ,medicine.anatomical_structure ,Medical Microbiology ,Female ,lcsh:Immunologic diseases. Allergy ,Adolescent ,Class I Phosphatidylinositol 3-Kinases ,T cell ,Primary Immunodeficiency Diseases ,Immunology ,Activated PI3K-delta syndrome ,primary immunodeficiency ,Immunophenotyping ,03 medical and health sciences ,Structure-Activity Relationship ,medicine ,Humans ,PI3K/AKT/mTOR pathway ,Alleles ,Inflammatory and immune system ,Molecular ,medicine.disease ,Biological ,CD4 Lymphocyte Count ,030104 developmental biology ,Amino Acid Substitution ,P110δ ,T cell differentiation ,Mutation ,Primary immunodeficiency ,biology.protein ,T follicular helper cells ,lcsh:RC581-607 ,030215 immunology - Abstract
Gain-of-function variants in p110δ, the catalytic subunit of phosphatidylinositol 3-kinase (PI3K) expressed in lymphocytes, cause activated PI3-kinase δ syndrome (APDS), a primary immunodeficiency that is characterized by recurrent infections, viremia, lymphadenopathy, and autoimmunity. The mechanism of autoimmunity in APDS has not been well-understood. Here, we show the profound skewing of peripheral CD4+ T cells to a T follicular helper (TFH) phenotype in a patient with APDS bearing a novel p110δ variant, Y524S. We also saw a diminishment of transient Foxp3 expression in activated T cells. Mechanistic studies revealed that both the new variant and a previously described, pathogenic variant (E81K) enhanced an interaction between intracellular Osteopontin and p85α. This interaction had been shown in mice to promote TFH differentiation. Our results demonstrate a new influence of PI3K on human T cell differentiation that is unrelated to its lipid-kinase activity and suggest that TFH should be monitored in APDS patients.
- Published
- 2019
47. The autoimmune targets in IPEX are dominated by gut epithelial proteins
- Author
-
Stephan Ehl, Olle Kämpe, Olov Ekwall, Outi Mäkitie, Eystein S. Husebye, Anders Rönnblom, Sophie Bensing, Federica Barzaghi, Nils Landegren, Fabian Sardh, Hörður Ingi Gunnarsson, Rosa Bacchetta, Frédéric Van Gool, Saila Laakso, Åsa Hallgren, Mark S. Anderson, Christina Lundqvist, Alice Y. Chan, and Daniel Eriksson
- Subjects
Adult ,Diarrhea ,Adolescent ,Respiratory Medicine and Allergy ,Immunology ,MEDLINE ,Autoantigens ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Diabetes mellitus ,medicine ,Humans ,Immunology and Allergy ,Intestinal Mucosa ,Young adult ,Child ,Autoantibodies ,030304 developmental biology ,Lungmedicin och allergi ,0303 health sciences ,business.industry ,Infant, Newborn ,Infant ,Proteins ,Immunology in the medical area ,Genetic Diseases, X-Linked ,medicine.disease ,Diabetes Mellitus, Type 1 ,Immune System Diseases ,Child, Preschool ,030220 oncology & carcinogenesis ,Immunologi inom det medicinska området ,business - Published
- 2019
48. From IPEX syndrome toFOXP3mutation: a lesson on immune dysregulation
- Author
-
Maria Grazia Roncarolo, Federica Barzaghi, and Rosa Bacchetta
- Subjects
0301 basic medicine ,business.industry ,General Neuroscience ,FOXP3 ,Disease ,Immune dysregulation ,IPEX syndrome ,Autoimmune enteropathy ,medicine.disease_cause ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Autoimmunity ,03 medical and health sciences ,030104 developmental biology ,History and Philosophy of Science ,Immunology ,medicine ,Primary immunodeficiency ,Enteropathy ,business - Abstract
Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a rare disorder that increasingly has gained attention as a model of genetic autoimmunity. Numerous papers documenting the key clinical and molecular characteristics of IPEX have provided a detailed understanding of this devastating disease. IPEX is a primary immunodeficiency caused by mutations in the gene FOXP3, which encodes an essential transcription factor required for maintenance of thymus-derived regulatory T (tTreg) cells. tTreg cell dysfunction is the main pathogenic event leading to multiorgan autoimmunity in IPEX. In addition to the traditional clinical presentation (i.e., severe enteropathy, type 1 diabetes, and eczema), IPEX may encompass other variable and distinct clinical manifestations. As IPEX awareness and characterization have increased, so has identification of FOXP3 mutations, with at least 70 to date. Thus, while FOXP3 is the unifying gene, IPEX is a complex and diverse clinical continuum of disorders. Despite understanding IPEX pathogenesis, new treatment options have remained elusive, although early diagnosis led to hematopoietic stem cell transplantation (HSCT) and immunosuppression treatment and improved patient outcomes. Here, we review current knowledge about IPEX syndrome and highlight findings that could lead to novel targeted treatments.
- Published
- 2016
49. Gene editing using CRISPR enables FOXP3 gene repair in HSPCs and IPEX patient T cells
- Author
-
S. Shipp, Matthew H. Porteus, Esmond Lee, Maria Grazia Roncarolo, U. Lakshmanan, Rosa Bacchetta, Marianne Goodwin, and Mara Pavel-Dinu
- Subjects
Cancer Research ,Transplantation ,business.industry ,Immunology ,FOXP3 ,chemical and pharmacologic phenomena ,Cell Biology ,IPEX syndrome ,medicine.disease ,Haematopoiesis ,Immune system ,Oncology ,Cord blood ,medicine ,Immunology and Allergy ,IL-2 receptor ,Stem cell ,Progenitor cell ,business ,Genetics (clinical) - Abstract
Background & Aim Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is the prototypical primary immune regulatory disorder caused by mutations in the forkhead box protein 3 (FOXP3) gene, a critical transcription factor required for regulatory T cells (Tregs). Patients typically present with severe early onset autoimmune manifestations that can be fatal within the first year of life. Currently, the only cure for IPEX is allogeneic hematopoietic stem cell transplant (HSCT), but this comes with risk of complications and the survival probability at 15 years is 73.2% (F. Barzaghi et al 2018). Moreover, a suitable donor is not always available. As a monogenic immune disease with limited treatment options, IPEX is an ideal candidate for a gene therapy approach whereby patient hematopoietic stem cells are gene edited and reinfused for autologous transplant. Methods, Results & Conclusion We developed a CRISPR/Cas9 approach combined with AAV delivery of a donor template to restore FOXP3 expression at the endogenous locus, permitting regulated expression of wild-type FOXP3 irrespective of downstream mutations. We were able to edit CD4+ effector T cells (Teff) and Tregs, showing that they maintain characteristic phenotypic markers and are functional in vitro. IPEX patient Tregs edited with the construct had partial restoration of both FOXP3 expression and functional suppression upon co-culture with responder T cells. Gene edited cord blood hematopoietic stem and progenitor Cells (HSPCs) are capable of long-term engraftment in humanized mice and differentiate into multiple hematopoietic subsets (CD13, CD19, CD56, CD3) and partially reconstitute Tregs (CD4+CD25+FOXP3+). These results demonstrate the feasibility of using gene edited HSPCs and hold promise for autologous transplant in genetic autoimmune diseases such as IPEX syndrome.
- Published
- 2020
50. Engineered Type-1 Regulatory T Cells as Cellular Therapy for Treatment of Immune Mediated Diseases
- Author
-
Jeffrey M Liu, Pauline Chen, Brandon Cieniewicz, Alma-Martina Cepika, Rosa Bacchetta, and Maria Grazia Roncarolo
- Subjects
Immunology ,Immunology and Allergy - Abstract
Type 1 regulatory cells (Tr1) are a promising cellular product for suppression of effector T cells in immune mediated diseases, including graft-versus-host-disease (GvHD) in allogeneic hematopoietic stem cell transplantation (allo-HSCT) (Roncarolo et al. Immunity 2018). We have developed an in vitro protocol to produce Tr1 cells by lentiviral transduction of the human IL10 with a constitutive promoter into human CD4+ T cells (Locafaro et al. Molecular Therapy 2017). These engineered Tr1 cells, called LV-10, acquire a characteristic Tr1 cytokine profile (IL-10 and IFN-g high, IL-4 low and expression of intracellular perforin and granzyme B). In vitro, LV-10 cells suppress the proliferation of responder CD4+ T cells upon activation by allogeneic dendritic cells. LV-10 cells also degranulate in response to and kill myeloid cells, including myeloid blasts from patients with acute myeloid leukemia, through a granzyme B- and perforin-dependent mechanism. Interestingly, the ability to degranulate and kill myeloid cells is not present when LV-10 are activated and expanded with anti-CD3 and anti-CD28 coated beads, suggesting that signals beyond TCR, CD28, and IL-10 receptor pathway activation are necessary to reprogram LV-10 cells into cytotoxic cells. In vivo, LV-10 cells injected into NSG mice do not induce xeno-GvHD, in contrast to control CD4+ T cells. In addition, LV-10 cells suppress CD4-induced xeno-GvHD and prevent expansion of myeloid leukemic cells. Experiments are ongoing to compare the potency and in vivo survival of allogeneic vs autologous LV-10 cells. These findings demonstrate the promise of using LV-10 to treat immune mediated diseases, including GvHD in AML patients receiving allo-HSCT.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.