39 results on '"Roberta E. Parrott"'
Search Results
2. Mesenchymal stromal cells reprogram monocytes and macrophages with processing bodies
- Author
-
Anthony J. Filiano, Hyunjung Min, Roberta E. Parrott, Rishi R. Rampersad, N. Meadows, Alban Gaultier, Emily M. Rabjohns, Christopher C. Overall, Joanne Kurtzberg, Anthony Fernandez-Castaneda, Li Xu, Teresa K. Tarrant, and M. Lillich
- Subjects
0301 basic medicine ,T cell ,Antigen presentation ,Inflammation ,Biology ,Mesenchymal Stem Cell Transplantation ,Monocytes ,Cell therapy ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Animals ,Humans ,Macrophages ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Cell Biology ,Cellular Reprogramming ,In vitro ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Heterografts ,Molecular Medicine ,medicine.symptom ,Reprogramming ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Mesenchymal stromal cells (MSCs) are widely used in clinical trials because of their ability to modulate inflammation. The success of MSCs has been variable over 25 years, most likely due to an incomplete understanding of their mechanism. After MSCs are injected, they traffic to the lungs and other tissues where they are rapidly cleared. Despite being cleared, MSCs suppress the inflammatory response in the long term. Using human cord tissue-derived MSCs (hCT-MSCs), we demonstrated that hCT-MSCs directly interact and reprogram monocytes and macrophages. After engaging hCT-MSCs, monocytes and macrophages engulfed cytoplasmic components of live hCT-MSCs, then downregulated gene programs for antigen presentation and costimulation, and functionally suppressed the activation of helper T cells. We determined that low-density lipoprotein receptor-related proteins on monocytes and macrophages mediated the engulfment of hCT-MSCs. Since a large amount of cellular information can be packaged in cytoplasmic RNA processing bodies (p-bodies), we generated p-body deficient hCT-MSCs and confirmed that they failed to reprogram monocytes and macrophages in vitro and in vivo. hCT-MSCs suppressed an inflammatory response caused by a nasal lipopolysaccharide challenge. Although both control and p-body deficient hCT-MSCs were engulfed by infiltrating lung monocytes and macrophages, p-body deficient hCT-MSCs failed to suppress inflammation and downregulate MHC-II. Overall, we identified a novel mechanism by which hCT-MSCs indirectly suppressed a T-cell response by directly interacting and reprogramming monocytes and macrophages via p-bodies. The results of this study suggest a novel mechanism for how MSCs can reprogram the inflammatory response and have long-term effects to suppress inflammation.
- Published
- 2020
- Full Text
- View/download PDF
3. Aberrant T-cell exhaustion in severe combined immunodeficiency survivors with poor T-cell reconstitution after transplantation
- Author
-
Roxane Labrosse, Ines Boufaied, Benoîte Bourdin, Saideep Gona, Haley E. Randolph, Brent R. Logan, Sara Bourbonnais, Chloé Berthe, Wendy Chan, Rebecca H. Buckley, Roberta E. Parrott, Geoffrey D.E. Cuvelier, Neena Kapoor, Sharat Chandra, Blachy J. Dávila Saldaña, Hesham Eissa, Fred D. Goldman, Jennifer Heimall, Richard O’Reilly, Sonali Chaudhury, Edward A. Kolb, Shalini Shenoy, Linda M. Griffith, Michael Pulsipher, Donald B. Kohn, Luigi D. Notarangelo, Sung-Yun Pai, Morton J. Cowan, Christopher C. Dvorak, Élie Haddad, Jennifer M. Puck, Luis B. Barreiro, and Hélène Decaluwe
- Subjects
Immunology ,Immunology and Allergy - Abstract
Severe combined immunodeficiency (SCID) comprises rare inherited disorders of immunity that require definitive treatment through hematopoietic cell transplantation (HCT) or gene therapy for survival. Despite successes of allogeneic HCT, many SCID patients experience incomplete immune reconstitution, persistent T-cell lymphopenia, and poor long-term outcomes.We hypothesized that CD4We analyzed markers of exhaustion in blood samples from 61 SCID patients at a median of 10.4 years after HCT.Compared to post-HCT SCID patients with normal CD4Recipients of unconditioned HCT for SCID may develop late post-HCT T-cell exhaustion as a result of diminished production of T-lineage cells. Elevated expression of inhibitory receptors on their T cells may be a biomarker of poor long-term T-cell reconstitution.
- Published
- 2022
4. Outcomes following treatment for ADA-deficient severe combined immunodeficiency: a report from the PIDTC
- Author
-
Geoffrey D. E. Cuvelier, Brent R. Logan, Susan E. Prockop, Rebecca H. Buckley, Caroline Y. Kuo, Linda M. Griffith, Xuerong Liu, Alison Yip, Michael S. Hershfield, Paul G. Ayoub, Theodore B. Moore, Morna J. Dorsey, Richard J. O’Reilly, Neena Kapoor, Sung-Yun Pai, Malika Kapadia, Christen L. Ebens, Lisa R. Forbes Satter, Lauri M. Burroughs, Aleksandra Petrovic, Deepak Chellapandian, Jennifer Heimall, David C. Shyr, Ahmad Rayes, Jeffrey J. Bednarski, Sharat Chandra, Shanmuganathan Chandrakasan, Alfred P. Gillio, Lisa Madden, Troy C. Quigg, Emi H. Caywood, Blachy J. Dávila Saldaña, Kenneth DeSantes, Hesham Eissa, Frederick D. Goldman, Jacob Rozmus, Ami J. Shah, Mark T. Vander Lugt, Monica S. Thakar, Roberta E. Parrott, Caridad Martinez, Jennifer W. Leiding, Troy R. Torgerson, Michael A. Pulsipher, Luigi D. Notarangelo, Morton J. Cowan, Christopher C. Dvorak, Elie Haddad, Jennifer M. Puck, and Donald B. Kohn
- Subjects
Adenosine Deaminase ,Agammaglobulinemia ,Child, Preschool ,Immunology ,Hematopoietic Stem Cell Transplantation ,Infant, Newborn ,Humans ,Infant ,Severe Combined Immunodeficiency ,Cell Biology ,Hematology ,Biochemistry - Abstract
Adenosine deaminase (ADA) deficiency causes ∼13% of cases of severe combined immune deficiency (SCID). Treatments include enzyme replacement therapy (ERT), hematopoietic cell transplant (HCT), and gene therapy (GT). We evaluated 131 patients with ADA-SCID diagnosed between 1982 and 2017 who were enrolled in the Primary Immune Deficiency Treatment Consortium SCID studies. Baseline clinical, immunologic, genetic characteristics, and treatment outcomes were analyzed. First definitive cellular therapy (FDCT) included 56 receiving HCT without preceding ERT (HCT); 31 HCT preceded by ERT (ERT-HCT); and 33 GT preceded by ERT (ERT-GT). Five-year event-free survival (EFS, alive, no need for further ERT or cellular therapy) was 49.5% (HCT), 73% (ERT-HCT), and 75.3% (ERT-GT; P < .01). Overall survival (OS) at 5 years after FDCT was 72.5% (HCT), 79.6% (ERT-HCT), and 100% (ERT-GT; P = .01). Five-year OS was superior for patients undergoing HCT at
- Published
- 2022
5. Microglial Suppression Assays for Cell Therapies
- Author
-
Li Xu, Hyunjung Min, Anthony J. Filiano, Joanne Kurtzberg, and Roberta E. Parrott
- Subjects
Cancer Research ,Transplantation ,Medicine (General) ,Preclinical Studies ,QH573-671 ,business.industry ,Immunology ,Cell ,Cell Biology ,General Medicine ,medicine.anatomical_structure ,R5-920 ,Oncology ,Cancer research ,medicine ,Immunology and Allergy ,business ,Cytology ,Genetics (clinical) ,Developmental Biology - Published
- 2021
6. Optimizing the Production of a Human Umbilical Cord Blood‐Derived Cell Therapy Product, DUOC‐01
- Author
-
Anthony J. Filiano, Joanne Kurtzberg, Roberta E. Parrott, Li Xu, and Madison French
- Subjects
Preclinical Studies ,Medicine (General) ,QH573-671 ,business.industry ,Cell Biology ,General Medicine ,Umbilical cord ,Cell therapy ,Andrology ,medicine.anatomical_structure ,R5-920 ,Product (mathematics) ,medicine ,Production (economics) ,business ,Cytology ,Developmental Biology - Published
- 2021
7. A Novel Mechanism for Mesenchymal Stromal Cells to Induce Long‐Term Suppression of Pulmonary Inflammation
- Author
-
Ashley Moseman, Sebastian A. Wellford, Roberta E. Parrott, Joanne Kurzberg, Anthony J. Filiano, Hyunjung Min, and Li Xu
- Subjects
Medicine (General) ,R5-920 ,QH573-671 ,Chemistry ,Mechanism (biology) ,Pulmonary inflammation ,Mesenchymal stem cell ,Cancer research ,Cell Biology ,General Medicine ,Cytology ,Developmental Biology - Published
- 2021
8. Optimization of T‐Cell Suppression Potency Assay for Human Cord Tissue Mesenchymal Stromal Cells
- Author
-
Roberta E. Parrott, Joanne Kurtzberg, Anthony J. Filiano, and Li Xu
- Subjects
Medicine (General) ,Cord ,QH573-671 ,Chemistry ,T cell ,Mesenchymal stem cell ,Cell Biology ,General Medicine ,Perinatal Tissue Banking and Therapies (Including Mscs) ,medicine.anatomical_structure ,R5-920 ,Cancer research ,medicine ,Potency ,Cytology ,Developmental Biology - Published
- 2021
9. Outcomes after Hematopoietic Cell Transplant and Gene Therapy for Adenosine Deaminase (ADA) Severe Combined Immune Deficiency: A Combined Analysis from the Primary Immune Deficiency Treatment Consortium (PIDTC) 6901 and 6902 Studies
- Author
-
Geoff D.E. Cuvelier, Brent Logan, Susan E. Prockop, Rebecca H. Buckley, Caroline Y. Kuo, Linda M. Griffith, Xuerong Liu, Alison Yip, Michael S. Hershfield, Roberta E Parrott, Christen L. Ebens, Theodore B. Moore, Richard J. O’Reilly, Sung-Yun Pai, Malika Kapadia, Neena Kapoor, Lisa R. Forbes Satter, Caridad Martinez, Lauri M. Burroughs, Aleksandra Petrovic, Monica S. Thakar, Deepak Chellapandian, Jennifer Heimall, David C. Shyr, Ahmad Rayes, Jeffrey J. Bednarski II, Sharat Chandra, Shanmuganathan Chandrakasan, Alfred P. Gillio, Lisa Madden, Troy C. Quigg, Emi H. Caywood, Blachy J Dávila Saldaña, Kenneth DeSantes, Hesham Eissa, Frederick Goldman, Jacob Rozmus, Ami J Shah, Mark T. Vander Lugt, Mike A. Pulsipher, Luigi D. Notarangelo, Morton J. Cowan, Christopher C. Dvorak, Elie Haddad, Jennifer Puck, and Donald B. Kohn
- Subjects
Transplantation ,Molecular Medicine ,Immunology and Allergy ,Cell Biology ,Hematology - Published
- 2022
- Full Text
- View/download PDF
10. SCID genotype and 6-month posttransplant CD4 count predict survival and immune recovery
- Author
-
Harry L. Malech, Roberta E. Parrott, Evan Shereck, Kenneth B. DeSantes, Troy C. Quigg, Thomas A. Fleisher, Alfred P. Gillio, Rebecca H. Buckley, Richard J. O'Reilly, Sung-Yun Pai, Luigi D. Notarangelo, Victor M. Aquino, Morton J. Cowan, Jeffrey J. Bednarski, Jennifer M. Puck, Donald B. Kohn, David C. Shyr, Soma Jyonouchi, Imelda C. Hanson, Pierre Teira, Matthew H. Porteus, Angela R. Smith, Paul Szabolcs, Candace Taylor, Jeffrey H. Davis, Mark Vander Lugt, Jack J. Bleesing, Morris Kletzel, Hélène Decaluwe, Megan Murnane, Christine M. Seroogy, Trudy N. Small, James A. Connelly, Audrey G. Tumlin, Sharat Chandra, Matthew E. Cavanaugh, Kathleen E. Sullivan, Ann E. Haight, Aleksandra Petrovic, Linda M. Griffith, Neena Kapoor, Brent R. Logan, John Craddock, Susan E. Prockop, Michael A. Pulsipher, Michael D. Keller, Geoffrey D.E. Cuvelier, Caridad Martinez, Jessica Chaisson, Frederick D. Goldman, Alan P. Knutsen, Monica S. Thakar, Lolie C. Yu, Ziyan Yin, Lauri Burroughs, William T. Shearer, Hisham Abdel-Azim, Jennifer W. Leiding, Jennifer Heimall, Elie Haddad, Christopher C. Dvorak, Theodore B. Moore, Blachy J. Dávila Saldaña, Elizabeth M. Kang, and Suzanne Skoda-Smith
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Genotype ,DCLRE1C ,medicine.medical_treatment ,Immunology ,Plenary Paper ,Hematopoietic stem cell transplantation ,Biochemistry ,03 medical and health sciences ,Immune Reconstitution ,Immune system ,Internal medicine ,medicine ,Humans ,Lymphocyte Count ,Retrospective Studies ,Severe combined immunodeficiency ,business.industry ,Hematopoietic Stem Cell Transplantation ,Immunosuppression ,DNA ,Cell Biology ,Hematology ,medicine.disease ,Omenn syndrome ,CD4 Lymphocyte Count ,Transplantation ,030104 developmental biology ,Severe Combined Immunodeficiency ,business - Abstract
The Primary Immune Deficiency Treatment Consortium (PIDTC) performed a retrospective analysis of 662 patients with severe combined immunodeficiency (SCID) who received a hematopoietic cell transplantation (HCT) as first-line treatment between 1982 and 2012 in 33 North American institutions. Overall survival was higher after HCT from matched-sibling donors (MSDs). Among recipients of non-MSD HCT, multivariate analysis showed that the SCID genotype strongly influenced survival and immune reconstitution. Overall survival was similar for patients with RAG, IL2RG, or JAK3 defects and was significantly better compared with patients with ADA or DCLRE1C mutations. Patients with RAG or DCLRE1C mutations had poorer immune reconstitution than other genotypes. Although survival did not correlate with the type of conditioning regimen, recipients of reduced-intensity or myeloablative conditioning had a lower incidence of treatment failure and better T- and B-cell reconstitution, but a higher risk for graft-versus-host disease, compared with those receiving no conditioning or immunosuppression only. Infection-free status and younger age at HCT were associated with improved survival. Typical SCID, leaky SCID, and Omenn syndrome had similar outcomes. Landmark analysis identified CD4(+) and CD4(+)CD45RA(+) cell counts at 6 and 12 months post-HCT as biomarkers predictive of overall survival and long-term T-cell reconstitution. Our data emphasize the need for patient-tailored treatment strategies depending upon the underlying SCID genotype. The prognostic significance of CD4(+) cell counts as early as 6 months after HCT emphasizes the importance of close follow-up of immune reconstitution to identify patients who may need additional intervention to prevent poor long-term outcome.
- Published
- 2018
- Full Text
- View/download PDF
11. B-cell differentiation and IL-21 response in IL2RG/JAK3 SCID patients after hematopoietic stem cell transplantation
- Author
-
Susan E. Prockop, Richard J. O'Reilly, Luigi D. Notarangelo, Alexandra Miggelbrink, Morton J. Cowan, Roberta E. Parrott, Linda M. Griffith, Christopher C. Dvorak, Neena Kapoor, Jolan E. Walter, David C. Shyr, Brent R. Logan, Jennifer M. Puck, Donald B. Kohn, Alfred P. Gillio, Sung-Yun Pai, Blachy J. Dávila Saldaña, Hermann Eibel, Gregory Hopkins, Hélène Decaluwe, Jennifer Whangbo, Hisham Abdel-Azim, Elie Haddad, Imelda C. Hanson, and Rebecca H. Buckley
- Subjects
0301 basic medicine ,Severe combined immunodeficiency ,biology ,business.industry ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Immunoglobulin secretion ,03 medical and health sciences ,Interleukin 21 ,surgical procedures, operative ,030104 developmental biology ,medicine.anatomical_structure ,Immune system ,Immunoglobulin M ,biology.protein ,medicine ,Antibody ,business ,B cell - Abstract
Allogeneic hematopoietic stem cell transplant (HSCT) typically results in donor T-cell engraftment and function in patients with severe combined immunodeficiency (SCID), but humoral immunity, particularly when using donors other than matched siblings, is variable. B-cell function after HSCT for SCID depends on the genetic cause, the use of pre-HSCT conditioning, and whether donor B-cell chimerism is achieved. Patients with defects in IL2RG or JAK3 undergoing HSCT without conditioning often have poor B-cell function post-HSCT, perhaps as a result of impairment of IL-21 signaling in host-derived B cells. To investigate the effect of pre-HSCT conditioning on B-cell function, and the relationship of in vitro B-cell function to clinical humoral immune status, we analyzed 48 patients with IL2RG/JAK3 SCID who were older than 2 years after HSCT with donors other than matched siblings. T follicular helper cells (TFH) developed in these patients with kinetics similar to healthy young children; thus, poor B-cell function could not be attributed to a failure of TFH development. In vitro differentiation of B cells into plasmablasts and immunoglobulin secretion in response to IL-21 strongly correlated with the use of conditioning, donor B-cell engraftment, freedom from immunoglobulin replacement, and response to tetanus vaccine. Patients receiving immunoglobulin replacement who had normal serum immunoglobulin M showed poor response to IL-21 in vitro, similar to those with low serum IgM. In vitro response of B cells to IL-21 may predict clinically relevant humoral immune function in patients with IL2RG/JAK3 SCID after HSCT.
- Published
- 2018
- Full Text
- View/download PDF
12. Mesenchymal stromal cells reprogram macrophages with processing bodies
- Author
-
A. Moseman, S. Wellford, Roberta E. Parrott, Anthony J. Filiano, Hyunjung Min, Li Xu, and Joanne Kurtzberg
- Subjects
Cancer Research ,Transplantation ,Lung ,Lipopolysaccharide ,business.industry ,Immunology ,Mesenchymal stem cell ,Inflammation ,Cell Biology ,Proinflammatory cytokine ,Cell therapy ,chemistry.chemical_compound ,Immune system ,medicine.anatomical_structure ,Oncology ,chemistry ,Cancer research ,Immunology and Allergy ,Medicine ,medicine.symptom ,Diffuse alveolar damage ,business ,Genetics (clinical) - Abstract
Background & Aim: Mesenchymal stromal cells (MSCs) are an investigational cell therapy for inflammatory diseases. Although they have robust anti-inflammatory properties, their success has been variable in clinical trials due to an unclear understanding of their mechanism. Once injected, a majority of MSCs traffic to the lung, where they are rapidly cleared, signifying an opportunity to target lung inflammatory [Figure presented] conditions such as acute respiratory distress syndrome (ARDS). ARDS is a catastrophic condition of the lungs, involving pulmonary inflammation that develops with severe SARS-CoV2 and other respiratory infections. MSCs are expected to prevent alveolar damage by suppressing the immune response and there is evidence that MSCs protect in phase I/IIa trials for ARDS associated with COVID-19. While these results are promising, understanding the mechanism is critical to determine dosing, maximize efficacy, and ultimately lead to an approved product. Methods, Results & Conclusion: We and others have demonstrated that within their short time in the lung, MSCs interact with monocytes and macrophages. Through direct cell contact, MSCs transfer cytoplasmic components, notably cytoplasmic processing-bodies (p-bodies) to monocytes and macrophages. P-bodies are membrane-less organelles that contain RNA binding proteins, microRNAs, and mRNAs enriched for genes that regulate the transcriptional landscape of cells. MSC interactions result in long-term transcriptional reprogramming of monocytes and macrophages to suppress a helper T cell response and upregulate tissue repair pathways. To investigate the mechanisms of MSCs in vivo, we utilized 2 mouse models of lung inflammation: 1. intranasal lipopolysaccharide (LPS) to study general acute inflammation and 2. an engineered vesicular stomatitis virus (VSV) with a SARS-CoV2 Spike protein. Using these models, we demonstrated that during inflammation cytoplasm of MSCs transferred to lung macrophages to decrease activation and the expression of MHC-II. Further, MSCs prevented a decrease in resident alveolar macrophages, suppressed proinflammatory macrophages, and blocked an influx in infiltrating monocytes (Fig 1). Depleting p-bodies from MSCs abolished the beneficial effects, despite transfer cytoplasmic component to macrophages at similar levels of control MSCs. Overall, our data suggest a novel form of cell communication that could explain how MSCs could lead to long-term beneficial effects on lung inflammation despite being rapidly cleared. [Figure presented]
- Published
- 2021
- Full Text
- View/download PDF
13. DUOC-01, a cord blood derived cell therapy product, ameliorates experimental autoimmune encephalomyelitis, a murine model for multiple sclerosis
- Author
-
Arjun Saha, Joanne Kurtzberg, Paula Scotland, Li Xu, Anthony J. Filiano, Jonathan Schwartzman, and Roberta E. Parrott
- Subjects
Cancer Research ,Transplantation ,Pathology ,medicine.medical_specialty ,biology ,business.industry ,Multiple sclerosis ,Immunology ,Experimental autoimmune encephalomyelitis ,Central nervous system ,Cell Biology ,medicine.disease ,Myelin oligodendrocyte glycoprotein ,Cell therapy ,Cellular infiltration ,medicine.anatomical_structure ,Oncology ,Gliosis ,medicine ,biology.protein ,Immunology and Allergy ,Remyelination ,medicine.symptom ,business ,Genetics (clinical) - Abstract
Background & Aim We have developed an umbilical cord-blood derived, macrophage-like cell therapy, DUOC-01, to treat demyelinating conditions of the central nervous system. Previously, we showed that DUOC-01 accelerated remyelination, decreased gliosis, and reduced cellular infiltration in the brain of immune-incompetent mice exposed to the demyelinating agent, cuprizone. To further explore the mechanism of action and investigate whether DUOC-01 will be effective in other experimental models of demyelination, we tested DUOC-01 in the experimental autoimmune encephalomyelitis (EAE) model, an animal model used to study multiple sclerosis (MS). Methods, Results & Conclusion EAE was induced in C57BL/6 mice by immunizing with myelin oligodendrocyte glycoprotein peptide (MOG35-55) in complete Freund's adjuvant. We have previously established in a phase I trial of intrathecally administered DUOC-01 cells in patients with leukodystrophies, that DUOC-01 is formulated in hydrocortisone (HC) for infusion in the clinic. Therefore, to mimic the practice in the clinic, DUOC-01 cells were incubated in Ringer's Lactate with 3mg/mL HC at a dilution of 1 × 106 cells/ml for 2 hours at room temperature prior to injection. DUOC-01 cells were then administered at a dose of 3 × 105cells/mouse into the cerebrospinal fluid by a single intra-cisterna magna injection at the point when mice started showing early disease symptoms of EAE. Clinical score was recorded for next several days. Compared to mice injected with Ringer's (n=9), mice injected with DUOC-01 (n=9) had decreased severity of the disease, as indicated by lower clinical scores based on severity of ascending paralysis. The spinal cords of mice in the cohorts were studied histopathologically and those with mice injected with DUOC-01 had reduced inflammation and lower cellular infiltration compared to spinal cords of mice injected with Ringer's. The overall number of CD45+ immune cells were less in spinal cords of mice treated with DUOC-01 cells compared to the Ringer's injected mice. Most strikingly, among the various cell types, the number of neutrophils present in the spinal cords from the DUOC-01 treated group was less than the Ringer's treated samples. Presently we are exploring the mechanism(s) through which DUOC-01 cells promote remyelination and decrease immune cell infiltration. In brief, our data suggest that DUOC-01 cell therapy product could be beneficial in treating MS and other diverse neurological conditions with demyelination.
- Published
- 2020
- Full Text
- View/download PDF
14. Reprogramming inflammatory macrophages with mesenchymal stromal cells
- Author
-
Roberta E. Parrott, Emily M. Rabjohns, Anthony J. Filiano, Christopher C. Overall, N. Meadows, M. Lillich, Rishi R. Rampersad, Joanne Kurtzberg, Hyunjung Min, Teresa K. Tarrant, and Li Xu
- Subjects
Cancer Research ,Transplantation ,Immunology ,Mesenchymal stem cell ,Cell ,Inflammation ,Cell Biology ,Biology ,Cell biology ,Cell therapy ,medicine.anatomical_structure ,Oncology ,Antigen ,In vivo ,medicine ,Immunology and Allergy ,medicine.symptom ,Reprogramming ,Cell culture assays ,Genetics (clinical) - Abstract
Background & Aim MSCs represent approximately 25% of all cell-based clinical trials with over 1,000 registered on clinicaltrials.gov. Although the safety of MSCs is well-documented, the success in trials over the past 25 years is variable. This is likely due to the differences in the source tissue to generate MSCs, manufacturing procedures, the heterogeneity of the MSC populations, and the disease being targeted. Regardless of these differences, it is absolutely critical to understand the mechanism of action in vivo to optimize the potential of MSCs as a FDA licensed cell therapy product. Recent reports highlight that MSCs are rapidly cleared in vivo yet modulate myeloid cell populations to limit inflammation well after they are no longer detectable. Human umbilical cord tissue is readily available and a rich source of rapidly proliferating MSCs. In this study, we hypothesized that human cord tissue-derived MSCs (hCT-MSCs) can reprogram inflammatory macrophages and limit disease progression in inflammatory diseases. Methods, Results & Conclusion Multiple lines of hCT-MSCs were manufactured from donated cord tissue in the Robertson GMP laboratory within the Marcus Center for Cellular Cures (MC3). Using cell culture assays, we determined that hCT-MSC indirectly suppressed T cells by directly interacting with macrophages. Macrophages did not phagocytosis live MSCs, rather they engulfed cytoplasmic components, which down-regulated genes responsible for presenting antigens and activating T cells. Since vast amounts of information can be stored within cytoplasmic granules call processing bodies (P-bodies), we tested their role in facilitating hCT-MSCs to reprogram macrophages. hCT-MSCs contained substantially numbers of P-bodies in the cytoplasm and when we genetically depleted P-bodies in MSCs, they failed to reprogram macrophages to suppress T cells (Fig. 1). Overall, our data demonstrate that hCT-MSCs reprogram macrophages to suppress a T cell response through P-bodies. This not only explains a potential in vivo mechanism how MSCs can confer long-term benefit after they are cleared but is a potential novel and manipulatable target to promote increased efficacy of MSCs in multiple inflammatory conditions.
- Published
- 2020
- Full Text
- View/download PDF
15. Hydrocortisone-Treated DUOC-01, a Cord Blood-Derived Cell Therapy Product, Ameliorates Experimental Autoimmune Encephalomyelitis
- Author
-
Roberta E. Parrott, Joanne Kurtzberg, Paula Scotland, Li Xu, Arjun Saha, Jonathan Schwartzman, and Anthony J. Filiano
- Subjects
lcsh:R5-920 ,Preclinical Studies ,lcsh:Cytology ,business.industry ,Experimental autoimmune encephalomyelitis ,Cell Biology ,General Medicine ,Pharmacology ,medicine.disease ,Cell therapy ,Cord blood ,Medicine ,lcsh:QH573-671 ,lcsh:Medicine (General) ,business ,Developmental Biology ,Hydrocortisone ,medicine.drug - Published
- 2020
- Full Text
- View/download PDF
16. Measuring Microglial Suppression with Multiple Assays
- Author
-
Roberta E. Parrott, Hyunjung Min, Anthony J. Filiano, Joanne Kurtzberg, and Li Xu
- Subjects
Preclinical Studies ,lcsh:R5-920 ,lcsh:Cytology ,Cell Biology ,General Medicine ,lcsh:QH573-671 ,lcsh:Medicine (General) ,Developmental Biology - Published
- 2020
- Full Text
- View/download PDF
17. B-cell differentiation and IL-21 response in
- Author
-
Alexandra M, Miggelbrink, Brent R, Logan, Rebecca H, Buckley, Roberta E, Parrott, Christopher C, Dvorak, Neena, Kapoor, Hisham, Abdel-Azim, Susan E, Prockop, David, Shyr, Hélène, Decaluwe, Imelda C, Hanson, Alfred, Gillio, Blachy J, Dávila Saldaña, Hermann, Eibel, Gregory, Hopkins, Jolan E, Walter, Jennifer S, Whangbo, Donald B, Kohn, Jennifer M, Puck, Morton J, Cowan, Linda M, Griffith, Elie, Haddad, Richard J, O'Reilly, Luigi D, Notarangelo, and Sung-Yun, Pai
- Subjects
Male ,B-Lymphocytes ,Transplantation ,Transplantation Conditioning ,Adolescent ,Interleukins ,T-Lymphocytes ,Hematopoietic Stem Cell Transplantation ,Janus Kinase 3 ,Cell Differentiation ,Lymphocyte Activation ,Immunity, Humoral ,Young Adult ,surgical procedures, operative ,Child, Preschool ,Humans ,Transplantation, Homologous ,Female ,Severe Combined Immunodeficiency ,Child ,Interleukin Receptor Common gamma Subunit - Abstract
Allogeneic hematopoietic stem cell transplant (HSCT) typically results in donor T-cell engraftment and function in patients with severe combined immunodeficiency (SCID), but humoral immunity, particularly when using donors other than matched siblings, is variable. B-cell function after HSCT for SCID depends on the genetic cause, the use of pre-HSCT conditioning, and whether donor B-cell chimerism is achieved. Patients with defects in IL2RG or JAK3 undergoing HSCT without conditioning often have poor B-cell function post-HSCT, perhaps as a result of impairment of IL-21 signaling in host-derived B cells. To investigate the effect of pre-HSCT conditioning on B-cell function, and the relationship of in vitro B-cell function to clinical humoral immune status, we analyzed 48 patients with IL2RG/JAK3 SCID who were older than 2 years after HSCT with donors other than matched siblings. T follicular helper cells (T(FH)) developed in these patients with kinetics similar to healthy young children; thus, poor B-cell function could not be attributed to a failure of T(FH) development. In vitro differentiation of B cells into plasmablasts and immunoglobulin secretion in response to IL-21 strongly correlated with the use of conditioning, donor B-cell engraftment, freedom from immunoglobulin replacement, and response to tetanus vaccine. Patients receiving immunoglobulin replacement who had normal serum immunoglobulin M showed poor response to IL-21 in vitro, similar to those with low serum IgM. In vitro response of B cells to IL-21 may predict clinically relevant humoral immune function in patients with IL2RG/JAK3 SCID after HSCT.
- Published
- 2017
18. Targeting neuroinflammation with human umbilical cord tissue-derived mesenchymal stromal cells
- Author
-
Hyunjung Min, Li Xu, N. Meadows, Arjun Saha, Pamela Noldner, Joanne Kurtzberg, Anthony J. Filiano, A. Rudisill, Aruni Gunaratne, M. Lillich, and Roberta E. Parrott
- Subjects
Cancer Research ,Transplantation ,Pathology ,medicine.medical_specialty ,business.industry ,Immunology ,Mesenchymal stem cell ,Cell Biology ,Oncology ,Umbilical cord tissue ,medicine ,Immunology and Allergy ,business ,Genetics (clinical) ,Neuroinflammation - Published
- 2018
- Full Text
- View/download PDF
19. Human Umbilical Cord Blood-Derived Cell Therapy Product, DUOC-01, Ameliorates Experimental Autoimmune Encephalomyelitis, a Mouse Model for Multiple Sclerosis
- Author
-
Joanne Kurtzberg, Paula Scotland, Jonathan Schwartzman, Jesse D. Troy, Roberta E. Parrott, Aruni Gunaratne, Nicole Williams, Li Xu, Anthony J. Filiano, Andrew E. Balber, and Arjun Saha
- Subjects
lcsh:R5-920 ,lcsh:Cytology ,business.industry ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,Cell Biology ,General Medicine ,medicine.disease ,Umbilical cord ,Cell therapy ,medicine.anatomical_structure ,Product (mathematics) ,Immunology ,medicine ,Scientific Abstracts ,lcsh:QH573-671 ,lcsh:Medicine (General) ,business ,Developmental Biology - Published
- 2019
- Full Text
- View/download PDF
20. Long-term outcome of non-ablative booster BMT in patients with SCID
- Author
-
Roberta E. Parrott, Claire L. Teigland, and Rebecca H. Buckley
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,T-Lymphocytes ,bone marrow transplantation ,Graft vs Host Disease ,2-parent bone marrow transplants ,Transplantation Chimera ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Humans ,Prospective Studies ,Prospective cohort study ,Retrospective Studies ,030304 developmental biology ,0303 health sciences ,Transplantation ,Severe combined immunodeficiency ,Booster (rocketry) ,business.industry ,Infant, Newborn ,Infant ,Retrospective cohort study ,Hematology ,severe combined immunodeficiency ,medicine.disease ,Booster ,3. Good health ,Surgery ,Treatment Outcome ,Graft-versus-host disease ,Female ,business ,030215 immunology - Abstract
Severe combined immunodeficiency (SCID) is a fatal syndrome caused by mutations in at least 13 different genes. It is characterized by the absence of T-cells. Immune reconstitution can be achieved through non-ablative related donor bone marrow transplantation. However, the first transplant may not provide sufficient immunity. In these cases, booster transplants may be helpful. A prospective/retrospective study was conducted of 49 SCID patients (28.7 percent of 171 SCIDs transplanted over 30 years) who had received booster transplants to define the long term outcome, factors contributing to a need for a booster and factors that predicted success. Of the 49 patients, 31 (63 percent) are alive for up to 28 years. Age at initial transplantation was found to have a significant effect on outcome (mean of 194 days old for patients currently alive, versus a mean of 273 days old for those now deceased, p=0.0401). Persistent viral infection was present in most deceased booster patients. In several patients, the use of two parents as sequential donors resulted in striking T and B cell immune reconstitution. A majority of the patients alive today have normal or adequate T-cell function and are healthy. Non-ablative booster bone marrow transplantation can be life-saving for SCID.
- Published
- 2013
- Full Text
- View/download PDF
21. Characterization of msc derived from umbilical cord tissues
- Author
-
M. Lillich, Joanne Kurtzberg, N. Meadows, Pamela Noldner, Lynn Cheatham, Robert W. Storms, and Roberta E. Parrott
- Subjects
0301 basic medicine ,Cancer Research ,Transplantation ,Pathology ,medicine.medical_specialty ,Immunology ,Cell Biology ,Biology ,Umbilical cord ,Cord lining ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Immunology and Allergy ,Genetics (clinical) - Published
- 2017
- Full Text
- View/download PDF
22. Genotype, Phenotype and T Cell Counts at One Year Predict Survival and Long Term Immune Reconstitution after Transplantation in Severe Combined Immune Deficiency (SCID)—The Primary Immune Deficiency Treatment Consortium (PIDTC)
- Author
-
Mark Vander Lugt, Sung-Yun Pai, Alan P. Knutsen, Morris Kletzel, Hélène Decaluwe, Brent R. Logan, Lolie C. Yu, Monica S. Thakar, Christopher C. Dvorak, Alfred P. Gillio, Imelda C. Hanson, John Craddock, Victor M. Aquino, Susan E. Prockop, Luigi D. Notarangelo, Lauri Burroughs, Angela R. Smith, Frederick D. Goldman, Jennifer M. Puck, James A. Connelly, William T. Shearer, Paul Szabolcs, Donald B. Kohn, Audrey G. Tumlin, Hisham Abdel-Azim, Sharat Chandra, Kathleen E. Sullivan, Theodore B. Moore, Elie Haddad, Marlis L. Schroeder, Ziyan Yin, Michael A. Pulsipher, Aleksandra Petrovic, Caridad Martinez, Rebecca H. Buckley, Matthew H. Porteus, Morton J. Cowan, Ann E. Haight, Jeffrey H. Davis, Blachy J. Dávila Saldaña, Elizabeth M. Kang, Jack J. Bleesing, Christine M. Seroogy, Harry L. Malech, Richard J. O'Reilly, Roberta E. Parrott, Evan Shereck, Jeffrey J. Bednarski, Linda M. Griffith, Troy C. Quigg, Thomas A. Fleisher, Neena Kapoor, David C. Shyr, and Soma Jyonouchi
- Subjects
030203 arthritis & rheumatology ,Transplantation ,business.industry ,T cell ,Hematology ,Genotype phenotype ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Immunology ,Medicine ,business ,030215 immunology - Published
- 2017
- Full Text
- View/download PDF
23. Manufacturing of allogeneic human cord tissue mesenchymal stromal cells: Initial results of a phase I clinical trial in children with autism spectrum disorder
- Author
-
Joanne Kurtzberg, Roberta E. Parrott, Vinod K. Prasad, Lynn Cheatham, J. Lew, Jessica Sun, N. Meadows, A. Rudisill, Anthony J. Filiano, Kristin Page, Pamela Noldner, M. Lillich, and Geraldine Dawson
- Subjects
Cancer Research ,Transplantation ,Pathology ,medicine.medical_specialty ,Cord ,business.industry ,Immunology ,Mesenchymal stem cell ,Phases of clinical research ,Cell Biology ,medicine.disease ,Oncology ,Autism spectrum disorder ,medicine ,Immunology and Allergy ,business ,Genetics (clinical) - Published
- 2018
- Full Text
- View/download PDF
24. Dominant Splice Site Mutations in PIK3R1 Cause Hyper IgM Syndrome, Lymphadenopathy and Short Stature
- Author
-
Balachandra K. Gorentla, Hongxiang Huang, Xiao-Ping Zhong, Talal Mousallem, David Goldstein, Joseph L. Roberts, Roberta E. Parrott, Martin Armstrong, Slavé Petrovski, Jialong Yang, Duncan McHale, Endi Wang, Nancie J. MacIver, and Rebecca H. Buckley
- Subjects
0301 basic medicine ,Male ,DNA Mutational Analysis ,Lymphadenopathy ,Gene mutation ,medicine.disease_cause ,Hyper-IgM Immunodeficiency Syndrome ,Craniofacial Abnormalities ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Immunology and Allergy ,Child ,Growth Disorders ,Cell Line, Transformed ,Genes, Dominant ,Mutation ,B-Lymphocytes ,TOR Serine-Threonine Kinases ,High-Throughput Nucleotide Sequencing ,Ear ,Thorax ,Class Ia Phosphatidylinositol 3-Kinase ,Nephrocalcinosis ,SHORT syndrome ,Child, Preschool ,Female ,medicine.symptom ,Signal Transduction ,medicine.medical_specialty ,Hyper IgM syndrome ,Immunology ,Dwarfism ,Biology ,Short stature ,Article ,03 medical and health sciences ,Metabolic Diseases ,Internal medicine ,medicine ,Humans ,PI3K/AKT/mTOR pathway ,CD40 ,Ribosomal Protein S6 Kinases ,medicine.disease ,Molecular biology ,Alternative Splicing ,030104 developmental biology ,Endocrinology ,biology.protein ,Hypercalcemia ,Neck ,030215 immunology - Abstract
The purpose of this research was to use next generation sequencing to identify mutations in patients with primary immunodeficiency diseases whose pathogenic gene mutations had not been identified. Remarkably, four unrelated patients were found by next generation sequencing to have the same heterozygous mutation in an essential donor splice site of PIK3R1 (NM_181523.2:c.1425 + 1G > A) found in three prior reports. All four had the Hyper IgM syndrome, lymphadenopathy and short stature, and one also had SHORT syndrome. They were investigated with in vitro immune studies, RT-PCR, and immunoblotting studies of the mutation's effect on mTOR pathway signaling. All patients had very low percentages of memory B cells and class-switched memory B cells and reduced numbers of naive CD4+ and CD8+ T cells. RT-PCR confirmed the presence of both an abnormal 273 base-pair (bp) size and a normal 399 bp size band in the patient and only the normal band was present in the parents. Following anti-CD40 stimulation, patient's EBV-B cells displayed higher levels of S6 phosphorylation (mTOR complex 1 dependent event), Akt phosphorylation at serine 473 (mTOR complex 2 dependent event), and Akt phosphorylation at threonine 308 (PI3K/PDK1 dependent event) than controls, suggesting elevated mTOR signaling downstream of CD40. These observations suggest that amino acids 435-474 in PIK3R1 are important for its stability and also its ability to restrain PI3K activity. Deletion of Exon 11 leads to constitutive activation of PI3K signaling. This is the first report of this mutation and immunologic abnormalities in SHORT syndrome.
- Published
- 2016
25. Unusual clinical and immunologic manifestations of transplacentally acquired maternal T cells in severe combined immunodeficiency
- Author
-
Roberta E. Parrott, Todd D. Green, Dong-Feng Chen, Kricia Palmer, Rebecca H. Buckley, Joseph L. Roberts, Nancy L. Reinsmoen, E.O. Sajaroff, and Myriah Cooney
- Subjects
Reoperation ,CD3 Complex ,Pancytopenia ,T-Lymphocytes ,Lymphocyte ,Immunology ,Autoimmune Diseases ,Antibodies, Monoclonal, Murine-Derived ,Immune system ,medicine ,Humans ,Immunologic Factors ,Immunology and Allergy ,Lymphocytes ,Bone Marrow Transplantation ,Cell Proliferation ,Severe combined immunodeficiency ,Receptors, Interleukin-7 ,business.industry ,Janus kinase 3 ,Antibodies, Monoclonal ,Infant ,Janus Kinase 3 ,T lymphocyte ,medicine.disease ,Virology ,Transplantation ,medicine.anatomical_structure ,Graft-versus-host disease ,Haplotypes ,Leukocyte Common Antigens ,Female ,Severe Combined Immunodeficiency ,Mitogens ,Rituximab ,business ,Immunity, Maternally-Acquired - Abstract
The persistence of transplacentally transferred maternal T cells is common in infants with severe combined immunodeficiency (SCID), occurring in more than half of patients with SCID undergoing transplantation at our institution. These T cells respond poorly to mitogens in vitro but can cause cutaneous graft-versus-host disease; however, other effects of these cells are unknown. We describe 2 infants with SCID who had unusual problems associated with transplacentally transferred maternal T cells. Patient 1 was a 5-month-old girl with Janus kinase 3-deficient SCID who had 4% circulating CD3(+) T cells but no lymphocyte proliferative response to mitogens. Although the number of T cells increased after 2 nonchemoablated, T cell-depleted, haploidentical, paternal bone marrow transplantations, T-cell function failed to develop, and she became pancytopenic. Restriction fragment length polymorphism studies of flow cytometry-sorted blood T cells revealed all to be of maternal origin. A subsequent nonchemoablated, T cell-depleted maternal transplantation resulted in normal T-cell function and marrow recovery. Patient 2 was a 9-month-old girl with IL-7Ralpha-deficient SCID who presented with autoimmune pancytopenia. She had 8% blood T cells (all CD45RO(+)) but no response to mitogens. High-resolution HLA sequence-specific priming typing detected both maternal haplotypes, indicating the presence of maternal cells. Her pancytopenia resolved after treatment with rituximab and was thought to be due to host B-cell activation by transplacentally acquired maternal T cells. Persistent transplacentally acquired maternal T cells in infants with SCID can mediate immunologic functions despite failing to respond to mitogens in vitro. We present evidence that these cells can cause allograft rejection and immune cytopenias.
- Published
- 2007
- Full Text
- View/download PDF
26. Inherited DOCK2 deficiency in patients with early-onset invasive infections
- Author
-
Ergin Çiftçi, Kerry Dobbs, David Goldstein, Roberta E. Parrott, Samuli Rounioja, Cecilia Domínguez Conde, Ornella Patrizi, Kaan Boztug, Satoshi Okada, Janna Saarela, Emma Haapaniemi, Janet Chou, Yuval Itan, Mikael Knip, Talal A. Chatila, Berna Savaş, Luigi D. Notarangelo, Axel Schambach, Anne Marie Comeau, Michael Schuster, Sule Haskologlu, Merja Helminen, Michel J. Massaad, Alessandro Moretta, Pierre Lebon, Ivan Bilic, Flore Rozenberg, Silvia Parolini, Raif S. Geha, Sevgi Keles, Jean-Laurent Casanova, Rebecca H. Buckley, Giovanna Tabellini, Figen Dogu, Toshiro K. Ohsumi, Kerstin Felgentreff, Laurent Abel, Magali Audry, Christoph Bock, Aydan Ikinciogullari, Nina K. Serwas, Shen-Ying Zhang, Waleed Al-Herz, Bertrand Boisson, Adel M Alwahadneh, Vincent Pedergnana, Arzu Ensari, Kelly Capuder, Slavé Petrovski, Herrada, Anthony, Boston Children's Hospital, Harvard Medical School [Boston] (HMS), Research Center for Molecular Medicine of the Austrian Academy of Sciences [Vienna, Austria] (CeMM ), Austrian Academy of Sciences (OeAW), St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University [New York], Imagine - Institut des maladies génétiques (IMAGINE - U1163), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), University of Brescia, Helsingin yliopisto = Helsingfors universitet = University of Helsinki, Necmettin Erbakan University [Konya, Turquie], Tampere University Hospital, Ankara University School of Medicine [Turkey], Massachusetts General Hospital [Boston, MA, USA], Università degli studi di Genova = University of Genoa (UniGe), Institute for Molecular Medicine Finland [Helsinki] (FIMM), Helsinki Institute of Life Science (HiLIFE), Helsingin yliopisto = Helsingfors universitet = University of Helsinki-Helsingin yliopisto = Helsingfors universitet = University of Helsinki, Children's Hospital, Folkhälsan Research Center, Faculty of Medecine [Helsinki], Columbia University [New York], Duke University [Durham], Hannover Medical School [Hannover] (MHH), University of Massachusetts Medical School [Worcester] (UMASS), University of Massachusetts System (UMASS), Kuwait University, CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Howard Hughes Medical Institute [Chevy Chase] (HHMI), Howard Hughes Medical Institute (HHMI), Medizinische Universität Wien = Medical University of Vienna, Harvard Stem Cell Institute [Cambridge, USA] (HSCI), and Harvard University [Cambridge]
- Subjects
Male ,rac1 GTP-Binding Protein ,MESH: Fatal Outcome ,[SDV]Life Sciences [q-bio] ,Lymphocyte ,medicine.medical_treatment ,T-Lymphocytes ,MESH: GTPase-Activating Proteins ,Hematopoietic stem cell transplantation ,Fatal Outcome ,Interferon ,MESH: Guanine Nucleotide Exchange Factors ,Guanine Nucleotide Exchange Factors ,Killer Cells ,Child ,B-Lymphocytes ,Medicine (all) ,Hematopoietic Stem Cell Transplantation ,General Medicine ,MESH: Infant ,3. Good health ,Pedigree ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,Genetic Diseases ,Natural ,Female ,Child, Preschool ,Genes, Recessive ,Genetic Diseases, Inborn ,Humans ,Immunologic Deficiency Syndromes ,Infant ,Killer Cells, Natural ,Mutation ,medicine.drug ,MESH: Killer Cells, Natural ,MESH: Genetic Diseases, Inborn ,MESH: Mutation ,MESH: Immunologic Deficiency Syndromes ,MESH: Pedigree ,Combined immunodeficiencies ,Immunity ,MESH: B-Lymphocytes ,medicine ,Recessive ,Preschool ,MESH: Genes, Recessive ,MESH: Hematopoietic Stem Cell Transplantation ,MESH: Humans ,MESH: rac1 GTP-Binding Protein ,business.industry ,MESH: Child, Preschool ,medicine.disease ,MESH: Male ,Transplantation ,MESH: T-Lymphocytes ,Inborn ,Genes ,Humoral immunity ,Immunology ,Primary immunodeficiency ,business ,MESH: Female - Abstract
International audience; Background Combined immunodeficiencies are marked by inborn errors of T-cell immunity in which the T cells that are present are quantitatively or functionally deficient. Impaired humoral immunity is also common. Patients have severe infections, autoimmunity, or both. The specific molecular, cellular, and clinical features of many types of combined immunodeficiencies remain unknown. Methods We performed genetic and cellular immunologic studies involving five unrelated children with early-onset invasive bacterial and viral infections, lymphopenia, and defective T-cell, B-cell, and natural killer (NK)-cell responses. Two patients died early in childhood; after allogeneic hematopoietic stem-cell transplantation, the other three had normalization of T-cell function and clinical improvement. Results We identified biallelic mutations in the dedicator of cytokinesis 2 gene (DOCK2) in these five patients. RAC1 activation was impaired in the T cells. Chemokine-induced migration and actin polymerization were defective in the T cells, B cells, and NK cells. NK-cell degranulation was also affected. Interferon-α and interferon-λ production by peripheral-blood mononuclear cells was diminished after viral infection. Moreover, in DOCK2-deficient fibroblasts, viral replication was increased and virus-induced cell death was enhanced; these conditions were normalized by treatment with interferon alfa-2b or after expression of wild-type DOCK2. Conclusions Autosomal recessive DOCK2 deficiency is a new mendelian disorder with pleiotropic defects of hematopoietic and nonhematopoietic immunity. Children with clinical features of combined immunodeficiencies, especially with early-onset, invasive infections, may have this condition. (Supported by the National Institutes of Health and others.).
- Published
- 2015
- Full Text
- View/download PDF
27. Transplantation outcomes for severe combined immunodeficiency, 2000-2009
- Author
-
James A. Connelly, Marlis L. Schroeder, Kathleen E. Sullivan, Imelda C. Hanson, Lauri Burroughs, Matthew H. Porteus, William T. Shearer, Jeffrey H. Davis, Angela R. Smith, Alexandra H. Filipovich, Michael A. Pulsipher, Brett Loechelt, Jennifer M. Puck, Victor M. Aquino, Donald B. Kohn, Roberta E. Parrott, Qun Xiang, Audrey Grizzle, Linda M. Griffith, Luigi D. Notarangelo, Neena Kapoor, Trudy N. Small, Ka Wah Chan, Sung-Yun Pai, Brent R. Logan, Soma Jyonouchi, Ann E. Haight, Rebecca H. Buckley, Ramsay Fuleihan, Richard J. O'Reilly, Alfred P. Gillio, Elie Haddad, Frederick D. Goldman, Alan P. Knutsen, Thomas A. Fleisher, Morton J. Cowan, Suzanne Skoda-Smith, Christopher C. Dvorak, and Theodore B. Moore
- Subjects
Male ,medicine.medical_specialty ,Pediatrics ,Transplantation Conditioning ,CD3 Complex ,medicine.medical_treatment ,T-Lymphocytes ,Graft vs Host Disease ,Hematopoietic stem cell transplantation ,Regenerative Medicine ,Medical and Health Sciences ,Article ,Rare Diseases ,Clinical Research ,General & Internal Medicine ,medicine ,Humans ,Lymphocyte Count ,Sibling ,Survival rate ,Retrospective Studies ,Pediatric ,Transplantation ,Severe combined immunodeficiency ,business.industry ,Incidence (epidemiology) ,Incidence ,Siblings ,Hematopoietic Stem Cell Transplantation ,Infant ,Retrospective cohort study ,Organ Transplantation ,General Medicine ,medicine.disease ,Surgery ,Immunoglobulin A ,Survival Rate ,Orphan Drug ,Good Health and Well Being ,Treatment Outcome ,Retreatment ,Severe Combined Immunodeficiency ,Female ,business - Abstract
Background: The Primary Immune Deficiency Treatment Consortium was formed to analyze the results of hematopoietic-cell transplantation in children with severe combined immunodeficiency (SCID) and other primary immunodeficiencies. Factors associated with a good transplantation outcome need to be identified in order to design safer and more effective curative therapy, particularly for children with SCID diagnosed at birth. Methods: We collected data retrospectively from 240 infants with SCID who had received transplants at 25 centers during a 10-year period (2000 through 2009). Results: Survival at 5 years, freedom from immunoglobulin substitution, and CD3+ T-cell and IgA recovery were more likely among recipients of grafts from matched sibling donors than among recipients of grafts from alternative donors. However, the survival rate was high regardless of donor type among infants who received transplants at 3.5 months of age or younger (94%) and among older infants without prior infection (90%) or with infection that had resolved (82%). Among actively infected infants without a matched sibling donor, survival was best among recipients of haploidentical T-cell - depleted transplants in the absence of any pretransplantation conditioning. Among survivors, reduced-intensity or myeloablative pretransplantation conditioning was associated with an increased likelihood of a CD3+ T-cell count of more than 1000 per cubic millimeter, freedom from immunoglobulin substitution, and IgA recovery but did not significantly affect CD4+ T-cell recovery or recovery of phytohemagglutinin- induced T-cell proliferation. The genetic subtype of SCID affected the quality of CD3+ T-cell recovery but not survival. Conclusions: Transplants from donors other than matched siblings were associated with excellent survival among infants with SCID identified before the onset of infection. All available graft sources are expected to lead to excellent survival among asymptomatic infants. (Funded by the National Institute of Allergy and Infectious Diseases and others.) Copyright © 2014 Massachusetts Medical Society.
- Published
- 2014
- Full Text
- View/download PDF
28. Clinical application of whole-genome sequencing in patients with primary immunodeficiency
- Author
-
Talal Mousallem, David Goldstein, Mingfu Zhu, Roberta E. Parrott, Brian J. Krueger, Joseph L. Roberts, Mehdi Adeli, Rebecca H. Buckley, Thomas J. Urban, Sarah E. Kleinstein, and K. Melodi McSweeney
- Subjects
Male ,T-Lymphocytes ,Immunology ,Molecular Sequence Data ,Disease ,Biology ,Genome ,DNA sequencing ,Article ,medicine ,Immunology and Allergy ,Humans ,Base sequence ,In patient ,Lymphocyte Count ,Child ,Whole genome sequencing ,Genetics ,B-Lymphocytes ,Base Sequence ,Extramural ,Genome, Human ,Immunologic Deficiency Syndromes ,High-Throughput Nucleotide Sequencing ,NADPH Oxidases ,Nuclear Proteins ,Exons ,medicine.disease ,Endonucleases ,Immunoglobulin A ,DNA-Binding Proteins ,Killer Cells, Natural ,Alternative Splicing ,Immunoglobulin G ,Mutation ,Primary immunodeficiency ,Female - Abstract
This report illustrates the value of whole genome sequencing (WGS) in elucidating the genetic cause of disease in patients with primary immunodeficiency (PID). As sequencing costs decline, we predict that utilization of next generation sequencing (NGS) in the clinical setting will increase.
- Published
- 2014
29. CD45-deficient severe combined immunodeficiency caused by uniparental disomy
- Author
-
Jianming Pei, David L. Wiest, Suraj Peri, Roberta E. Parrott, Joseph R. Testa, Alla Lapidus, Jinwook Shin, Qiong Wei, Joseph L. Roberts, Biao Luo, Roland L. Dunbrack, Rebecca H. Buckley, and Xiao-Ping Zhong
- Subjects
Genetics ,Mutation ,Severe combined immunodeficiency ,Heterozygote ,Multidisciplinary ,Loss of Heterozygosity ,Heterozygote advantage ,Biology ,Uniparental Disomy ,Biological Sciences ,medicine.disease_cause ,medicine.disease ,Polymorphism, Single Nucleotide ,Uniparental disomy ,Loss of heterozygosity ,medicine ,Humans ,Leukocyte Common Antigens ,Severe Combined Immunodeficiency ,Allele ,Gene ,Exome sequencing - Abstract
Analysis of the molecular etiologies of SCID has led to important insights into the control of immune cell development. Most cases of SCID result from either X-linked or autosomal recessive inheritance of mutations in a known causative gene. However, in some cases, the molecular etiology remains unclear. To identify the cause of SCID in a patient known to lack the protein-tyrosine phosphatase CD45, we used SNP arrays and whole-exome sequencing. The patient’s mother was heterozygous for an inactivating mutation in CD45 but the paternal alleles exhibited no detectable mutations. The patient exhibited a single CD45 mutation identical to the maternal allele. Patient SNP array analysis revealed no change in copy number but loss of heterozygosity for the entire length of chromosome 1 (Chr1), indicating that disease was caused by uniparental disomy (UPD) with isodisomy of the entire maternal Chr1 bearing the mutant CD45 allele. Nonlymphoid blood cells and other mesoderm- and ectoderm-derived tissues retained UPD of the entire maternal Chr1 in this patient, who had undergone successful bone marrow transplantation. Exome sequencing revealed mutations in seven additional genes bearing nonsynonymous SNPs predicted to have deleterious effects. These findings are unique in representing a reported case of SCID caused by UPD and suggest UPD should be considered in SCID and other recessive disorders, especially when the patient appears homozygous for an abnormal gene found in only one parent. Evaluation for alterations in other genes affected by UPD should also be considered in such cases.
- Published
- 2012
30. Thymic output, T-cell diversity, and T-cell function in long-term human SCID chimeras
- Author
-
Chan M. Win, Marcella Sarzotti-Kelsoe, Barry K. Moser, Gregory D. Sempowski, Myriah Cooney, Roberta E. Parrott, Rebecca H. Buckley, and Joseph L. Roberts
- Subjects
Male ,medicine.medical_specialty ,Time Factors ,T cell ,T-Lymphocytes ,Immunology ,Receptors, Antigen, T-Cell ,Transplantation Chimera ,Thymus Gland ,Biology ,Biochemistry ,Chimera (genetics) ,Internal medicine ,medicine ,Humans ,Transplantation, Homologous ,Receptor ,Bone Marrow Transplantation ,Retrospective Studies ,Severe combined immunodeficiency ,Hematology ,Infant ,Cell Biology ,T lymphocyte ,medicine.disease ,Transplantation ,medicine.anatomical_structure ,Female ,Severe Combined Immunodeficiency ,Follow-Up Studies - Abstract
Severe combined immunodeficiency (SCID) is a syndrome of diverse genetic cause characterized by profound deficiencies of T, B, and sometimes NK-cell function. Nonablative human leukocyte antigen–identical or rigorously T cell–depleted haploidentical parental bone marrow transplantation (BMT) results in thymus-dependent genetically donor T-cell development in the recipients, leading to long-term survival. We reported previously that normal T-cell numbers, function, and repertoire developed by 3 to 4 months after transplantation in SCID patients, and the repertoire remained highly diverse for the first 10 years after BMT. The T-cell receptor diversity positively correlated with T-cell receptor excision circle levels, a reflection of thymic output. However, the fate of thymic function in SCID patients beyond 10 to 12 years after BMT remained to be determined. In this greater than 25-year follow-up study of 128 patients with 11 different molecular types of SCID after nonconditioned BMT, we provide evidence that T-cell function, thymic output, and T-cell clonal diversity are maintained long-term.
- Published
- 2009
31. T-B+NK+ severe combined immunodeficiency caused by complete deficiency of the CD3zeta subunit of the T-cell antigen receptor complex
- Author
-
David L. Wiest, Roberta E. Parrott, Jeffery H. Carpenter, Marcella Sarzotti, Michael S. Krangel, Myriah Cooney, Chan M. Win, Jens Peter Holst Lauritsen, Juan Carabana, Xiao-Ping Zhong, E.O. Sajaroff, Michael D. Keller, Joseph L. Roberts, and Rebecca H. Buckley
- Subjects
Receptor complex ,CD3 Complex ,Clinical Trials and Observations ,CD8 Antigens ,T-Lymphocytes ,Immunology ,Population ,Receptors, Antigen, T-Cell ,Biology ,Biochemistry ,Cell Line ,Antigen ,Transduction, Genetic ,medicine ,Humans ,education ,Severe combined immunodeficiency ,education.field_of_study ,B-Lymphocytes ,T-cell receptor ,Infant ,Cell Biology ,Hematology ,T lymphocyte ,Exons ,medicine.disease ,Virology ,Molecular biology ,Killer Cells, Natural ,Mutagenesis, Insertional ,Retroviridae ,Gene Expression Regulation ,Cell culture ,Multiprotein Complexes ,CD4 Antigens ,Neural cell adhesion molecule ,Female ,Severe Combined Immunodeficiency - Abstract
CD3zeta is a subunit of the T-cell antigen receptor (TCR) complex required for its assembly and surface expression that also plays an important role in TCR-mediated signal transduction. We report here a patient with T(-)B(+)NK(+) severe combined immunodeficiency (SCID) who was homozygous for a single C insertion following nucleotide 411 in exon 7 of the CD3zeta gene. The few T cells present contained no detectable CD3zeta protein, expressed low levels of cell surface CD3epsilon, and were nonfunctional. CD4(+)CD8(-)CD3epsilon(low), CD4(-)CD8(+)CD3epsilon(low), and CD4(-)CD8(-)CD3epsilon(low) cells were detected in the periphery, and the patient also exhibited an unusual population of CD56(-)CD16(+) NK cells with diminished cytolytic activity. Additional studies demonstrated that retrovirally transduced patient mutant CD3zeta cDNA failed to rescue assembly of nascent complete TCR complexes or surface TCR expression in CD3zeta-deficient MA5.8 murine T-cell hybridoma cells. Nascent transduced mutant CD3zeta protein was also not detected in metabolically labeled MA5.8 cells, suggesting that it was unstable and rapidly degraded. Taken together, these findings provide the first demonstration that complete CD3zeta deficiency in humans can cause SCID by preventing normal TCR assembly and surface expression.
- Published
- 2006
32. Retrospective Study of 240 Patients with Severe Combined Immunodeficiency Transplanted from 2000-2009: A Report from the Primary Immune Deficiency Treatment Consortium of North America
- Author
-
Roberta E. Parrott, Morton J. Cowan, Jeffrey H. Davis, Qun Xiang, Marlis L. Schroeder, James A. Connelly, Ka Wah Chan, Linda M. Griffith, Frederick D. Goldman, Neena Kapoor, Ann E. Haight, Alexandra H. Filipovich, Christopher C. Dvorak, Michael A. Pulsipher, Elie Haddad, Brent R. Logan, Theodore B. Moore, Imelda C. Hanson, Alan P. Knutsen, Jennifer M. Puck, Donald B. Kohn, Trudy N. Small, Soma Jyonouchi, Rebecca H. Buckley, Ramsay Fuleihan, Sung-Yun Pai, Richard J. O'Reilly, Matthew H. Porteus, Thomas A. Fleisher, Luigi D. Notarangelo, Angela R. Smith, Brett Loechelt, Lauri Burroughs, William T. Shearer, Alfred P. Gillio, and Victor M. Aquino
- Subjects
Transplantation ,Severe combined immunodeficiency ,Pediatrics ,medicine.medical_specialty ,Immune system ,business.industry ,Medicine ,chemical and pharmacologic phenomena ,Retrospective cohort study ,Hematology ,business ,medicine.disease - Published
- 2014
- Full Text
- View/download PDF
33. T cell repertoire development in humans with SCID after nonablative allogeneic marrow transplantation
- Author
-
Scott Langdon, Roberta E. Parrott, Shui Cao, Katherine Coyne, Dhavalkumar D. Patel, Rebecca H. Buckley, Marcella Sarzotti, Daniel A. Ozaki, and Xiaojing Li
- Subjects
T cell ,CD3 ,Receptors, Antigen, T-Cell, alpha-beta ,Immunology ,chemical and pharmacologic phenomena ,Thymus Gland ,CD8-Positive T-Lymphocytes ,Immunophenotyping ,T-Lymphocyte Subsets ,Lymphopenia ,Preoperative Care ,medicine ,Immunology and Allergy ,Humans ,Longitudinal Studies ,Lymphocyte Count ,Postoperative Period ,Gene Rearrangement, beta-Chain T-Cell Antigen Receptor ,Bone Marrow Transplantation ,biology ,Repertoire ,T-cell receptor ,hemic and immune systems ,Cell Differentiation ,Gene rearrangement ,Complementarity Determining Regions ,Clone Cells ,Transplantation ,medicine.anatomical_structure ,biology.protein ,Leukocytes, Mononuclear ,Leukocyte Common Antigens ,Severe Combined Immunodeficiency ,CD8 ,Cell Division - Abstract
Transplantation of HLA-identical or haploidentical T cell-depleted allogeneic bone marrow (BM) into SCID infants results in thymus-dependent T cell development in the recipients. Immunoscope analysis of the TCR Vβ repertoire was performed on 15 SCID patients given BM transplants. Before and within the first 100 days after bone marrow transplantation (BMT), patients’ PBMC displayed an oligoclonal or skewed T cell repertoire, low TCR excision circles (TREC) values, and a predominance of CD45RO+ T cells. In contrast, the presence of high numbers of CD45RA+ cells in the circulation of SCID patients >100 days post-BMT correlated with active T cell output by the thymus as revealed by high TREC values and a polyclonal T cell repertoire demonstrated by a Gaussian distribution of Vβ-specific peaks. Ten years after BMT, we observed a decrease of the normal polyclonal T cell repertoire and an increase of a more skewed T cell repertoire. A decline of TREC levels and a decrease in the number of CD45RA+ cells beyond 10 years after BMT was concomitant with the detection of oligoclonal CD3+CD8+CD45RO+ cells. The switch from a polyclonal to a more skewed repertoire, observed in the CD3+CD8+CD45RO+ T cell subset, is a phenomenon that occurs normally with decreased thymic output during aging, but not as rapidly as in this patient population. We conclude that a normal T cell repertoire develops in SCID patients as a result of thymic output and the repertoire remains highly diverse for the first 10 years after BMT. The TCR diversity positively correlates in these patients with TREC levels.
- Published
- 2003
34. Thymic function after hematopoietic stem-cell transplantation for the treatment of severe combined immunodeficiency
- Author
-
Barton F. Haynes, Maria E. Gooding, Rebecca H. Buckley, Roberta E. Parrott, Kimberly M. Curtis, and Dhavalkumar D. Patel
- Subjects
medicine.medical_treatment ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,Hematopoietic stem cell transplantation ,Thymus Gland ,Immune system ,Antigen ,medicine ,Humans ,Lymphocyte Count ,Severe combined immunodeficiency ,business.industry ,Hematopoietic Stem Cell Transplantation ,Infant ,General Medicine ,medicine.disease ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Phenotype ,Immunology ,Leukocyte Common Antigens ,Severe Combined Immunodeficiency ,Bone marrow ,Stem cell ,business ,Plasmids - Abstract
Immune function can be restored in infants with severe combined immunodeficiency by transplantation of unfractionated bone marrow from HLA-identical donors or T-cell-depleted marrow stem cells from haploidentical donors, with whom there is a single haplotype mismatch, without the need for chemotherapy before transplantation or prophylaxis against graft-versus-host disease. The role of the thymus in this process is unknown.We analyzed the phenotypes of circulating T cells and the proliferative responses of peripheral-blood mononuclear cells to phytohemagglutinin in 83 patients with severe combined immunodeficiency who received allogeneic marrow transplants without T-cell ablation from related donors over an 18-year period. We also tested for the presence of episomes of T-cell antigen receptors (extrachromosomal DNA circles formed during intrathymic T-cell development) to assess thymus-dependent T-cell reconstitution.Before and early after transplantation, the numbers of circulating T cells were low, with a predominance of mature CD45RO+ T cells (primarily resulting from the transplacental transfer of maternal cells); T-cell antigen-receptor episomes were undetectable in peripheral-blood mononuclear cells. In 73 of the infants, thymus-derived T cells expressing CD45RA and T-cell antigen-receptor episomes were detected within three to six weeks after transplantation. The mean (+/-SD) value for thymus-dependent T-cell antigen-receptor episomes peaked (at 7311+/-8652 per microgram of peripheral-blood mononuclear-cell DNA) 1 to 2 years after transplantation and declined to low levels (less than 100 episomes per microgram of DNA) within 14 years, as compared with a gradual decline from birth to the age of about 80 years in normal subjects.The vestigial thymus in infants with severe combined immunodeficiency is functional and can produce enough T cells after bone marrow transplantation to provide normal immune function.
- Published
- 2000
35. Whole-Exome Sequencing Reveals IKBKB As a Cause of Combined Immunodeficiency
- Author
-
Thomas J. Urban, Talal Mousallem, Roberta E. Parrott, Xiao-Ping Zhong, David Goldstein, Hongxia Wang, Joseph L. Roberts, Rebecca H. Buckley, and Jialong Yang
- Subjects
Immunology ,medicine ,Immunology and Allergy ,Computational biology ,Biology ,medicine.disease ,Immunodeficiency ,Exome sequencing - Published
- 2014
- Full Text
- View/download PDF
36. Next Generation Sequencing May Be More Efficient and Economical Than Targeted Gene Testing in Patients with Primary Immune Deficiency (PID)
- Author
-
Talal Mousallem, Joseph L. Roberts, Rebecca H. Buckley, Roberta E. Parrott, David Goldstein, and Thomas J. Urban
- Subjects
Genetics ,Primary (chemistry) ,Immune system ,Immunology ,Immunology and Allergy ,PID controller ,In patient ,Biology ,Gene ,DNA sequencing - Published
- 2013
- Full Text
- View/download PDF
37. CD45 Deficiency Caused by Uniparental Disomy, a Novel Cause of Severe Combined Immunodeficiency
- Author
-
Qiong Wei, David L. Wiest, Suraj Peri, Roberta E. Parrott, Alla Lapidus, Jinwook Shin, Joseph L. Roberts, Roland L. Dunbrack, Joseph R. Testa, Jianming Pei, Xiao-Ping Zhong, Biao Luo, and Rebecca H. Buckley
- Subjects
Severe combined immunodeficiency ,business.industry ,Immunology ,medicine ,Immunology and Allergy ,medicine.disease ,business ,Uniparental disomy - Published
- 2012
- Full Text
- View/download PDF
38. 784 Thymic output after non-ablative allogeneic hematopoietic stem cell transplantation in infants with severe combined immunodeficiency (SCID)
- Author
-
Maria E. Gooding, Roberta E. Parrott, Barton F. Haynes, Kimberly M. Curtis, Rebecca H. Buckley, and Dhavalkumar D. Patel
- Subjects
Severe combined immunodeficiency ,business.industry ,medicine.medical_treatment ,Immunology ,medicine ,Immunology and Allergy ,Non ablative ,Hematopoietic stem cell transplantation ,medicine.disease ,business - Published
- 2000
- Full Text
- View/download PDF
39. T-B+NK+ Severe Combined Immunodeficiency Caused by Complete Deficiency of the CD3zeta Subunit of the T Cell Antigen Receptor Complex
- Author
-
R.H. Bucley, Chan M. Win, Michael D. Keller, David L. Wiest, Roberta E. Parrott, Joseph L. Roberts, Jeffery H. Carpenter, Marcella Sarzotti, Myriah Cooney, Xiao-Ping Zhong, E.O. Sajaroff, and Jens Peter Holst Lauritsen
- Subjects
Receptor complex ,Severe combined immunodeficiency ,Protein subunit ,Immunology ,T-cell receptor ,medicine ,Immunology and Allergy ,T-cell Antigen ,Biology ,medicine.disease ,Virology - Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.