Clinical Focus


  • Pediatric Hematology-Oncology
  • Pediatric Stem Cell Transplantation, Genetic Immune Defects

Administrative Appointments


  • Attending Physician, Pediatric SCTRM Stanford School of Medicine (2016 - Present)
  • Assistant Professor, Pediatrics Stanford School of Medicine (2012 - 2016)

Honors & Awards


  • Uytengsu-Hamilton Faculty Scholar Award, Stanford Maternal & Child Health Research Institute (2019-Endowed)
  • California Institute of Regenerative Medicine Discovery 2 Award, CIRM (2019-2020)
  • Baxter Faculty Scholar, Stanford Medicine (2017-2018)
  • Mentored Clinical Scientist Research Career Development Award (K08), NIAID (2016-21)
  • Research Fellowship Award, Charles King Trust (2013-15)
  • Amy-Potter Research Fellowship Award, Boston Children’s Hospital, Harvard Medical School (2013-14)
  • Research Fellowship Award, Primary Immunodeficiency Treatment Consortium (2013-14)
  • Amy-Potter Research Fellowship Award, Boston Children’s Hospital, Harvard Medical School (2012-13)
  • Dan Heller Teaching Award, Harvard Medical School (2009)

Boards, Advisory Committees, Professional Organizations


  • Member, 22q11.2 Society (2017 - Present)
  • Member, International Society of Stem Cell Research (ISSCR) (2014 - Present)
  • Member, Primary Immunodeficiency Treatment Consortium (PIDTC) (2012 - Present)
  • Member, American Society of Pediatric Hematology/Oncology (ASPHO) (2009 - Present)
  • Member, Children’s Oncology Group (COG) (2009 - Present)
  • Associate Member, American Society of Hematology (ASH) (2009 - Present)

Professional Education


  • Board Certification: American Board of Pediatrics, Pediatrics (2021)
  • Board Certification: American Board of Pediatrics, Pediatric Hematology-Oncology (2020)
  • Fellowship: Boston Childrens Hospital Pediatric Hematology and Oncology Fellowship (2012) MA
  • Residency: Mass General Hospital for Children Pediatric Residency (2009) MA
  • Board Certification, American Board of Pediatric Hematology-Oncology, 2013
  • Board Certification, American Board of Pediatrics, 2009
  • Research Fellowship, Pediatric Immunology, Laboratory Dr. Luigi D. Notarangelo, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 2010-2012
  • Clinical Fellowship, Pediatric Hematology/Oncology, Boston Children’s Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 2009-2012
  • Residency, Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 2006-2009
  • Postdoctoral Fellow, Channing Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 2005-2006
  • PhD (Medicine), Technische Universitaet Muenchen, School of Medicine, Munich, Germany, 2004
  • MD, Technische Universitaet Muenchen, School of Medicine, Munich, Germany, 2002

Patents


  • Comstock LE, Coyne MJ, Weinacht KG, Kasper DL, Tzianabos AO. "United States Patent 10/388,390 Method of Overexpression of Zwitterionic Polysaccharides", Brigham and Women’s Hospital, Mar 13, 2003

Current Research and Scholarly Interests


I am a pediatric hematologist-oncologist with special interest in the niche of diseases that intersect immune dysfunction, primary immunodeficiency and bone marrow failure. My clinical practice focuses on pediatric patients requiring a hematopoietic stem cell transplantation, patients with DiGeorge Syndrome and patients with genetic immune diseases presenting with autoimmunity. As a physician-scientist, I strive to advance our insights into the mechanisms leading to immunodeficiency, autoimmunity and tolerance on a molecular level and to translate our research into novel targeted therapies patients.

My work is a natural extension of my clinical training in pediatric hematology-oncology combined with my scientific background in immunology and microbiology. After completing my clinical training at Boston Children’s Hospital/Dana-Farber Cancer Institute, I joined the laboratory of Luigi D. Notarangelo in the division of immunology, Boston Children’s Hospital/Harvard Stem Cell Institute, where I have acquired skills in the field of reprogramming, tissue engineering and gene correction. In my laboratory, we now use iPSC-based disease models to study how defects in mitochondrial metabolism and oxidative stress affect hematopoietic stem and progenitor cell development and cell death with the goal of identifying therapeutic targets. A separate focus of my laboratory is devoted to understanding the thymic developmental defects in DiGeorge syndrome.

Clinical Trials


  • Natural History Study of SCID Disorders Recruiting

    This study is a prospective evaluation of children with Severe Combined Immune Deficiency (SCID) who are treated under a variety of protocols used by participating institutions. In order to determine the patient, recipient and transplant-related variables that are most important in determining outcome, study investigators will uniformly collect pre-, post- and peri-transplant (or other treatment) information on all children enrolled into this study. Children will be divided into three strata: - Stratum A: Typical SCID with virtual absence of autologous T cells and poor T cell function - Stratum B: Atypical SCID (leaky SCID, Omenn syndrome and reticular dysgenesis with limited T cell diversity or number and reduced function), and - Stratum C: ADA deficient SCID and XSCID patients receiving alternative therapy including PEG-ADA ERT or gene therapy. Each Group/Cohort Stratum will be analyzed separately.

    View full details

  • Patients Treated for Chronic Granulomatous Disease (CGD) Since 1995 Recruiting

    Chronic granulomatous disease (CGD) is an inherited immune system abnormality in which bone marrow transplantation (BMT) has been shown to be curative. However the risks of transplantation are high and not all patients with CGD may need to undergo this high risk procedure. This study will determine the long term medical condition and daily functioning of participants with CGD after a transplant and if possible, compare these results to participants who do not undergo a transplant.

    View full details

  • Patients Treated for SCID (1968-Present) Recruiting

    Individuals with a past diagnosis of severe combined immune deficiency (including many cases of "leaky SCID", Omenn syndrome, and reticular dysgenesis) who have undergone blood and marrow transplant, gene therapy, or enzyme replacement in the past may be eligible for this study. The purpose of study is to look backwards at what has already been done in the. Over 800 patients with SCID are expected to be enrolled, making this one of the largest studies ever to describe outcomes for patients with SCID treated at many different hospitals around North America.

    View full details

  • Patients Treated for Wiskott-Aldrich Syndrome (WAS) Since 1990 Not Recruiting

    Wiskott - Aldrich syndrome (WAS) is a rare serious medical condition that causes problems both with the immune system and with easy bruising and bleeding. The immune abnormalities cause patients with WAS to be very susceptible to infections. Depending on the specific type of primary immune deficiency diseases, there are effective treatments, including antibiotics, cellular therapy and gene therapy, but studies of large numbers of patients are needed to determine the full range of causes, natural history, or the best methods of treatment for long term success. This multicenter study combines retrospective, prospective and cross-sectional analyses of the transplant experiences for patients with WAS who have already received HCT since 1990, or who will undergo Hematopoietic cell transplant (HCT) during the study period. The retrospective and prospective portions of the study will address the impact of a number of pre and post-transplant factors on post-transplant disease correction and ultimate benefit from HCT and the cross-sectional portion of the study will assess the benefit of HCT 2 years post-HCT in consenting surviving patients.

    Stanford is currently not accepting patients for this trial. For more information, please contact Matthew Porteus, MD, 650-725-6520.

    View full details

Stanford Advisees


All Publications


  • The rise of haplo: a quest for the perfect graft. Blood Weinacht, K. G. 2024; 143 (3): 193-195

    View details for DOI 10.1182/blood.2023022428

    View details for PubMedID 38236615

  • Accelerating Immune Reconstitution in HSCT Patients through iPSC-Derived Thymic Epithelial Cells Mohammed, A., Hanh Dan Nguyen, Hubka, K., Wang, W., Slepicka, P., Solomon, B., Arreola, M., Zheng, Z., Gentles, A., Weinacht, K. G. AMER SOC HEMATOLOGY. 2023
  • Loss of Metabolic Control Beyond the Promyelocyte Stage Resolves Myeloid Maturation Arrest in Reticular Dysgenesis Wang, W., Arreola, M., Mathews, T., Devilbiss, A., Zhao, Z., Morrison, S., Weinacht, K. G. AMER SOC HEMATOLOGY. 2023
  • Precision Delivery of Steroids as a Rescue Therapy for Gastrointestinal Graft-versus-Host Disease in Pediatric Stem Cell Transplant Recipients. Journal of clinical medicine Levitte, S., Ganguly, A., Frolik, S., Guevara-Tique, A. A., Patel, S., Tadas, A., Klein, O., Shyr, D., Agarwal-Hashmi, R., Beach, L., Callard, E., Weinacht, K., Bertaina, A., Thakor, A. S. 2023; 12 (13)

    Abstract

    Graft versus host disease (GVHD) is one of the most serious complications following stem cell transplant in children and is a major cause of morbidity and mortality. Corticosteroids remain the mainstay of treatment, and although a majority of children respond to systemic steroids, those refractory to or dependent upon corticosteroids suffer from complications secondary to long-term steroid administration. This problem has prompted consideration of steroid-sparing treatment strategies, although the time to clinical remission can be variable. Intraarterial corticosteroid delivery has been used in adults as a rescue therapy in steroid-resistant patients, but its use in children has been limited. We investigated the feasibility of intraarterial steroid administration into the bowel and/or liver in a cohort of six pediatric patients with acute GVHD. All patients successfully underwent treatment with no serious adverse effects. Five of five (100%) patients with gastrointestinal bleeding due to GVHD had rapid symptom improvement by 48 h, which was durable up to three weeks. Three of four (75%) patients with hepatic GVHD had improved cholestasis following intraarterial steroid administration. Our experience with this small cohort preliminarily demonstrated the feasibility and safety of intraarterial steroid administration in children with acute GVHD. This approach warrants consideration as a rescue therapy in steroid-refractory cases and as a "bridge" therapy for children with severe acute GVHD who are transitioning to steroid-sparing regimens.

    View details for DOI 10.3390/jcm12134229

    View details for PubMedID 37445274

  • Precision Delivery of Steroids as a Rescue Therapy for Gastrointestinal Graft-versus-Host Disease in Pediatric Stem Cell Transplant Recipients Journal of Clinical Medicine Levitte, s., Ganguly, A., Frolik, S., Guevara-Tique, A., et al 2023; 12 (4229)

    View details for DOI 10.3390/jcm12134229

  • Case report: Refractory Evans syndrome in two patients with spondyloenchondrodysplasia with immune dysregulation treated successfully with JAK1/JAK2 inhibition. Frontiers in immunology Gernez, Y., Narula, M., Cepika, A., Valdes Camacho, J., Hoyte, E. G., Mouradian, K., Glader, B., Singh, D., Sathi, B., Rao, L., Tolin, A. L., Weinberg, K. I., Lewis, D. B., Bacchetta, R., Weinacht, K. G. 2023; 14: 1328005

    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.

    View details for DOI 10.3389/fimmu.2023.1328005

    View details for PubMedID 38347954

  • Human JAK1 gain of function causes dysregulated myelopoeisis and severe allergic inflammation. JCI insight Biggs, C. M., Cordeiro-Santanach, A., Prykhozhij, S. V., Deveau, A. P., Lin, Y., Del Bel, K. L., Orben, F., Ragotte, R. J., Saferali, A., Mostafavi, S., Dinh, L., Dai, D., Weinacht, K. G., Dobbs, K., Ott de Bruin, L., Sharma, M., Tsai, K., Priatel, J. J., Schreiber, R. A., Rozmus, J., Hosking, M. C., Shopsowitz, K. E., McKinnon, M. L., Vercauteren, S., Seear, M., Notarangelo, L. D., Lynn, F. C., Berman, J. N., Turvey, S. E. 2022; 7 (24)

    Abstract

    Primary atopic disorders are a group of inborn errors of immunity that skew the immune system toward severe allergic disease. Defining the biology underlying these extreme monogenic phenotypes reveals shared mechanisms underlying common polygenic allergic disease and identifies potential drug targets. Germline gain-of-function (GOF) variants in JAK1 are a cause of severe atopy and eosinophilia. Modeling the JAK1GOF (p.A634D) variant in both zebrafish and human induced pluripotent stem cells (iPSCs) revealed enhanced myelopoiesis. RNA-Seq of JAK1GOF human whole blood, iPSCs, and transgenic zebrafish revealed a shared core set of dysregulated genes involved in IL-4, IL-13, and IFN signaling. Immunophenotypic and transcriptomic analysis of patients carrying a JAK1GOF variant revealed marked Th cell skewing. Moreover, long-term ruxolitinib treatment of 2 children carrying the JAK1GOF (p.A634D) variant remarkably improved their growth, eosinophilia, and clinical features of allergic inflammation. This work highlights the role of JAK1 signaling in atopic immune dysregulation and the clinical impact of JAK1/2 inhibition in treating eosinophilic and allergic disease.

    View details for DOI 10.1172/jci.insight.150849

    View details for PubMedID 36546480

  • Functional Human iPSC-Derived Thymic Epithelial Progenitor Cells Reconstitute T Cell Development and Function in an In Vivo Model of Thymic Aplasia Slepicka, P., Hubka, K. M., Hanh Dan Nguyen, Mohammed, A., Wang, J., Gifford, C., Sebastiano, V., Weinacht, K. G. AMER SOC HEMATOLOGY. 2022: 7340-7341
  • Epigenetic and Immunological Indicators of IPEX Disease in subjects with FOXP3 gene mutation. The Journal of allergy and clinical immunology Narula, M., Lakshmanan, U., Borna, S., Schulze, J. J., Holmes, T. H., Harre, N., Kirkey, M., Ramachandran, A., Tagi, V. M., Barzaghi, F., Grunebaum, E., Upton, J. E., Hong-Diep Kim, V., Wysocki, C., Dimitriades, V. R., Weinberg, K., Weinacht, K. G., Gernez, Y., Sathi, B. K., Schelotto, M., Johnson, M., Olek, S., Sachsenmaier, C., Roncarolo, M. G., Bacchetta, R. 2022

    Abstract

    Forkhead-Box-Protein-3 (FOXP3) is the master transcription factor in CD4+CD25hiCD127lo regulatory T (Treg) cells. Mutations in FOXP3 result in IPEX (Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-linked) syndrome. Clinical presentation of IPEX syndrome is broader than initially described, challenging the understanding of the disease, its evolution and treatment choice.To study the type and extent of immunological abnormalities which remain ill-defined in IPEX, across genetic and clinical heterogeneity.We performed Treg-specific epigenetic quantification and immunological characterization of severe "typical" (n=6) and "atypical" or asymptomatic (n=9) IPEX patients.Increased number of cells with Treg-Specific Demethylated Region (TSDR) demethylation in FOXP3 is a consistent feature in IPEX patients, with i) highest values in those with typical IPEX, ii) increased values in subjects with pathogenic FOXP3 but still no symptoms, and iii) gradual increase over the course of disease progression. Large scale profiling using Luminex identified plasma inflammatory signature of macrophage activation and Th2 polarization, with cytokines previously not associated with IPEX pathology, including CCL22, CCL17, CCL15, and IL-13, and the inflammatory markers TNFα, IL-1A, IL-8, sFasL, and CXCL9. Similarly, both Treg and Teff compartments, studied by CyTOF, were skewed towards the Th2 compartment, especially in typical IPEX.Elevated 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.IPEX-specific epigenetic and immunologic changes provide invaluable tools that, complementing the genetic diagnosis, allow monitoring disease progression and enable early treatment interventions.

    View details for DOI 10.1016/j.jaci.2022.09.013

    View details for PubMedID 36152823

  • A case of Spondyloenchondrodysplasia with immune dysregulation presenting as Systemic Lupus Erythematous Camacho, J., Singh, D., Bacchetta, R., Weinberg, K., Cepika, A., Narula, M., Lewis, D. B., Gernez, Y., Weinacht, K. SPRINGER/PLENUM PUBLISHERS. 2021: S18–S19
  • A challenging case of recurrent idiopathic hemophagocytic lymphohistiocytosis (HLH) initially presenting in an infant with Pneumocystis jirovecii pneumonia Solomon, B., Balagtas, J., Chien, M., Pooni, R., Balboni, I., Weinacht, K., Gernez, Y. SPRINGER/PLENUM PUBLISHERS. 2021: S55
  • Correction: Chronic Granulomatous Disease-Associated IBD Resolves and Does Not Adversely Impact Survival Following Allogeneic HCT. Journal of clinical immunology Marsh, R. A., Leiding, J. W., Logan, B. R., Griffith, L. M., Arnold, D. E., Haddad, E., Falcone, E. L., Yin, Z., Patel, K., Arbuckle, E., Bleesing, J. J., Sullivan, K. E., Heimall, J., Burroughs, L. M., Skoda-Smith, S., Chandrakasan, S., Yu, L. C., Oshrine, B. R., Cuvelier, G. D., Thakar, M. S., Chen, K., Teira, P., Shenoy, S., Phelan, R., Forbes, L. R., Martinez, C., Chellapandian, D., Davila Saldana, B. J., Shah, A. J., Weinacht, K. G., Joshi, A., Boulad, F., Quigg, T. C., Dvorak, C. C., Grossman, D., Torgerson, T., Graham, P., Prasad, V., Knutsen, A., Chong, H., Miller, H., de la Morena, M. T., DeSantes, K., Cowan, M. J., Notarangelo, L. D., Kohn, D. B., Stenger, E., Pai, S., Routes, J. M., Puck, J. M., Kapoor, N., Pulsipher, M. A., Malech, H. L., Parikh, S., Kang, E. M., submitted on behalf of the Primary Immune Deficiency Treatment Consortium 2020

    Abstract

    The original version of this article unfortunately contained the missing author, Caridad Martinez. The authors would like to correct the list. We apologize for any inconvenience that this may have caused. The correct author list is shown above.

    View details for DOI 10.1007/s10875-020-00852-0

    View details for PubMedID 32860171

  • Insight into the Pediatric and Adult Dichotomy of COVID-19: Age-Related Differences in the Immune Response to SARS-CoV-2 infection. Pediatric pulmonology Fialkowski, A., Gernez, Y., Arya, P., Weinacht, K. G., Bernard Kinane, T., Yonker, L. M. 2020

    Abstract

    The difference in morbidity and mortality between adult and pediatric COVID-19 infections is dramatic. Understanding pediatric-specific acute and delayed immune responses to SARS-CoV-2 is critical for the development of vaccination strategies, immune-targeted therapies, and treatment and prevention of MIS-C. The goal of this review is to highlight research developments in understanding of the immune responses to SARS-CoV-2 infections, with a specific focus on age-related immune responses. This article is protected by copyright. All rights reserved.

    View details for DOI 10.1002/ppul.24981

    View details for PubMedID 32710693

  • Artificial thymic organoids represent a reliable tool to study T-cell differentiation in patients with severe T-cell lymphopenia. Blood advances Bosticardo, M., Pala, F., Calzoni, E., Delmonte, O. M., Dobbs, K., Gardner, C. L., Sacchetti, N., Kawai, T., Garabedian, E. K., Draper, D., Bergerson, J. R., DeRavin, S. S., Freeman, A. F., Gungor, T., Hartog, N., Holland, S. M., Kohn, D. B., Malech, H. L., Markert, M. L., Weinacht, K. G., Villa, A., Seet, C. S., Montel-Hagen, A., Crooks, G. M., Notarangelo, L. D. 2020; 4 (12): 2611–16

    Abstract

    The study of early T-cell development in humans is challenging because of limited availability of thymic samples and the limitations of in vitro T-cell differentiation assays. We used an artificial thymic organoid (ATO) platform generated by aggregating a DLL4-expressing stromal cell line (MS5-hDLL4) with CD34+ cells isolated from bone marrow or mobilized peripheral blood to study T-cell development from CD34+ cells of patients carrying hematopoietic intrinsic or thymic defects that cause T-cell lymphopenia. We found that AK2 deficiency is associated with decreased cell viability and an early block in T-cell development. We observed a similar defect in a patient carrying a null IL2RG mutation. In contrast, CD34+ cells from a patient carrying a missense IL2RG mutation reached full T-cell maturation, although cell numbers were significantly lower than in controls. CD34+ cells from patients carrying RAG mutations were able to differentiate to CD4+CD8+ cells, but not to CD3+TCRalphabeta+ cells. Finally, normal T-cell differentiation was observed in a patient with complete DiGeorge syndrome, consistent with the extra-hematopoietic nature of the defect. The ATO system may help determine whether T-cell deficiency reflects hematopoietic or thymic intrinsic abnormalities and define the exact stage at which T-cell differentiation is blocked.

    View details for DOI 10.1182/bloodadvances.2020001730

    View details for PubMedID 32556283

  • Excellent Outcomes Following Hematopoietic Cell Transplantation for Wiskott-Aldrich Syndrome: A PIDTC Report. Blood Burroughs, L., Petrovic, A., Brazauskas, R., Liu, X., Griffith, L. M., Ochs, H. D., Bleesing, J., Edwards, S., Dvorak, C. C., Chaudhury, S., Prockop, S., Quinones, R., Goldman, F., Quigg, T., Chandrakasan, S., Smith, A. R., Parikh, S. H., Davila Saldana, B. J., Thakar, M. S., Phelan, R., Shenoy, S., Forbes, L. R., Martinez, C. A., Chellapandian, D., Shereck, E., Miller, H., Kapoor, N., Barnum, J. L., Chong, H., Shyr, D., Chen, K., Abu-Arja, R. F., Shah, A., Weinacht, K., Moore, T. B., Joshi, A., DeSantes, K., Gillio, A. P., Cuvelier, G. D., Keller, M. D., Rozmus, J., Torgerson, T. R., Pulsipher, M. A., Haddad, E., Sullivan, K., Logan, B. R., Kohn, D. B., Puck, J. M., Notarangelo, L. D., Pai, S., Rawlings, D., Cowan, M. J. 2020

    Abstract

    Wiskott-Aldrich syndrome (WAS) is an X-linked disease caused by mutations in the WAS gene leading to thrombocytopenia, eczema, recurrent infections, autoimmune disease, and malignancy. Hematopoietic cell transplantation (HCT) is the primary curative approach, with the goal of correcting the underlying immunodeficiency and thrombocytopenia. HCT outcomes have improved over time, particularly for patients with HLA-matched sibling and unrelated donors. Here we report the outcomes of 129 patients with WAS who underwent HCT at 29 Primary Immune Deficiency Treatment Consortium centers between 2005 and 2015. Median age at HCT was 1.2 years. Most patients (65%) received myeloablative busulfan-based conditioning. With a median follow-up of 4.5 years, the 5-year overall survival (OS) was 91%. Superior 5-year OS was observed in patients <5 vs. ≥5 years old at the time of HCT (94% vs. 66%, overall p=0.0008). OS was excellent regardless of donor type even in cord blood recipients (90%). Conditioning intensity did not impact OS, but was associated with donor T-cell and myeloid engraftment post-HCT. Specifically, patients who received fludarabine/melphalan-based reduced-intensity regimens were more likely to have donor myeloid chimerism <50% early post-HCT. In addition, higher platelet counts were observed among recipients who achieved full (>95%) versus low-level (5%-49%) donor myeloid engraftment. In summary, HCT outcomes for WAS have improved since 2005 compared to prior reports. HCT at a younger age continues to be associated with superior outcomes supporting the recommendation for early HCT. High-level donor myeloid engraftment is important for platelet reconstitution following either myeloablative or busulfan-containing reduced intensity conditioning. (www.clinicaltrials.gov NCT02064933.).

    View details for DOI 10.1182/blood.2019002939

    View details for PubMedID 32268350

  • Autosomal Dominant JAK1 Gain-Of-Function Mutation Drives Myelopoiesis and Dysregulated T Helper Responses Leading To Severe Allergic Inflammation That Is Clinically Responsive To Ruxolitinib Biggs, C., Lin, E., Del Bel, K. L., Orben, F., Ragotte, R., Saferali, A., Mostafavi, S., Weinacht, K., de Bruin, L., Sharma, M., Shopsowitz, K., McKinnon, M. L., Vercauteren, S., Notarangelo, L., Lynn, F., Turvey, S. SPRINGER/PLENUM PUBLISHERS. 2020: S120
  • A Case of G6PC3 Congenital Neutropenia, Misdiagnosed As Evans Syndrome Camacho, J., Brar, R., Chapman, C., Fernandez-Pol, S., Weinacht, K., Gernez, Y. SPRINGER/PLENUM PUBLISHERS. 2020: S131–S132
  • Hematopoietic Cell Transplantation in Patients With Primary Immune Regulatory Disorders (PIRD): A Primary Immune Deficiency Treatment Consortium (PIDTC) Survey. Frontiers in immunology Chan, A. Y., Leiding, J. W., Liu, X., Logan, B. R., Burroughs, L. M., Allenspach, E. J., Skoda-Smith, S., Uzel, G., Notarangelo, L. D., Slatter, M., Gennery, A. R., Smith, A. R., Pai, S., Jordan, M. B., Marsh, R. A., Cowan, M. J., Dvorak, C. C., Craddock, J. A., Prockop, S. E., Chandrakasan, S., Kapoor, N., Buckley, R. H., Parikh, S., Chellapandian, D., Oshrine, B. R., Bednarski, J. J., Cooper, M. A., Shenoy, S., Davila Saldana, B. J., Forbes, L. R., Martinez, C., Haddad, E., Shyr, D. C., Chen, K., Sullivan, K. E., Heimall, J., Wright, N., Bhatia, M., Cuvelier, G. D., Goldman, F. D., Meyts, I., Miller, H. K., Seidel, M. G., Vander Lugt, M. T., Bacchetta, R., Weinacht, K. G., Andolina, J. R., Caywood, E., Chong, H., de la Morena, M. T., Aquino, V. M., Shereck, E., Walter, J. E., Dorsey, M. J., Seroogy, C. M., Griffith, L. M., Kohn, D. B., Puck, J. M., Pulsipher, M. A., Torgerson, T. R. 2020; 11: 239

    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.

    View details for DOI 10.3389/fimmu.2020.00239

    View details for PubMedID 32153572

  • Adenylate Kinase 2 Links Energy Metabolism and Cell Fate in Hematopoietic Stem and Progenitor Cells Wang, W., Awani, A., Devilbiss, A., Mathews, T., Thomas, D., Dever, D. P., Porteus, M., Morrison, S., Weinacht, K. G. AMER SOC HEMATOLOGY. 2019
  • Chronic Granulomatous Disease-Associated IBD Resolves and Does Not Adversely Impact Survival Following Allogeneic HCT. Journal of clinical immunology Marsh, R. A., Leiding, J. W., Logan, B. R., Griffith, L. M., Arnold, D. E., Haddad, E., Falcone, E. L., Yin, Z., Patel, K., Arbuckle, E., Bleesing, J. J., Sullivan, K. E., Heimall, J., Burroughs, L. M., Skoda-Smith, S., Chandrakasan, S., Yu, L. C., Oshrine, B. R., Cuvelier, G. D., Thakar, M. S., Chen, K., Teira, P., Shenoy, S., Phelan, R., Forbes, L. R., Chellapandian, D., Davila Saldana, B. J., Shah, A. J., Weinacht, K. G., Joshi, A., Boulad, F., Quigg, T. C., Dvorak, C. C., Grossman, D., Torgerson, T., Graham, P., Prasad, V., Knutsen, A., Chong, H., Miller, H., de la Morena, M. T., DeSantes, K., Cowan, M. J., Notarangelo, L. D., Kohn, D. B., Stenger, E., Pai, S., Routes, J. M., Puck, J. M., Kapoor, N., Pulsipher, M. A., Malech, H. L., Parikh, S., Kang, E. M., submitted on behalf of the Primary Immune Deficiency Treatment Consortium 2019

    Abstract

    INTRODUCTION: Inflammatory bowel disease (IBD) affects approximately 1/3 of patients with chronic granulomatous disease (CGD). Comprehensive investigation of the effect of allogeneic hematopoietic cell transplantation (HCT) on CGD IBD and the impact of IBD on transplant outcomes is lacking.METHODS: We collected data retrospectively from 145 patients with CGD who had received allogeneic HCT at 26 Primary Immune Deficiency Treatment Consortium (PIDTC) centers between January 1, 2005 and June 30, 2016.RESULTS: Forty-nine CGD patients with IBD and 96 patients without IBD underwent allogeneic HCT. Eighty-nine percent of patients with IBD and 93% of patients without IBD engrafted (p=0.476). Upper gastrointestinal acute GVHD occurred in 8.5% of patients with IBD and 3.5% of patients without IBD (p=0.246). Lower gastrointestinal acute GVHD occurred in 10.6% of patients with IBD and 11.8% of patients without IBD (p=0.845). The cumulative incidence of acute GVHD grades II-IV was 30% (CI 17-43%) in patients with IBD and 20% (CI 12-29%) in patients without IBD (p=0.09). Five-year overall survival was equivalent for patients with and without IBD: 80% [CI 66-89%] and 83% [CI 72-90%], respectively (p=0.689). All 33 surviving evaluable patients with a history of IBD experienced resolution of IBD by 2years following allogeneic HCT.CONCLUSIONS: In this cohort, allogeneic HCT was curative for CGD-associated IBD. IBD should not contraindicate HCT, as it does not lead to an increased risk of mortality. This study is registered at clinicaltrials.gov NCT02082353.

    View details for DOI 10.1007/s10875-019-00659-8

    View details for PubMedID 31376032

  • Severe autoinflammation in four patients with C-terminal variants in CDC42 successfully treated with IL-1beta inhibition. The Journal of allergy and clinical immunology Gernez, Y., de Jesus, A. A., Alsaleem, H., Macaubas, C., Roy, A., Lovell, D., Jagadeesh, K. A., Alehashemi, S., Erdman, L., Grimley, M., Talarico, S., Bacchetta, R., Lewis, D. B., Canna, S. W., Laxer, R. M., Mellins, E. D., Goldbach-Mansky, R., Weinacht, K. G. 2019

    Abstract

    Four patients with novel variants in the C-terminal domain of CDC42, severe autoinflammation, constitutive elevation of serum interleukin 18, and predisposition to macrophage activation syndrome respond to treatment with interleukin-1beta signaling inhibition.

    View details for DOI 10.1016/j.jaci.2019.06.017

    View details for PubMedID 31271789

  • Combined liver and hematopoietic stem cell transplantation in patients with X-linked hyper-IgM syndrome JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY Bucciol, G., Nicholas, S. K., Calvo, P., Cant, A., Edgar, J. M., Espanol, T., Ferrua, F., Galicchio, M., Gennery, A. R., Hadzic, N., Hanson, I., Kusminsky, G., Lange, A., Lanternier, F., Mahlaoui, N., Moshous, D., Nademi, Z., Neven, B., Oleastro, M., Porta, F., Quarello, P., Silva, M., Slatter, M. A., Soncini, E., Stefanowicz, M., Tandoi, F., Teisseyre, M., Torgerson, T. R., Veys, P., Weinacht, K. G., Wolska-Kusnierz, B., Pirenne, J., de la Morena, M., Meyts, I. 2019; 143 (5): 1952-+
  • Calm in the midst of cytokine storm: a collaborative approach to the diagnosis and treatment of hemophagocytic lymphohistiocytosis and macrophage activation syndrome. Pediatric rheumatology online journal Halyabar, O., Chang, M. H., Schoettler, M. L., Schwartz, M. A., Baris, E. H., Benson, L. A., Biggs, C. M., Gorman, M., Lehmann, L., Lo, M. S., Nigrovic, P. A., Platt, C. D., Priebe, G. P., Rowe, J., Sundel, R. P., Surana, N. K., Weinacht, K. G., Mann, A., Yuen, J. C., Meleedy-Rey, P., Starmer, A., Banerjee, T., Dedeoglu, F., Degar, B. A., Hazen, M. M., Henderson, L. A. 2019; 17 (1): 7

    Abstract

    BACKGROUND: Hemophagocytic lymphohistiocytosis (HLH) and macrophage activation syndrome (MAS) were historically thought to be distinct entities, often managed in isolation. In fact, these conditions are closely related. A collaborative approach, which incorporates expertise from subspecialties that previously treated HLH/MAS independently, is needed. We leveraged quality improvement (QI) techniques in the form of an Evidence-Based Guideline (EBG) to build consensus across disciplines on the diagnosis and treatment of HLH/MAS.METHODS: A multidisciplinary work group was convened that met monthly to develop the HLH/MAS EBG. Literature review and expert opinion were used to develop a management strategy for HLH/MAS. The EBG was implemented, and quality metrics were selected to monitor outcomes.RESULTS: An HLH/MAS clinical team was formed with representatives from subspecialties involved in the care of patients with HLH/MAS. Broad entry criteria for the HLH/MAS EBG were established and included fever and ferritin ≥500ng/mL. The rheumatology team was identified as the "gate-keeper," charged with overseeing the diagnostic evaluation recommended in the EBG. First-line medications were recommended based on the acuity of illness and risk of concurrent infection. Quality metrics to be tracked prospectively based on time to initiation of treatment and clinical response were selected.CONCLUSION: HLH/MAS are increasingly considered to be a spectrum of related conditions, and joint management across subspecialties could improve patient outcomes. Our experience in creating a multidisciplinary approach to HLH/MAS management can serve as a model for care at other institutions.

    View details for DOI 10.1186/s12969-019-0309-6

    View details for PubMedID 30764840

  • Novel CDC42 Mutation Causes Severe Autoinflammatory Syndrome Responsive to IL-1 Inhibition Gernez, Y., Kirbey, M., Balagas, J. M., Macaubas, C. I., Canna, S., Lewis, D. B., Mellins, E., Bacchetta, R., Weinacht, K. G. SPRINGER/PLENUM PUBLISHERS. 2019: S69
  • Artificial Thymic Organoids Represent a Reliable and Quick Tool to Study T Cell Differentiation in Human Bone Marrow Samples from Patients with Severe T Cell Immunodeficiency Bosticardo, M., Pala, F., Calzoni, E., Gardner, C., Dobbs, K., De Ravin, S., Hartog, N., Markert, M., Weinacht, K. G., Malech, H. L., Seet, C., Montel-Hagen, A., Crooks, G. M., Notarangelo, L. D. SPRINGER/PLENUM PUBLISHERS. 2019: S51–S52
  • Epigenetic Immune Cell Quantification for Diagnosis and Monitoring of Patients with Primary Immune Deficiencies and Immune Regulatory Disorders Schulze, J., Werner, J., Schildknecht, K., Lakshmanan, U., Gruetzkau, A., Chu, J., Gernez, Y., Speckmann, C., Weinacht, K. G., Bertaina, A., Baron, U., Borte, S., Olek, S., Bacchetta, R. SPRINGER/PLENUM PUBLISHERS. 2019: S30
  • Combined liver and hematopoietic stem cell transplantation in X-linked hyper IgM syndrome. The Journal of allergy and clinical immunology Bucciol, G., Nicholas, S. K., Calvo, P. L., Cant, A., Edgar, J. D., Espanol, T., Ferrua, F., Galicchio, M., Gennery, A. R., Hadzic, N., Hanson, I. C., Kusminsky, G., Lange, A., Lanternier, F., Mahlaoui, N., Moshous, D., Nademi, Z., Neven, B., Oleastro, M., Porta, F., Quarello, P., Silva, M., Slatter, M. A., Soncini, E., Stefanowicz, M., Tandoi, F., Teisseyre, M., Torgerson, T. R., Veys, P., Weinacht, K. G., Wolska-Kusnierz, B., Pirenne, J., de la Morena, M. T., Meyts, I. 2019

    Abstract

    Liver disease in X-linked hyper IgM syndrome (XHIGM) is an important predictor of mortality. In case liver transplantation (LT) is required, a survival benefit is observed when LT is combined with HSCT.

    View details for PubMedID 30682461

  • An Engineered Cell-Traceable Model of Reticular Dysgenesis in Human Hematopoietic Stem Cells Linking Metabolism and Differentiation Wang, W., Awani, A., Reich, L., Nakauchi, Y., Thomas, D., Dever, D. P., Porteus, M., Weinacht, K. G. AMER SOC HEMATOLOGY. 2018
  • Engineering Regenerative Thymic Tissues to Restore Long-Term T Cell Lymphopoiesis Gai, H., Gras-Pena, R., Verma, Y., Fateh, V., Ikeda, K., Dejene, B., Min, D., Wang, J., Swigut, T., Weinberg, K. I., Hollander, G. A., Heilshorn, S., Roncarolo, M., Sebastiano, V., Weinacht, K. G. AMER SOC HEMATOLOGY. 2018
  • Jakinibs for the treatment of immune dysregulation in patients with gain-of-function signal transducer and activator of transcription 1 (STAT1) or STAT3 mutations. The Journal of allergy and clinical immunology Forbes, L. R., Vogel, T. P., Cooper, M. A., Castro-Wagner, J., Schussler, E., Weinacht, K. G., Plant, A. S., Su, H. C., Allenspach, E. J., Slatter, M., Abinun, M., Lilic, D., Cunningham-Rundles, C., Eckstein, O., Olbrich, P., Guillerman, R. P., Patel, N. C., Demirdag, Y. Y., Zerbe, C., Freeman, A. F., Holland, S. M., Szabolcs, P., Gennery, A., Torgerson, T. R., Milner, J. D., Leiding, J. W. 2018

    View details for PubMedID 30092289

  • Recent outcome of hematopoietic cell transplantation for Wiskott-Aldrich syndrome is excellent in all donor types: A Primary Immune Deficiency Treatment Consortium (PIDTC) Study Pai, S., Brazauskas, R., Bleesing, J., Dvorak, C. C., Kletzel, M., Petrovic, A., Prockop, S. E., Quinones, R., Goldman, F. D., Quigg, T. C., Hanson, I. C., Stenger, E., Thakar, M. S., Saldana, B., Bednarski, J. J., Smith, A. R., Parikh, S. H., Leiding, J. W., Miller, H. K., Shereck, E., Chong, H. J., DeSantes, K., Cuvelier, G. E., Chen, K., Weinacht, K. G., Abu-Arja, R. F., Kapoor, N., Moore, T. B., Gillio, A. P., Joshi, A. Y., Barnum, J. L., Chandrakasan, S., Haddad, E., Keller, M., Rozmus, J., Sullivan, K., Griffith, L. M., Ochs, H., Notarangelo, L. D., Puck, J., Kohn, D. B., Cowan, M. J., Rawlings, D., Burroughs, L. SPRINGER/PLENUM PUBLISHERS. 2018: 399–400
  • Human Ipsc-Derived Thymic Epithelial Progenitor Cells as Stem Cell-Based Therapyto Restore Thymic Function in Hematopoietic Stem Cell Transplant Recipients Gai, H., Sebastiano, V., Weinacht, K. G. ELSEVIER SCIENCE INC. 2018: S364
  • Resolution of CGD Related Colitis after Allogeneic Hematopoietic Stem Cell Transplantation in Patients with Chronic Granulomatous Disease-Early Results From the 6903 Study of the Primary Immune Deficiency Treatment Consortium (PIDTC) Leiding, J. W., Logan, B. R., Yin, Z., Arbuckle, E., Bleesing, J. J., Sullivan, K. E., Heimall, J., Burroughs, L., Skoda-Smith, S., Chandrakasan, S., Yu, L. C., Oshrine, B. R., Cuvelier, G. E., Thakar, M., Chen, K., Shenoy, S., Saldana, B., Weinacht, K. G., Joshi, A., Boulad, F., Quigg, T. C., Dvorak, C. C., Knutsen, A., Chong, H., Miller, H. K., De la Morena, M., DeSantes, K., Cowan, M. J., Notarangelo, L. D., Kohn, D. B., Pai, S., Stenger, E., Puck, J., Kapoor, N., Pulsipher, M. A., Haddad, E., Griffith, L. M., Shearer, W., Malech, H. L., Parikh, S., Marsh, R. A., Kang, E. M. ELSEVIER SCIENCE INC. 2018: S53–S54
  • Ruxolitinib reverses Dysregulated T Helper Cell Responses and controls Autoimmunity caused by a Novel STAT1 Gain of Function Mutation Journal of Allergy and Clinical Immunology Weinacht, K. G., Charbonnier, L. M., Alroqi, F., Plant, A., Qiao, Q., Wu, H., Ma, C., Torgerson, T. R., Rosenweig, S. D., Flesier, T. A., Notarangelo, L. D., Hanson, I. C., Forbes, L., Chatila, T. A. 2017
  • Reticular dysgenesis-associated AK2 protects hematopoietic stem and progenitor cell development from oxidative stress JOURNAL OF EXPERIMENTAL MEDICINE Rissone, A., Weinacht, K. G., La Marca, G., Bishop, K., Giocaliere, E., Jagadeesh, J., Felgentreff, K., Dobbs, K., Al-Herz, W., Jones, M., Chandrasekharappa, S., Kirby, M., Wincovitch, S., Simon, K. L., Itan, Y., Devine, A., Schlaeger, T., Schambach, A., Sood, R., Notarangelo, L. D., Candotti, F. 2015; 212 (8): 1185-1202

    Abstract

    Adenylate kinases (AKs) are phosphotransferases that regulate the cellular adenine nucleotide composition and play a critical role in the energy homeostasis of all tissues. The AK2 isoenzyme is expressed in the mitochondrial intermembrane space and is mutated in reticular dysgenesis (RD), a rare form of severe combined immunodeficiency (SCID) in humans. RD is characterized by a maturation arrest in the myeloid and lymphoid lineages, leading to early onset, recurrent, and overwhelming infections. To gain insight into the pathophysiology of RD, we studied the effects of AK2 deficiency using the zebrafish model and induced pluripotent stem cells (iPSCs) derived from fibroblasts of an RD patient. In zebrafish, Ak2 deficiency affected hematopoietic stem and progenitor cell (HSPC) development with increased oxidative stress and apoptosis. AK2-deficient iPSCs recapitulated the characteristic myeloid maturation arrest at the promyelocyte stage and demonstrated an increased AMP/ADP ratio, indicative of an energy-depleted adenine nucleotide profile. Antioxidant treatment rescued the hematopoietic phenotypes in vivo in ak2 mutant zebrafish and restored differentiation of AK2-deficient iPSCs into mature granulocytes. Our results link hematopoietic cell fate in AK2 deficiency to cellular energy depletion and increased oxidative stress. This points to the potential use of antioxidants as a supportive therapeutic modality for patients with RD.

    View details for DOI 10.1084/jem.20141286

    View details for Web of Science ID 000360379400004

    View details for PubMedID 26150473

  • Diagnosis of immunodeficiency caused by a purine nucleoside phosphorylase defect by using tandem mass spectrometry on dried blood spots JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY La Marca, G., Canessa, C., Giocaliere, E., Romano, F., Malvagia, S., Funghini, S., Moriondo, M., Valleriani, C., Lippi, F., Ombrone, D., Della Bona, M. L., Speckmann, C., Borte, S., Brodszki, N., Gennery, A. R., Weinacht, K., Celmeli, F., Pagel, J., de Martino, M., Guerrini, R., Wittkowski, H., Santisteban, I., Bali, P., Ikinciogullari, A., Hershfield, M., Notarangelo, L. D., Resti, M., Azzari, C. 2014; 134 (1): 155-?

    Abstract

    Purine nucleoside phosphorylase (PNP) deficiency is a rare form of autosomal recessive combined primary immunodeficiency caused by a enzyme defect leading to the accumulation of inosine, 2'-deoxy-inosine (dIno), guanosine, and 2'-deoxy-guanosine (dGuo) in all cells, especially lymphocytes. Treatments are available and curative for PNP deficiency, but their efficacy depends on the early approach. PNP-combined immunodeficiency complies with the criteria for inclusion in a newborn screening program.This study evaluate whether mass spectrometry can identify metabolite abnormalities in dried blood spots (DBSs) from affected patients, with the final goal of individuating the disease at birth during routine newborn screening.DBS samples from 9 patients with genetically confirmed PNP-combined immunodeficiency, 10,000 DBS samples from healthy newborns, and 240 DBSs from healthy donors of different age ranges were examined. Inosine, dIno, guanosine, and dGuo were tested by using tandem mass spectrometry (TMS). T-cell receptor excision circle (TREC) and kappa-deleting recombination excision circle (KREC) levels were evaluated by using quantitative RT-PCR only for the 2 patients (patients 8 and 9) whose neonatal DBSs were available.Mean levels of guanosine, inosine, dGuo, and dIno were 4.4, 133.3, 3.6, and 3.8 μmol/L, respectively, in affected patients. No indeterminate or false-positive results were found. In patient 8 TREC levels were borderline and KREC levels were abnormal; in patient 9 TRECs were undetectable, whereas KREC levels were normal.TMS is a valid method for diagnosis of PNP deficiency on DBSs of affected patients at a negligible cost. TMS identifies newborns with PNP deficiency, whereas TREC or KREC measurement alone can fail.

    View details for DOI 10.1016/j.jaci.2014.01.040

    View details for Web of Science ID 000338930300020

    View details for PubMedID 24767876

  • Differential role of nonhomologous end joining factors in the generation, DNA damage response, and myeloid differentiation of human induced pluripotent stem cells PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Felgentreff, K., Du, L., Weinacht, K. G., Dobbs, K., Bartish, M., Giliani, S., Schlaeger, T., DeVine, A., Schambach, A., Woodbine, L. J., Davies, G., Baxi, S. N., van der Burg, M., Bleesing, J., Gennery, A., Manis, J., Pan-Hammarstrom, Q., Notarangelo, L. D. 2014; 111 (24): 8889-8894

    Abstract

    Nonhomologous end-joining (NHEJ) is a key pathway for efficient repair of DNA double-strand breaks (DSBs) and V(D)J recombination. NHEJ defects in humans cause immunodeficiency and increased cellular sensitivity to ionizing irradiation (IR) and are variably associated with growth retardation, microcephaly, and neurodevelopmental delay. Repair of DNA DSBs is important for reprogramming of somatic cells into induced pluripotent stem cells (iPSCs). To compare the specific contribution of DNA ligase 4 (LIG4), Artemis, and DNA-protein kinase catalytic subunit (PKcs) in this process and to gain insights into phenotypic variability associated with these disorders, we reprogrammed patient-derived fibroblast cell lines with NHEJ defects. Deficiencies of LIG4 and of DNA-PK catalytic activity, but not Artemis deficiency, were associated with markedly reduced reprogramming efficiency, which could be partially rescued by genetic complementation. Moreover, we identified increased genomic instability in LIG4-deficient iPSCs. Cell cycle synchronization revealed a severe defect of DNA repair and a G0/G1 cell cycle arrest, particularly in LIG4- and DNA-PK catalytically deficient iPSCs. Impaired myeloid differentiation was observed in LIG4-, but not Artemis- or DNA-PK-mutated iPSCs. These results indicate a critical importance of the NHEJ pathway for somatic cell reprogramming, with a major role for LIG4 and DNA-PKcs and a minor, if any, for Artemis.

    View details for DOI 10.1073/pnas.1323649111

    View details for Web of Science ID 000337300100048

    View details for PubMedID 24889605

  • Primary Immune Deficiency Treatment Consortium (PIDTC) report. journal of allergy and clinical immunology Griffith, L. M., Cowan, M. J., Notarangelo, L. D., Kohn, D. B., Puck, J. M., Pai, S., Ballard, B., Bauer, S. C., Bleesing, J. J., Boyle, M., Brower, A., Buckley, R. H., van der Burg, M., Burroughs, L. M., Candotti, F., Cant, A. J., Chatila, T., Cunningham-Rundles, C., Dinauer, M. C., Dvorak, C. C., Filipovich, A. H., Fleisher, T. A., Bobby Gaspar, H., Gungor, T., Haddad, E., Hovermale, E., Huang, F., Hurley, A., Hurley, M., Iyengar, S., Kang, E. M., Logan, B. R., Long-Boyle, J. R., Malech, H. L., McGhee, S. A., Modell, F., Modell, V., Ochs, H. D., O'Reilly, R. J., Parkman, R., Rawlings, D. J., Routes, J. M., Shearer, W. T., Small, T. N., Smith, H., Sullivan, K. E., Szabolcs, P., Thrasher, A., Torgerson, T. R., Veys, P., Weinberg, K., Zuniga-Pflucker, J. C. 2014; 133 (2): 335-347 e11

    Abstract

    The Primary Immune Deficiency Treatment Consortium (PIDTC) is a network of 33 centers in North America that study the treatment of rare and severe primary immunodeficiency diseases. Current protocols address the natural history of patients treated for severe combined immunodeficiency (SCID), Wiskott-Aldrich syndrome, and chronic granulomatous disease through retrospective, prospective, and cross-sectional studies. The PIDTC additionally seeks to encourage training of junior investigators, establish partnerships with European and other International colleagues, work with patient advocacy groups to promote community awareness, and conduct pilot demonstration projects. Future goals include the conduct of prospective treatment studies to determine optimal therapies for primary immunodeficiency diseases. To date, the PIDTC has funded 2 pilot projects: newborn screening for SCID in Navajo Native Americans and B-cell reconstitution in patients with SCID after hematopoietic stem cell transplantation. Ten junior investigators have received grant awards. The PIDTC Annual Scientific Workshop has brought together consortium members, outside speakers, patient advocacy groups, and young investigators and trainees to report progress of the protocols and discuss common interests and goals, including new scientific developments and future directions of clinical research. Here we report the progress of the PIDTC to date, highlights of the first 2 PIDTC workshops, and consideration of future consortium objectives.

    View details for DOI 10.1016/j.jaci.2013.07.052

    View details for PubMedID 24139498

  • Whole-exome sequencing identifies tetratricopeptide repeat domain 7A (TTC7A) mutations for combined immunodeficiency with intestinal atresias JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY Chen, R., Giliani, S., Lanzi, G., Mias, G. I., Lonardi, S., Dobbs, K., Manis, J., Im, H., Gallagher, J. E., Phanstiel, D. H., Euskirchen, G., Lacroute, P., Bettinger, K., Moratto, D., Weinacht, K., Montin, D., Gallo, E., Mangili, G., Porta, F., Notarangelo, L. D., Pedretti, S., Al-Herz, W., Alfahdli, W., Comeau, A. M., Traister, R. S., Pai, S., Carella, G., Facchetti, F., Nadeau, K. C., Snyder, M., Notarangelo, L. D. 2013; 132 (3): 656-?

    Abstract

    Combined immunodeficiency with multiple intestinal atresias (CID-MIA) is a rare hereditary disease characterized by intestinal obstructions and profound immune defects.We sought to determine the underlying genetic causes of CID-MIA by analyzing the exomic sequences of 5 patients and their healthy direct relatives from 5 unrelated families.We performed whole-exome sequencing on 5 patients with CID-MIA and 10 healthy direct family members belonging to 5 unrelated families with CID-MIA. We also performed targeted Sanger sequencing for the candidate gene tetratricopeptide repeat domain 7A (TTC7A) on 3 additional patients with CID-MIA.Through analysis and comparison of the exomic sequence of the subjects from these 5 families, we identified biallelic damaging mutations in the TTC7A gene, for a total of 7 distinct mutations. Targeted TTC7A gene sequencing in 3 additional unrelated patients with CID-MIA revealed biallelic deleterious mutations in 2 of them, as well as an aberrant splice product in the third patient. Staining of normal thymus showed that the TTC7A protein is expressed in thymic epithelial cells, as well as in thymocytes. Moreover, severe lymphoid depletion was observed in the thymus and peripheral lymphoid tissues from 2 patients with CID-MIA.We identified deleterious mutations of the TTC7A gene in 8 unrelated patients with CID-MIA and demonstrated that the TTC7A protein is expressed in the thymus. Our results strongly suggest that TTC7A gene defects cause CID-MIA.

    View details for DOI 10.1016/j.jaci.2013.06.013

    View details for Web of Science ID 000323612000018

    View details for PubMedID 23830146

  • Whole-exome sequencing identifies tetratricopeptide repeat domain 7A (TTC7A) mutations for combined immunodeficiency with intestinal atresias. journal of allergy and clinical immunology Chen, R., Giliani, S., Lanzi, G., Mias, G. I., Lonardi, S., Dobbs, K., Manis, J., Im, H., Gallagher, J. E., Phanstiel, D. H., Euskirchen, G., Lacroute, P., Bettinger, K., Moratto, D., Weinacht, K., Montin, D., Gallo, E., Mangili, G., Porta, F., Notarangelo, L. D., Pedretti, S., Al-Herz, W., Alfahdli, W., Comeau, A. M., Traister, R. S., Pai, S., Carella, G., Facchetti, F., Nadeau, K. C., Snyder, M., Notarangelo, L. D. 2013; 132 (3): 656-664 e17

    Abstract

    Combined immunodeficiency with multiple intestinal atresias (CID-MIA) is a rare hereditary disease characterized by intestinal obstructions and profound immune defects.We sought to determine the underlying genetic causes of CID-MIA by analyzing the exomic sequences of 5 patients and their healthy direct relatives from 5 unrelated families.We performed whole-exome sequencing on 5 patients with CID-MIA and 10 healthy direct family members belonging to 5 unrelated families with CID-MIA. We also performed targeted Sanger sequencing for the candidate gene tetratricopeptide repeat domain 7A (TTC7A) on 3 additional patients with CID-MIA.Through analysis and comparison of the exomic sequence of the subjects from these 5 families, we identified biallelic damaging mutations in the TTC7A gene, for a total of 7 distinct mutations. Targeted TTC7A gene sequencing in 3 additional unrelated patients with CID-MIA revealed biallelic deleterious mutations in 2 of them, as well as an aberrant splice product in the third patient. Staining of normal thymus showed that the TTC7A protein is expressed in thymic epithelial cells, as well as in thymocytes. Moreover, severe lymphoid depletion was observed in the thymus and peripheral lymphoid tissues from 2 patients with CID-MIA.We identified deleterious mutations of the TTC7A gene in 8 unrelated patients with CID-MIA and demonstrated that the TTC7A protein is expressed in the thymus. Our results strongly suggest that TTC7A gene defects cause CID-MIA.

    View details for DOI 10.1016/j.jaci.2013.06.013

    View details for PubMedID 23830146

  • First reported case of Omenn syndrome in a patient with reticular dysgenesis JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY Henderson, L. A., Frugoni, F., Hopkins, G., Al-Herz, W., Weinacht, K., Comeau, A. M., Bonilla, F. A., Notarangelo, L. D., Pai, S. 2013; 131 (4): 1227-1230

    View details for DOI 10.1016/j.jaci.2012.07.045

    View details for Web of Science ID 000317187200034

    View details for PubMedID 23014587

  • The role of induced pluripotent stem cells in research and therapy of primary immunodeficiencies. Current opinion in immunology Weinacht, K. G., Brauer, P. M., Felgentreff, K., Devine, A., Gennery, A. R., Giliani, S., Al-Herz, W., Schambach, A., Zúñiga-Pflücker, J. C., Notarangelo, L. D. 2012; 24 (5): 617-624

    Abstract

    The advent of reprogramming technology has greatly advanced the field of stem cell biology and nurtured our hope to create patient specific renewable stem cell sources. While the number of reports of disease specific induced pluripotent stem cells is continuously rising, the field becomes increasingly more aware that induced pluripotent stem cells are not as similar to embryonic stem cells as initially assumed. Our state of the art understanding of human induced pluripotent stem cells, their capacity, their limitations and their promise as it pertains to the study and treatment of primary immunodeficiencies, is the content of this review.

    View details for DOI 10.1016/j.coi.2012.07.001

    View details for PubMedID 22841347

  • Trans locus inhibitors limit concomitant polysaccharide synthesis in the human gut symbiont Bacteroides fragilis PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Chatzidaki-Livanis, M., Weinacht, K. G., Comstock, L. E. 2010; 107 (26): 11976-11980

    Abstract

    Bacteroides is an abundant genus of bacteria of the human intestinal microbiota. Bacteroides species synthesize a large number of capsular polysaccharides (PS), a biological property not shared with closely related oral species, suggesting importance for intestinal survival. Bacteroides fragilis, for example, synthesizes eight capsular polysaccharides per strain, each of which phase varies via inversion of the promoters located upstream of seven of the eight polysaccharide biosynthesis operons. In a single cell, many of these polysaccharide loci promoters can be simultaneously oriented on for transcription of the downstream biosynthesis operons. Here, we demonstrate that despite the promoter orientations, concomitant transcription of multiple polysaccharide loci within a cell is inhibited. The proteins encoded by the second gene of each of these eight loci, collectively designated the UpxZ proteins, inhibit the synthesis of heterologous polysaccharides. These unique proteins interfere with the ability of UpxY proteins encoded by other polysaccharide loci to function in transcriptional antitermination of their respective operon. The eight UpxZs have different inhibitory spectra, thus establishing a hierarchical regulatory network for polysaccharide synthesis. Limitation of concurrent polysaccharide synthesis strongly suggests that these bacteria evolved this property as an evasion-type mechanism to avoid killing by polysaccharide-targeting factors in the ecosystem.

    View details for DOI 10.1073/pnas.1005039107

    View details for Web of Science ID 000279332300058

    View details for PubMedID 20547868

  • Tyrosine site-specific recombinases mediate DNA inversions affecting the expression of outer surface proteins of Bacteroides fragilis MOLECULAR MICROBIOLOGY Weinacht, K. G., Roche, H., Krinos, C. M., Coyne, M. J., Parkhill, J., Comstock, L. E. 2004; 53 (5): 1319-1330

    Abstract

    The chromosome of Bacteroides fragilis has been shown to undergo 13 distinct DNA inversions affecting the expression of capsular polysaccharides and mediated by a serine site-specific recombinase designated Mpi. In this study, we demonstrate that members of the tyrosine site-specific recombinase family, conserved in B. fragilis, mediate additional DNA inversions of the B. fragilis genome. These DNA invertases flip promoter regions in their immediate downstream region. The genetic organization of the genes regulated by these invertible promoter regions suggests that they are operons and many of the products are predicted to be outer membrane proteins. Phenotypic analysis of a deletion mutant of one of these DNA invertases, tsr15 (aapI), which resulted in the promoter region for the downstream genes being locked ON, confirmed the synthesis of multiple surface proteins by this operon. In addition, this deletion mutant demonstrated an autoaggregative phenotype and showed significantly greater adherence than wild-type organisms in a biofilm assay, suggesting a possible functional role for these phase-variable outer surface proteins. This study demonstrates that DNA inversion is a universal mechanism used by this commensal microorganism to phase vary expression of its surface molecules and involves at least three conserved DNA invertases from two evolutionarily distinct families.

    View details for DOI 10.1111/j.1365-2958.2004.04219.x

    View details for Web of Science ID 000223495100004

    View details for PubMedID 15387812

  • Mpi recombinase globally modulates the surface architecture of a human commensal bacterium PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Coyne, M. J., Weinacht, K. G., Krinos, C. M., Comstock, L. E. 2003; 100 (18): 10446-10451

    Abstract

    The mammalian gut represents a complex and diverse ecosystem, consisting of unique interactions between the host and microbial residents. Bacterial surfaces serve as an interface that promotes and responds to this dynamic exchange, a process essential to the biology of both symbionts. The human intestinal microorganism, Bacteroides fragilis, is able to extensively modulate its surface. Analysis of the B. fragilis genomic sequence, together with genetic conservation analyses, cross-species cloning experiments, and mutational studies, revealed that this organism utilizes an endogenous DNA inversion factor to globally modulate the expression of its surface structures. This DNA invertase is necessary for the inversion of at least 13 regions located throughout the genome, including the promoter regions for seven of the capsular polysaccharide biosynthesis loci, an accessory polysaccharide biosynthesis locus, and five other regions containing consensus promoter sequences. Bacterial DNA invertases of the serine site-specific recombinase family are typically encoded by imported elements such as phage and plasmids, and act locally on a single region of the imported element. In contrast, the conservation and unique global regulatory nature of the process in B. fragilis suggest an evolutionarily ancient mechanism for surface adaptation to the changing intestinal milieu during commensalism.

    View details for DOI 10.1073/pnas.1832655100

    View details for Web of Science ID 000185119300057

    View details for PubMedID 12915735

  • Extensive surface diversity of a commensal microorganism by multiple DNA inversions NATURE Krinos, C. M., Coyne, M. J., Weinacht, K. G., Tzianabos, A. O., Kasper, D. L., Comstock, L. E. 2001; 414 (6863): 555-558

    Abstract

    The dynamic interactions between a host and its intestinal microflora that lead to commensalism are unclear. Bacteria that colonize the intestinal tract do so despite the development of a specific immune response by the host. The mechanisms used by commensal organisms to circumvent this immune response have yet to be established. Here we demonstrate that the human colonic microorganism, Bacteroides fragilis, is able to modulate its surface antigenicity by producing at least eight distinct capsular polysaccharides-a number greater than any previously reported for a bacterium-and is able to regulate their expression in an on-off manner by the reversible inversion of DNA segments containing the promoters for their expression. This means of generating surface diversity allows the organism to exhibit a wide array of distinct surface polysaccharide combinations, and may have broad implications for how the predominant human colonic microorganisms, the Bacteroides species, maintain an ecological niche in the intestinal tract.

    View details for Web of Science ID 000172405900050

    View details for PubMedID 11734857

  • [Drug hypersensitivity]. Terapevticheskii? arkhiv TAREEV, E. M., VINOGRADOVA, O. M., Semenkova, E. N., SOLOV'EVA, A. P. 1975; 47 (4): 5-12

    View details for PubMedID 238301