Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hanna IJspeert is active.

Publication


Featured researches published by Hanna IJspeert.


Journal of Clinical Investigation | 2008

A DNA-PKcs mutation in a radiosensitive T-B- SCID patient inhibits Artemis activation and nonhomologous end-joining

Mirjam van der Burg; Hanna IJspeert; Nicole S. Verkaik; Tuba Turul; Wouter W. Wiegant; Keiko Morotomi-Yano; Pierre Olivier Mari; Ilhan Tezcan; David J. Chen; Małgorzata Z. Zdzienicka; Jacques J.M. van Dongen; Dik C. van Gent

Radiosensitive T-B- severe combined immunodeficiency (RS-SCID) is caused by defects in the nonhomologous end-joining (NHEJ) DNA repair pathway, which results in failure of functional V(D)J recombination. Here we have identified the first human RS-SCID patient to our knowledge with a DNA-PKcs missense mutation (L3062R). The causative mutation did not affect the kinase activity or DNA end-binding capacity of DNA-PKcs itself; rather, the presence of long P-nucleotide stretches in the immunoglobulin coding joints indicated that it caused insufficient Artemis activation, something that is dependent on Artemis interaction with autophosphorylated DNA-PKcs. Moreover, overall end-joining activity was hampered, suggesting that Artemis-independent DNA-PKcs functions were also inhibited. This study demonstrates that the presence of DNA-PKcs kinase activity is not sufficient to rule out a defect in this gene during diagnosis and treatment of RS-SCID patients. Further, the data suggest that residual DNA-PKcs activity is indispensable in humans.


Blood | 2012

Ribosomal deficiencies in Diamond-Blackfan anemia impair translation of transcripts essential for differentiation of murine and human erythroblasts

Rastislav Horos; Hanna IJspeert; Dagmar Pospisilova; Regine Sendtner; Charlotte Andrieu-Soler; Erdogan Taskesen; Andrzej Nieradka; Radek Cmejla; Michael Sendtner; Ivo P. Touw; Marieke von Lindern

Diamond-Blackfan anemia (DBA) is associated with developmental defects and profound anemia. Mutations in genes encoding a ribosomal protein of the small (e.g., RPS19) or large (e.g., RPL11) ribosomal subunit are found in more than half of these patients. The mutations cause ribosomal haploinsufficiency, which reduces overall translation efficiency of cellular mRNAs. We reduced the expression of Rps19 or Rpl11 in mouse erythroblasts and investigated mRNA polyribosome association, which revealed deregulated translation initiation of specific transcripts. Among these were Bag1, encoding a Hsp70 cochaperone, and Csde1, encoding an RNA-binding protein, and both were expressed at increased levels in erythroblasts. Their translation initiation is cap independent and starts from an internal ribosomal entry site, which appeared sensitive to knockdown of Rps19 or Rpl11. Mouse embryos lacking Bag1 die at embryonic day 13.5, with reduced erythroid colony forming cells in the fetal liver, and low Bag1 expression impairs erythroid differentiation in vitro. Reduced expression of Csde1 impairs the proliferation and differentiation of erythroid blasts. Protein but not mRNA expression of BAG1 and CSDE1 was reduced in erythroblasts cultured from DBA patients. Our data suggest that impaired internal ribosomal entry site-mediated translation of mRNAs expressed at increased levels in erythroblasts contributes to the erythroid phenotype of DBA.


Blood | 2011

Idiopathic CD4+ T lymphopenia without autoimmunity or granulomatous disease in the slipstream of RAG mutations

Taco W. Kuijpers; Hanna IJspeert; Ester M. M. van Leeuwen; Machiel H. Jansen; Mette D. Hazenberg; Kees Weijer; René A. W. van Lier; Mirjam van der Burg

A girl presented during childhood with a single course of extensive chickenpox and moderate albeit recurrent pneumonia in the presence of idiopathic CD4+ T lymphocytopenia (ICL). Her clinical condition remained stable over the past 10 years without infections, any granulomatous disease, or autoimmunity. Immunophenotyping demonstrated strongly reduced naive T and B cells with intact proliferative capacity. Antibody reactivity on in vivo immunizations was normal. T-cell receptor-Vβ repertoire was polyclonal with a very low content of T-cell receptor excision circles (TRECs). Kappa-deleting recombination excision circles (KRECs) were also abnormal in the B cells. Both reflect extensive in vivo proliferation. Patient-derived CD34+ hematopoietic stem cells could not repopulate RAG2(-/-)IL2Rγc(-/-) mice, indicating the lymphoid origin of the defect. We identified 2 novel missense mutations in RAG1 (p.Arg474Cys and p.Leu506Phe) resulting in reduced RAG activity. This report gives the first genetic clue for ICL and extends the clinical spectrum of RAG mutations from severe immune defects to an almost normal condition.


American Journal of Human Genetics | 2015

Mutations in the NHEJ Component XRCC4 Cause Primordial Dwarfism

Jennie E. Murray; Mirjam van der Burg; Hanna IJspeert; Paula Carroll; Qian Wu; Takashi Ochi; Andrea Leitch; Edward S. Miller; Boris Kysela; Alireza Jawad; Armand Bottani; Francesco Brancati; Marco Cappa; Valérie Cormier-Daire; Charu Deshpande; Eissa Faqeih; Gail E. Graham; Emmanuelle Ranza; Tom L. Blundell; Andrew P. Jackson; Grant S. Stewart; Louise S. Bicknell

Non-homologous end joining (NHEJ) is a key cellular process ensuring genome integrity. Mutations in several components of the NHEJ pathway have been identified, often associated with severe combined immunodeficiency (SCID), consistent with the requirement for NHEJ during V(D)J recombination to ensure diversity of the adaptive immune system. In contrast, we have recently found that biallelic mutations in LIG4 are a common cause of microcephalic primordial dwarfism (MPD), a phenotype characterized by prenatal-onset extreme global growth failure. Here we provide definitive molecular genetic evidence supported by biochemical, cellular, and immunological data for mutations in XRCC4, encoding the obligate binding partner of LIG4, causing MPD. We report the identification of biallelic mutations in XRCC4 in five families. Biochemical and cellular studies demonstrate that these alterations substantially decrease XRCC4 protein levels leading to reduced cellular ligase IV activity. Consequently, NHEJ-dependent repair of ionizing-radiation-induced DNA double-strand breaks is compromised in XRCC4 cells. Similarly, immunoglobulin junctional diversification is impaired in cells. However, immunoglobulin levels are normal, and individuals lack overt signs of immunodeficiency. Additionally, in contrast to individuals with LIG4 mutations, pancytopenia leading to bone marrow failure has not been observed. Hence, alterations that alter different NHEJ proteins give rise to a phenotypic spectrum, from SCID to extreme growth failure, with deficiencies in certain key components of this repair pathway predominantly exhibiting growth deficits, reflecting differential developmental requirements for NHEJ proteins to support growth and immune maturation.


Blood | 2016

XLF deficiency results in reduced N-nucleotide addition during V(D)J recombination

Hanna IJspeert; Jacob Rozmus; Klaus Schwarz; René L. Warren; David van Zessen; Robert A. Holt; Ingrid Pico-Knijnenburg; Erik J. Simons; Isabel Jerchel; Angela Wawer; Myriam Ricarda Lorenz; Turkan Patiroglu; Himmet Haluk Akar; Ricardo Leite; Nicole S. Verkaik; Andrew Stubbs; Dik C. van Gent; Jacques J.M. van Dongen; Mirjam van der Burg

Repair of DNA double-strand breaks (DSBs) by the nonhomologous end-joining pathway (NHEJ) is important not only for repair of spontaneous breaks but also for breaks induced in developing lymphocytes during V(D)J (variable [V], diversity [D], and joining [J] genes) recombination of their antigen receptor loci to create a diverse repertoire. Mutations in the NHEJ factor XLF result in extreme sensitivity for ionizing radiation, microcephaly, and growth retardation comparable to mutations in LIG4 and XRCC4, which together form the NHEJ ligation complex. However, the effect on the immune system is variable (mild to severe immunodeficiency) and less prominent than that seen in deficiencies of NHEJ factors ARTEMIS and DNA-dependent protein kinase catalytic subunit, with defects in the hairpin opening step, which is crucial and unique for V(D)J recombination. Therefore, we aimed to study the role of XLF during V(D)J recombination. We obtained clinical data from 9 XLF-deficient patients and performed immune phenotyping and antigen receptor repertoire analysis of immunoglobulin (Ig) and T-cell receptor (TR) rearrangements, using next-generation sequencing in 6 patients. The results were compared with XRCC4 and LIG4 deficiency. Both Ig and TR rearrangements showed a significant decrease in the number of nontemplated (N) nucleotides inserted by terminal deoxynucleotidyl transferase, which resulted in a decrease of 2 to 3 amino acids in the CDR3. Such a reduction in the number of N-nucleotides has a great effect on the junctional diversity, and thereby on the total diversity of the Ig and TR repertoire. This shows that XLF has an important role during V(D)J recombination in creating diversity of the repertoire by stimulating N-nucleotide insertion.


The Journal of Allergy and Clinical Immunology | 2013

Antibody deficiency in patients with ataxia telangiectasia is caused by disturbed B- and T-cell homeostasis and reduced immune repertoire diversity

Gertjan J. Driessen; Hanna IJspeert; Corry M. R. Weemaes; Ásgeir Haraldsson; Margreet Trip; Adilia Warris; Michiel van der Flier; Nico Wulffraat; M.M.M. Verhagen; Malcolm Taylor; Menno C. van Zelm; Jacques J.M. van Dongen; Marcel van Deuren; Mirjam van der Burg

BACKGROUND Ataxia telangiectasia (AT) is a multisystem DNA-repair disorder caused by mutations in the ataxia telangiectasia mutated (ATM) gene. Patients with AT have reduced B- and T-cell numbers and a highly variable immunodeficiency. ATM is important for V(D)J recombination and immunoglobulin class-switch recombination (CSR); however, little is known about the mechanisms resulting in antibody deficiency severity. OBJECTIVE We sought to examine the immunologic mechanisms responsible for antibody deficiency heterogeneity in patients with AT. METHODS In this study we included patients with classical AT plus early-onset hypogammaglobulinemia (n = 3), classical AT (n = 8), and variant AT (late onset, n = 4). We studied peripheral B- and T-cell subsets, B-cell subset replication history, somatic hypermutation frequencies, CSR patterns, B-cell repertoire, and ATM kinase activity. RESULTS Patients with classical AT lacked ATM kinase activity, whereas patients with variant AT showed residual function. Most patients had disturbed naive B-cell and T-cell homeostasis, as evidenced by low cell numbers, increased proliferation, a large proportion CD21(low)CD38(low) anergic B cells, and decreased antigen receptor repertoire diversity. Impaired formation of T cell-dependent memory B cells was predominantly found in patients with AT plus hypogammaglobulinemia. These patients had extremely low naive CD4(+) T-cell counts, which were more severely reduced compared with those seen in patients with classical AT without hypogammaglobulinemia. Finally, AT deficiency resulted in defective CSR to distal constant regions that might reflect an impaired ability of B cells to undergo multiple germinal center reactions. CONCLUSION The severity of the antibody deficiency in patients with AT correlates with disturbances in B- and T-cell homeostasis resulting in reduced immune repertoire diversity, which consequently affects the chance of successful antigen-dependent cognate B-T interaction.


Human Mutation | 2013

Clinical Spectrum of LIG4 Deficiency Is Broadened with Severe Dysmaturity, Primordial Dwarfism, and Neurological Abnormalities.

Hanna IJspeert; Adilia Warris; Michiel van der Flier; Ismail Reisli; Sevgi Keles; Sandra Chishimba; Jacques J.M. van Dongen; Dik C. van Gent; Mirjam van der Burg

DNA double‐strand break repair via non‐homologous end joining (NHEJ) is involved in recombination of immunoglobulin and T‐cell receptor genes. Mutations in NHEJ components result in syndromes that are characterized by microcephaly and immunodeficiency. We present a patient with lymphopenia, extreme radiosensitivity, severe dysmaturity, corpus callosum agenesis, polysyndactily, dysmorphic appearance, and erythema, which are suggestive of a new type of NHEJ deficiency. We identified two heterozygous mutations in LIG4. The p.S205LfsX29 mutation results in lack of the nuclear localization signal and appears to be a null mutation. The second mutation p.K635RfsX10 lacks the C‐terminal region responsible for XRCC4 binding and LIG4 stability and activity, and therefore this mutant might be a null mutation as well or have very low residual activity. This is remarkable since Lig4 knockout mice are embryonic lethal and so far in humans no complete LIG4 deficiencies have been described. This case broadens the clinical spectrum of LIG4 deficiencies.


Frontiers in Immunology | 2016

Evaluation of the Antigen-Experienced B-Cell Receptor Repertoire in Healthy Children and Adults

Hanna IJspeert; Pauline A. van Schouwenburg; David van Zessen; Ingrid Pico-Knijnenburg; Gertjan J. Driessen; Andrew Stubbs; Mirjam van der Burg

Upon antigen recognition via their B cell receptor (BR), B cells migrate to the germinal center where they undergo somatic hypermutation (SHM) to increase their affinity for the antigen, and class switch recombination (CSR) to change the effector function of the secreted antibodies. These steps are essential to create an antigen-experienced BR repertoire that efficiently protects the body against pathogens. At the same time, the BR repertoire should be selected to protect against responses to self-antigen or harmless antigens. Insights into the processes of SHM, selection, and CSR can be obtained by studying the antigen-experienced BR repertoire. Currently, a large reference data set of healthy children and adults, which ranges from neonates to the elderly, is not available. In this study, we analyzed the antigen-experienced repertoire of 38 healthy donors (HD), ranging from cord blood to 74 years old, by sequencing IGA and IGG transcripts using next generation sequencing. This resulted in a large, freely available reference data set containing 412,890 IGA and IGG transcripts. We used this data set to study mutation levels, SHM patterns, antigenic selection, and CSR from birth to elderly HD. Only small differences were observed in SHM patterns, while the mutation levels increase in early childhood and stabilize at 6 years of age at around 7%. Furthermore, comparison of the antigen-experienced repertoire with sequences from the naive immune repertoire showed that features associated with autoimmunity such as long CDR3 length and IGHV4-34 usage are reduced in the antigen-experienced repertoire. Moreover, IGA2 and IGG2 usage was increased in HD in higher age categories, while IGG1 usage was decreased. In addition, we studied clonal relationship in the different samples. Clonally related sequences were found with different subclasses. Interestingly, we found transcripts with the same CDR1–CDR3 sequence, but different subclasses. Together, these data suggest that a single antigen can provoke a B-cell response with BR of different subclasses and that, during the course of an immune response, some B cells change their isotype without acquiring additional SHM or can directly switch to different isotypes.


The Journal of Allergy and Clinical Immunology | 2016

Increased PI3K/Akt activity and deregulated humoral immune response in human PTEN deficiency.

Gertjan J. Driessen; Hanna IJspeert; Marjolein Wentink; Helger G. Yntema; P. Martin van Hagen; Arthur van Strien; Giorgia Bucciol; Ozgur Cogulu; Margreet Trip; Willy M. Nillesen; Els Peeters; Ingrid Pico-Knijnenburg; Barbara H. Barendregt; Marta Rizzi; Jacques J.M. van Dongen; Necil Kutukculer; Mirjam van der Burg

_To the Editor:_ Autosomal-dominant germline mutations in _PTEN_ are associated with phosphate and tensin homologue deleted on chromosome 10 (PTEN) hamartoma tumor syndromes (PHTS), including Cowden syndrome, characterized by hamartomas, malignant tumors, macrocephaly, and neurodevelopmental delay. Immunodeficiency has recently been reported in PHTS, but the mechanism of disease is not clear. Therefore, we performed a detailed study of the peripheral B-cell development of 9 patients with PHTS, to explore the role of phosphatidyl inositol 3-kinase (PI3K)/Akt signaling in the humoral immune response in these patients (details are described in this article’s Methods section in the Online Repository at _www.jacionline.org_).


Blood | 2016

Disturbed B-lymphocyte selection in autoimmune lymphoproliferative syndrome

Ales Janda; Klaus Schwarz; Mirjam van der Burg; Werner Vach; Hanna IJspeert; Myriam Ricarda Lorenz; Magdeldin Elgizouli; Kathrin Pieper; Paul Fisch; Joachim Hagel; Raquel Lorenzetti; Maximilian Seidl; Joachim Roesler; Fabian Hauck; Elisabetta Traggiai; Carsten Speckmann; Anne Rensing-Ehl; Stephan Ehl; Hermann Eibel; Marta Rizzi

Fas is a transmembrane receptor involved in the maintenance of tolerance and immune homeostasis. In murine models, it has been shown to be essential for deletion of autoreactive B cells in the germinal center. The role of Fas in human B-cell selection and in development of autoimmunity in patients carrying FAS mutations is unclear. We analyzed patients with either a somatic FAS mutation or a germline FAS mutation and somatic loss-of-heterozygosity, which allows comparing the fate of B cells with impaired vs normal Fas signaling within the same individual. Class-switched memory B cells showed: accumulation of FAS-mutated B cells; failure to enrich single V, D, J genes and single V-D, D-J gene combinations of the B-cell receptor variable region; increased frequency of variable regions with higher content of positively charged amino acids; and longer CDR3 and maintenance of polyreactive specificities. Importantly, Fas-deficient switched memory B cells showed increased rates of somatic hypermutation. Our data uncover a defect in B-cell selection in patients with FAS mutations, which has implications for the understanding of the pathogenesis of autoimmunity and lymphomagenesis of autoimmune lymphoproliferative syndrome.

Collaboration


Dive into the Hanna IJspeert's collaboration.

Top Co-Authors

Avatar

Mirjam van der Burg

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dik C. van Gent

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Andrew Stubbs

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Gertjan J. Driessen

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anton W. Langerak

Erasmus University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arjan C. Lankester

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

David van Zessen

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Erik J. Simons

Erasmus University Rotterdam

View shared research outputs
Researchain Logo
Decentralizing Knowledge