Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
1.
Nucleic Acids Res ; 51(12): 6337-6354, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37224534

ABSTRACT

Accurate genome replication is essential for all life and a key mechanism of disease prevention, underpinned by the ability of cells to respond to replicative stress (RS) and protect replication forks. These responses rely on the formation of Replication Protein A (RPA)-single stranded (ss) DNA complexes, yet this process remains largely uncharacterized. Here, we establish that actin nucleation-promoting factors (NPFs) associate with replication forks, promote efficient DNA replication and facilitate association of RPA with ssDNA at sites of RS. Accordingly, their loss leads to deprotection of ssDNA at perturbed forks, impaired ATR activation, global replication defects and fork collapse. Supplying an excess of RPA restores RPA foci formation and fork protection, suggesting a chaperoning role for actin nucleators (ANs) (i.e. Arp2/3, DIAPH1) and NPFs (i.e, WASp, N-WASp) in regulating RPA availability upon RS. We also discover that ß-actin interacts with RPA directly in vitro, and in vivo a hyper-depolymerizing ß-actin mutant displays a heightened association with RPA and the same dysfunctional replication phenotypes as loss of ANs/NPFs, which contrasts with the phenotype of a hyper-polymerizing ß-actin mutant. Thus, we identify components of actin polymerization pathways that are essential for preventing ectopic nucleolytic degradation of perturbed forks by modulating RPA activity.


Subject(s)
Actins , DNA Replication , Actins/genetics , Replication Protein A/genetics , Replication Protein A/metabolism , DNA, Single-Stranded/genetics , Molecular Chaperones/genetics
2.
bioRxiv ; 2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36711944

ABSTRACT

Accurate genome replication is essential for all life and a key mechanism of disease prevention, underpinned by the ability of cells to respond to replicative stress (RS) and protect replication forks. These responses rely on the formation of Replication Protein A (RPA)-single stranded (ss) DNA complexes, yet this process remains largely uncharacterized. Here we establish that actin nucleation-promoting factors (NPFs) associate with replication forks, promote efficient DNA replication and facilitate association of RPA with ssDNA at sites of RS. Accordingly, their loss leads to deprotection of ssDNA at perturbed forks, impaired ATR activation, global replication defects and fork collapse. Supplying an excess of RPA restores RPA foci formation and fork protection, suggesting a chaperoning role for actin nucleators (ANs) (i.e., Arp2/3, DIAPH1) and NPFs (i.e, WASp, N-WASp) in regulating RPA availability upon RS. We also discover that ß-actin interacts with RPA directly in vitro , and in vivo a hyper-depolymerizing ß-actin mutant displays a heightened association with RPA and the same dysfunctional replication phenotypes as loss of ANs/NPFs, which contrasts with the phenotype of a hyper-polymerizing ß-actin mutant. Thus, we identify components of actin polymerization pathways that are essential for preventing ectopic nucleolytic degradation of perturbed forks by modulating RPA activity.

3.
Nat Commun ; 13(1): 3743, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35768435

ABSTRACT

Perturbation in the replication-stress response (RSR) and DNA-damage response (DDR) causes genomic instability. Genomic instability occurs in Wiskott-Aldrich syndrome (WAS), a primary immunodeficiency disorder, yet the mechanism remains largely uncharacterized. Replication protein A (RPA), a single-strand DNA (ssDNA) binding protein, has key roles in the RSR and DDR. Here we show that human WAS-protein (WASp) modulates RPA functions at perturbed replication forks (RFs). Following genotoxic insult, WASp accumulates at RFs, associates with RPA, and promotes RPA:ssDNA complexation. WASp deficiency in human lymphocytes destabilizes RPA:ssDNA-complexes, impairs accumulation of RPA, ATR, ETAA1, and TOPBP1 at genotoxin-perturbed RFs, decreases CHK1 activation, and provokes global RF dysfunction. las17 (yeast WAS-homolog)-deficient S. cerevisiae also show decreased ScRPA accumulation at perturbed RFs, impaired DNA recombination, and increased frequency of DNA double-strand break (DSB)-induced single-strand annealing (SSA). Consequently, WASp (or Las17)-deficient cells show increased frequency of DSBs upon genotoxic insult. Our study reveals an evolutionarily conserved, essential role of WASp in the DNA stress-resolution pathway, such that WASp deficiency provokes RPA dysfunction-coupled genomic instability.


Subject(s)
DNA Breaks, Double-Stranded , DNA Replication , DNA, Single-Stranded , Replication Protein A , Saccharomyces cerevisiae Proteins , Wiskott-Aldrich Syndrome Protein , Animals , Antigens, Surface/metabolism , DNA Repair , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Instability , Humans , Protein Binding , Replication Protein A/genetics , Replication Protein A/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism
4.
Oncogene ; 40(2): 345-354, 2021 01.
Article in English | MEDLINE | ID: mdl-33139832

ABSTRACT

Immunodeficiency is associated with cancer risk. Accordingly, hematolymphoid cancers develop in Wiskott-Aldrich syndrome (WAS), an X-linked primary immunodeficiency disorder (PID) resulting from the deficiency of WAS-protein (WASp) expressed predominantly in the hematolymphoid cell lineages. Despite the correlation between WASp deficiency and hematolymphoid cancers, the molecular mechanism underlying the oncogenic role of WASp is incompletely understood. Employing the WASp-sufficient and WASp-deficient cell-pair model of human T and B lymphocytes, we show that WASp deficiency differentially influences hyperactivation versus inhibition of both CDC42:ERK1/2 and NF-κB:AP-1 pro-oncogenic signaling pathways in nonmalignant versus malignant T and B lymphocytes. Furthermore, WASp deficiency induces a cell-type specific up/down-modulation of the DNA-binding activities of NF-κB, AP-1, and multiple other transcription factors with known roles in oncogenesis. We propose that WASp functions as a putative "tumor-suppressor" protein in normal T and B cells, and "oncoprotein" in a subset of established T and B cell malignancies that are not associated with the NPM-ALK fusion.


Subject(s)
B-Lymphocytes/pathology , Oncogene Proteins/metabolism , T-Lymphocytes/pathology , Tumor Suppressor Proteins/metabolism , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription Factor AP-1/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/metabolism , cdc42 GTP-Binding Protein/metabolism
5.
J Allergy Clin Immunol ; 145(1): 324-334, 2020 01.
Article in English | MEDLINE | ID: mdl-31604087

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is an X-linked primary immune deficiency disorder resulting from Wiskott-Aldrich syndrome protein (WASp) deficiency. Lymphocytes from patients with WAS manifest increased DNA damage and lymphopenia from cell death, yet how WASp influences DNA damage-linked cell survival is unknown. A recently described mechanism promoting cell survival after ionizing radiation (IR)-induced DNA damage involves fragmentation and dispersal of the Golgi apparatus, known as the Golgi-dispersal response (GDR), which uses the Golgi phosphoprotein 3 (GOLPH3)-DNA-dependent protein kinase (DNA-PK)-myosin XVIIIA-F-actin signaling pathway. OBJECTIVE: We sought to define WASp's role in the DNA damage-induced GDR and its disruption as a contributor to the development of radiosensitivity-linked immunodeficiency in patients with WAS. METHODS: In human TH and B-cell culture systems, DNA damage-induced GDR elicited by IR or radiomimetic chemotherapy was monitored in the presence or absence of WASp or GOLPH3 alone or both together. RESULTS: WASp deficiency completely prevents the development of IR-induced GDR in human TH and B cells, despite the high DNA damage load. Loss of WASp impedes nuclear translocation of GOLPH3 and its colocalization with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Surprisingly, however, depletion of GOLPH3 alone or depolymerization of F-actin in WASp-sufficient TH cells still allows development of robust GDR, suggesting that WASp, but not GOLPH3, is essential for GDR and cell survival after IR-induced DNA-damage in human lymphocytes. CONCLUSION: The study identifies WASp as a novel effector of the nucleus-to-Golgi cell-survival pathway triggered by IR-induced DNA damage in cells of the hematolymphoid lineage and proposes an impaired GDR as a new cause for development of a "radiosensitive" form of immune dysregulation in patients with WAS.


Subject(s)
B-Lymphocytes/immunology , DNA Damage/immunology , Golgi Apparatus/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome Protein Family/immunology , DNA Damage/genetics , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/immunology , Golgi Apparatus/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein Family/genetics
6.
J Allergy Clin Immunol ; 142(1): 219-234, 2018 07.
Article in English | MEDLINE | ID: mdl-29248492

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS), X-linked thrombocytopenia (XLT), and X-linked neutropenia, which are caused by WAS mutations affecting Wiskott-Aldrich syndrome protein (WASp) expression or activity, manifest in immunodeficiency, autoimmunity, genomic instability, and lymphoid and other cancers. WASp supports filamentous actin formation in the cytoplasm and gene transcription in the nucleus. Although the genetic basis for XLT/WAS has been clarified, the relationships between mutant forms of WASp and the diverse features of these disorders remain ill-defined. OBJECTIVE: We sought to define how dysfunctional gene transcription is causally linked to the degree of TH cell deficiency and genomic instability in the XLT/WAS clinical spectrum. METHODS: In human TH1- or TH2-skewing cell culture systems, cotranscriptional R-loops (RNA/DNA duplex and displaced single-stranded DNA) and DNA double-strand breaks (DSBs) were monitored in multiple samples from patients with XLT and WAS and in normal T cells depleted of WASp. RESULTS: WASp deficiency provokes increased R-loops and R-loop-mediated DSBs in TH1 cells relative to TH2 cells. Mechanistically, chromatin occupancy of serine 2-unphosphorylated RNA polymerase II is increased, and that of topoisomerase 1, an R-loop preventing factor, is decreased at R-loop-enriched regions of IFNG and TBX21 (TH1 genes) in TH1 cells. These aberrations accompany increased unspliced (intron-retained) and decreased spliced mRNA of IFNG and TBX21 but not IL13 (TH2 gene). Significantly, increased cellular load of R-loops and DSBs, which are normalized on RNaseH1-mediated suppression of ectopic R-loops, inversely correlates with disease severity scores. CONCLUSION: Transcriptional R-loop imbalance is a novel molecular defect causative in TH1 immunodeficiency and genomic instability in patients with WAS. The study proposes that cellular R-loop load could be used as a potential biomarker for monitoring symptom severity and prognostic outcome in the XLT-WAS clinical spectrum and could be targeted therapeutically.


Subject(s)
Genomic Instability/genetics , Th1 Cells/pathology , Wiskott-Aldrich Syndrome/genetics , Cells, Cultured , DNA Damage/genetics , Humans , Transcription, Genetic , Wiskott-Aldrich Syndrome/pathology
7.
Indian J Med Microbiol ; 33(4): 588-90, 2015.
Article in English | MEDLINE | ID: mdl-26470972

ABSTRACT

Scedosporium prolificans have been reported to be resistant to all antifungals including the newer azoles and echinocandins. We report an unusual case of repeated S. prolificans infection of the heart valves in an immunocompetent patient.


Subject(s)
Antifungal Agents/therapeutic use , Endocarditis/diagnosis , Endocarditis/drug therapy , Mycoses/diagnosis , Mycoses/drug therapy , Scedosporium/isolation & purification , Humans , Male , Middle Aged , Recurrence
8.
Blood ; 126(14): 1670-82, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26261240

ABSTRACT

In Wiskott-Aldrich syndrome (WAS), immunodeficiency and autoimmunity often comanifest, yet how WAS mutations misregulate chromatin-signaling in Thelper (TH) cells favoring development of auto-inflammation over protective immunity is unclear. Previously, we identified an essential promoter-specific, coactivator role of nuclear-WASp in TH1 gene transcription. Here we identify small ubiquitin-related modifier (SUMO)ylation as a novel posttranslational modification of WASp, impairment of which converts nuclear-WASp from a transcriptional coactivator to a corepressor of nuclear factor (NF)-κB response genes in human (TH)1-differentiating cells. V75M, one of many disease-causing mutations occurring in SUMO*motif (72-ψψψψKDxxxxSY-83) of WASp, compromises WASp-SUMOylation, associates with COMMD1 to attenuate NF-κB signaling, and recruits histone deacetylases-6 (HDAC6) to p300-marked promoters of NF-κB response genes that pattern immunity but not inflammation. Consequently, proteins mediating adaptive immunity (IFNG, STAT1, TLR1) are deficient, whereas those mediating auto-inflammation (GM-CSF, TNFAIP2, IL-1ß) are paradoxically increased in TH1 cells expressing SUMOylation-deficient WASp. Moreover, SUMOylation-deficient WASp favors ectopic development of the TH17-like phenotype (↑IL17A, IL21, IL22, IL23R, RORC, and CSF2) under TH1-skewing conditions, suggesting a role for WASp in modulating TH1/TH17 plasticity. Notably, pan-histone deacetylase inhibitors lift promoter-specific repression imposed by SUMOylation-deficient WASp and restore misregulated gene expression. Our findings uncovering a SUMOylation-based mechanism controlling WASp's dichotomous roles in transcription may have implications for personalized therapy for patients carrying mutations that perturb WASp-SUMOylation.


Subject(s)
Gene Expression Regulation/immunology , Mutation , NF-kappa B/immunology , Th1 Cells/immunology , Transcriptional Activation/physiology , Wiskott-Aldrich Syndrome Protein/genetics , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Blotting, Western , Cell Line , Electrophoretic Mobility Shift Assay , Flow Cytometry , Humans , Immunoprecipitation , Mass Spectrometry , Mutagenesis, Site-Directed , NF-kappa B/genetics , Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Sumoylation , Transfection
9.
Blood ; 124(23): 3409-19, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-25253772

ABSTRACT

Wiskott-Aldrich syndrome (WAS), an immunodeficiency disorder, and X-linked thrombocytopenia (XLT), a bleeding disorder, both arise from nonsynonymous mutations in WAS, which encodes a hematopoietic-specific WASp. Intriguingly, XLT evolves into WAS in some patients but not in others; yet the biological basis for this cross-phenotype (CP) effect remains unclear. Using human T-helper (TH) cells expressing different disease-causing WAS mutations, we demonstrated that hSWI/SNF-like complexes require nuclear-WASp to execute their chromatin-remodeling activity at promoters of WASp-target, immune function genes during TH1 differentiation. Hot-spot WAS mutations Thr45Met and Arg86Cys, which result in XLT-to-WAS disease progression, impair recruitment of hBRM- but not BRG1-enriched BAF complexes to IFNG and TBX21 promoters. Moreover, promoter enrichment of histone H2A.Z and its catalyzing enzyme EP400 are both impaired. Consequently, activation of Notch signaling, a hBRM-regulated event, and its downstream effector NF-κB are both compromised, along with decreased accessibility of nucleosomal DNA and inefficient transcription-elongation of WASp-target TH1 genes. In contrast, patient mutations Ala236Gly and Arg477Lys that manifest in XLT without progressing to WAS do not disrupt chromatin remodeling or transcriptional reprogramming of TH1 genes. Our study defines an indispensable relationship between nuclear-WASp- and hSWI/SNF-complexes in gene activation and reveals molecular distinctions in TH cells that might contribute to disease severity in the XLT/WAS clinical spectrum.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Genetic Diseases, X-Linked/diagnosis , T-Lymphocytes/metabolism , Thrombocytopenia/diagnosis , Transcription Factors/genetics , Transcription Factors/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/diagnosis , Cell Nucleus/genetics , Cells, Cultured , Diagnosis, Differential , Genetic Diseases, X-Linked/genetics , Humans , Mutation , Promoter Regions, Genetic , Th1 Cells/metabolism , Thrombocytopenia/genetics , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome Protein/metabolism
10.
J Immunol ; 193(1): 150-60, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24872192

ABSTRACT

Defects in Wiskott-Aldrich Syndrome protein (WASp) underlie development of WAS, an X-linked immunodeficiency and autoimmunity disorder of childhood. Nucleation-promoting factors (NPFs) of the WASp family generate F-actin in the cytosol via the VCA (verprolin-homology, cofilin-homology, and acidic) domain and support RNA polymerase II-dependent transcription in the nucleus. Whether nuclear-WASp requires the integration of its actin-related protein (ARP)2/3-dependent cytoplasmic function to reprogram gene transcription, however, remains unresolved. Using the model of human TH cell differentiation, we find that WASp has a functional nuclear localizing and nuclear exit sequences, and accordingly, its effects on transcription are controlled mainly at the level of its nuclear entry and exit via the nuclear pore. Human WASp does not use its VCA-dependent, ARP2/3-driven, cytoplasmic effector mechanisms to support histone H3K4 methyltransferase activity in the nucleus of TH1-skewed cells. Accordingly, an isolated deficiency of nuclear-WASp is sufficient to impair the transcriptional reprogramming of TBX21 and IFNG promoters in TH1-skewed cells, whereas an isolated deficiency of cytosolic-WASp does not impair this process. In contrast, nuclear presence of WASp in TH2-skewed cells is small, and its loss does not impair transcriptional reprogramming of GATA3 and IL4 promoters. Our study unveils an ARP2/3:VCA-independent function of nuclear-WASp in TH1 gene activation that is uncoupled from its cytoplasmic role in actin polymerization.


Subject(s)
Actin-Related Protein 2-3 Complex/immunology , Actins/immunology , Cell Nucleus/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Transcription, Genetic/immunology , Wiskott-Aldrich Syndrome Protein/immunology , Actin-Related Protein 2-3 Complex/genetics , Actins/genetics , Cell Nucleus/genetics , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , HeLa Cells , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Jurkat Cells , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Transcription, Genetic/genetics , Wiskott-Aldrich Syndrome Protein/genetics
11.
Clin Orthop Relat Res ; 472(12): 3869-78, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24682741

ABSTRACT

BACKGROUND: Previous designs of internal bone lengthening devices have been fraught with imprecise distraction, resulting in nerve injuries, joint contractures, nonunions, and other complications. Recently, a magnet-operated PRECICE nail (Ellipse Technologies, Inc, Irvine, CA, USA) was approved by the FDA; however, its clinical efficacy is unknown. QUESTIONS/PURPOSES: We evaluated this nail in terms of (1) accuracy and precision of distraction, (2) effects on bone alignment, (3) effects on adjacent-joint ROM, and (4) frequency of implant-related and non-implant-related complications. METHODS: We reviewed medical and radiographic records of 24 patients who underwent femoral and/or tibial lengthening procedures using the PRECICE nail from August 2012 to July 2013 for conditions of varied etiology, the most common being congenital limb length discrepancy, posttraumatic growth arrest, and fracture malunion. This group represented 29% of patients (24 of 82) who underwent a limb lengthening procedure for a similar diagnosis during the review period. At each postoperative visit, the accuracy and precision of distraction, bone alignment, joint ROM, and any complications were recorded by the senior surgeon (SRR). Accuracy reflected how close the measured lengthening was to the prescribed distraction at each postoperative visit, while precision reflected how close the repeated measurements were to each other over the course of total lengthening period. No patients were lost to followup. Minimum followup from surgery was 3 weeks (mean, 14 weeks; range, 3-29 weeks). RESULTS: Mean total lengthening was 35 mm (range, 14-65 mm), with an accuracy of 96% and precision of 86%. All patients achieved target lengthening with minimal unintentional effects on bone alignment. The knee and ankle ROM were minimally affected. Of the complications requiring return to the operating room for an additional surgical procedure, there was one (4%) implant failure caused by a nonfunctional distraction mechanism and six (24%) non-implant-related complications, including premature consolidation in one patient (4%), delayed bone healing in two (8%), delayed equinus contracture in two (8%), and toe clawing in one (4%). CONCLUSIONS: We conclude that this internal lengthening nail is a valid option to achieve accurate and precise limb lengthening to treat a variety of conditions with limb shortening or length discrepancy. Randomized, larger-sample, long-term studies are required to further confirm clinical efficacy of these devices, monitor for any late failures and complications, and compare with other internal lengthening devices with different mechanisms of operation. LEVEL OF EVIDENCE: Level IV, therapeutic study. See Instructions for Authors for a complete description of levels of evidence.


Subject(s)
Bone Nails , Femur/surgery , Leg Length Inequality/surgery , Magnets , Osteogenesis, Distraction/instrumentation , Tibia/surgery , Adolescent , Adult , Aged , Biomechanical Phenomena , Female , Femur/abnormalities , Femur/diagnostic imaging , Femur/injuries , Femur/physiopathology , Humans , Joints/physiopathology , Leg Length Inequality/diagnosis , Leg Length Inequality/etiology , Leg Length Inequality/physiopathology , Male , Middle Aged , Osteogenesis, Distraction/adverse effects , Postoperative Complications/etiology , Prosthesis Design , Radiography , Range of Motion, Articular , Retrospective Studies , Tibia/abnormalities , Tibia/diagnostic imaging , Tibia/injuries , Tibia/physiopathology , Time Factors , Treatment Outcome , Young Adult
12.
Pediatr Blood Cancer ; 61(8): 1460-2, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24420417

ABSTRACT

Congenital dyserythropoietic anemia (CDA) type-1 is a rare genetic disorder of ineffective erythropoiesis, which manifests in macrocytic anemia. We report a CDA1 patient who as a newborn presented with macrocytic anemia and persistent pulmonary hypertension of the newborn (PPHN) requiring mechanical ventilation. Post-infancy, the patient developed acral dysmorphism and pectus excavatum the latter rarely found in CDA1. Patient is a compound heterozygote for a known maternal-derived missense-mutation (c.1796A > G/p.Asn589Ser) and a novel paternal-derived deletion-mutation (c.1104_1106del/Phe365del) in CDAN1. This report highlights the importance of recognizing PPHN as a presenting symptom of CDA1 and expands the repertoire of the accompanying mutations and axial skeletal malformations.


Subject(s)
Anemia, Dyserythropoietic, Congenital , Glycoproteins/genetics , Hypertension, Pulmonary , Mutation, Missense , Thorax/abnormalities , Amino Acid Substitution , Anemia, Dyserythropoietic, Congenital/complications , Anemia, Dyserythropoietic, Congenital/genetics , Anemia, Dyserythropoietic, Congenital/pathology , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Infant, Newborn , Male , Nuclear Proteins
13.
J Shoulder Elbow Surg ; 21(1): 105-15, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21420320

ABSTRACT

BACKGROUND: Posterior glenoid defects increase the risk of glenoid component loosening after total shoulder arthroplasty (TSA). The goal of this work was to evaluate the mechanical performance of a novel posterior-step glenoid prosthesis, designed to compensate for biconcave (type B2) glenoid defects. Two prototypes ("Poly-step" and "Ti-step") were constructed by attaching polyethylene or titanium step-blocks onto standard (STD) glenoid prostheses. We hypothesized that the mechanical performance of the experimental prostheses in the presence of a B2 defect would be similar to that of an STD prosthesis in the absence of a defect. METHODS: Fifteen normal shoulder specimens were consistently loaded under simulated muscle activity while peri-glenoid bone strains were measured. In 5 specimens, arthroplasty was performed with an STD glenoid prosthesis. In the remaining 10 specimens, a 20° B2 glenoid defect was created before arthroplasty was performed with the Poly-step or Ti-step prosthesis. RESULTS: Load-induced peri-glenoid strains after TSA with either the STD or Poly-step prosthesis did not show statistical differences as compared with the native joints (P > .05). A posterior defect decreased superior glenoid strain as compared with the intact specimens (P < .05). The change in strains after Poly-step prosthesis implantation in the presence of a biconcave glenoid defect was not different than the change induced by STD prosthesis implantation in the absence of a defect. In contrast, strains after Ti-step prosthesis implantation were statistically different from those induced by the STD and Poly-step prostheses (P < .05). CONCLUSIONS: The Poly-step prosthesis may be a viable option for treating posterior glenoid defects.


Subject(s)
Arthroplasty, Replacement/methods , Humerus/surgery , Joint Diseases/surgery , Joint Prosthesis , Shoulder Joint/surgery , Biomechanical Phenomena , Cadaver , Humans , Joint Diseases/physiopathology , Materials Testing , Prosthesis Design , Shoulder Joint/physiopathology
14.
J Clin Invest ; 120(9): 3242-54, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20714107

ABSTRACT

Allergic bronchopulmonary aspergillosis (ABPA) is caused by a dominant Th2 immune response to antigens derived from the opportunistic mold Aspergillus, most commonly Aspergillus fumigatus. It occurs in 4%-15% of patients with cystic fibrosis (CF); however, not all patients with CF infected with A. fumigatus develop ABPA. Therefore, we compared cohorts of A. fumigatus-colonized CF patients with and without ABPA to identify factors mediating tolerance versus sensitization. We found that the costimulatory molecule OX40 ligand (OX40L) was critical in driving Th2 responses to A. fumigatus in peripheral CD4+ T cells isolated from patients with ABPA. In contrast, CD4+ T cells from the non-ABPA cohort did not mount enhanced Th2 responses in vitro and contained a higher frequency of TGF-beta-expressing regulatory T cells. Heightened Th2 reactivity in the ABPA cohort correlated with lower mean serum vitamin D levels. Further, in vitro addition of 1,25 OH-vitamin D3 substantially reduced DC expression of OX40L and increased DC expression of TGF-beta. This in vitro treatment also resulted in increased Treg TGF-beta expression and reduced Th2 responses by CD4+ T cells from patients with ABPA. These data provide rationale for a therapeutic trial of vitamin D to prevent or treat ABPA in patients with CF.


Subject(s)
Aspergillosis, Allergic Bronchopulmonary/immunology , Aspergillus fumigatus/immunology , CD4-Positive T-Lymphocytes/immunology , Cholecalciferol/pharmacology , Cystic Fibrosis/immunology , Th2 Cells/immunology , Adult , Aspergillus/immunology , Female , Humans , Male , Receptors, OX40/metabolism , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology
15.
Sci Transl Med ; 2(37): 37ra44, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20574068

ABSTRACT

The clinical symptomatology in the X-linked Wiskott-Aldrich syndrome (WAS), a combined immunodeficiency and autoimmune disease resulting from WAS protein (WASp) deficiency, reflects the underlying coexistence of an impaired T helper 1 (TH1) immunity alongside intact TH2 immunity. This suggests a role for WASp in patterning T(H) subtype immunity, yet the molecular basis for the TH1-TH2 imbalance in human WAS is unknown. We have discovered a nuclear role for WASp in the transcriptional regulation of the TH1 regulator gene TBX21 at the chromatin level. In primary TH1-differentiating cells, a fraction of WASp is found in the nucleus, where it is recruited to the proximal promoter locus of the TBX21 gene, but not to the core promoter of GATA3 (a TH2 regulator gene) or RORc (a TH17 regulator gene). Genome-wide mapping demonstrates association of WASp in vivo with the gene-regulatory network that orchestrates TH1 cell fate choice in the human TH cell genome. Functionally, nuclear WASp associates with H3K4 trimethyltransferase [RBBP5 (retinoblastoma-binding protein 5)] and H3K9/H3K36 tridemethylase [JMJD2A (Jumonji domain-containing protein 2A)] proteins, and their enzymatic activity in vitro and in vivo is required for achieving transcription-permissive chromatin dynamics at the TBX21 proximal promoter in primary differentiating TH1 cells. During TH1 differentiation, the loss of WASp accompanies decreased enrichment of RBBP5 and, in a subset of WAS patients, also of filamentous actin at the TBX21 proximal promoter locus. Accordingly, human WASp-deficient TH cells, from natural mutation or RNA interference-mediated depletion, demonstrate repressed TBX21 promoter dynamics when driven under TH1-differentiating conditions. These chromatin derangements accompany deficient T-BET messenger RNA and protein expression and impaired TH1 function, defects that are ameliorated by reintroducing WASp. Our findings reveal a previously unappreciated role of WASp in the epigenetic control of T-BET transcription and provide a new mechanism for the pathogenesis of WAS by linking aberrant histone methylation at the TBX21 promoter to dysregulated adaptive immunity.


Subject(s)
Cell Nucleus/metabolism , Immunity/immunology , Th1 Cells/immunology , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/physiopathology , Actins/metabolism , Cell Differentiation , Cells, Cultured , Cellular Reprogramming/genetics , Chromatin/metabolism , DNA/metabolism , Epigenesis, Genetic , Genetic Loci/genetics , Genome, Human/genetics , Histones/metabolism , Humans , Methylation , Mutant Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Transport , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th1 Cells/cytology , Transcription, Genetic , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/deficiency
16.
J Immunol ; 180(7): 4668-78, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18354190

ABSTRACT

Cigarette smoke, a major risk factor in emphysema, causes cell death by incompletely understood mechanisms. Death-inducing signaling complex (DISC) formation is an initial event in Fas-mediated apoptosis. We demonstrate that cigarette smoke extract (CSE) induces DISC formation in human lung fibroblasts (MRC-5) and promotes DISC trafficking from the Golgi complex to membrane lipid rafts. We demonstrate a novel role of protein kinase C (PKC) in the regulation of DISC formation and trafficking. The PKC isoforms, PKCalpha, zeta, epsilon, and eta, were activated by CSE exposure. Overexpression of wild-type PKCalpha inhibited, while PKCzeta promoted, CSE-induced cell death. Dominant-negative (dn)PKCzeta protected against CSE-induced cell death by suppressing DISC formation and caspase-3 activation, while dnPKCalpha enhanced cell death by promoting these events. DISC formation was augmented by wortmannin, an inhibitor of PI3K. CSE-induced Akt phosphorylation was reduced by dnPKCalpha, but it was increased by dnPKCzeta. Expression of PKCalpha in vivo inhibited DISC formation, caspase-3/8 activation, lung injury, and cell death after prolonged cigarette smoke exposure, whereas expression of PKCzeta promoted caspase-3 activation. In conclusion, CSE-induced DISC formation is differentially regulated by PKCalpha and PKCzeta via the PI3K/Akt pathway. These results suggest that modulation of PKC may have therapeutic potential in the prevention of smoke-related lung injury.


Subject(s)
Apoptosis/drug effects , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Nicotiana/chemistry , Protein Kinase C-alpha/metabolism , Protein Kinase C/metabolism , Tobacco Smoke Pollution , Animals , Caspases/metabolism , Cell Line , Enzyme Activation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Phosphoserine/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism
17.
Future Cardiol ; 4(4): 409-26, 2008 Jul.
Article in English | MEDLINE | ID: mdl-19804321

ABSTRACT

Neovacularization is an important biological process whereby new blood vessels develop in both health and disease. During development, blood vessels are formed from mesodermal cells in a process called vasculogenesis. The vascular network then expands by the sprouting of new vessel networks from pre-established vessels in a process known as angiogenesis. However, in adult life, undesirable neovascularization is associated with tumor development and a growing list of 'angiogenesis-dependent' diseases, including cardiovascular complications. Furthermore, diseases characterized by ischemia-induced tissue damage cause a neovascularization response to facilitate tissue repair. Recent research has identified novel molecular and cellular mediators of neovascularization that, in adult life, recapitulate angiogenic processes observed during embryonic development. The discovery of vascular progenitor cells and new molecules that display selective functions in modulating endothelial cell fate, migration and patterning, vessel morphogenesis and the amplification of angiogenic signaling by regulating the master signal VEGF, opens the door to new clinical strategies that target angiogenesis-dependent diseases or that can promote therapeutic neovascularization.

18.
J Immunol ; 179(2): 819-29, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617572

ABSTRACT

Direct T cell allorecognition underlies the development of a vigorous immune response in the clinical setting of acute graft rejection. The Notch pathway is an important regulator of Th immune responses, yet the molecular underpinnings of directional Notch signaling, otherwise critical for binary cell fate decisions, are unknown during autologous or allogeneic Th:DC interactions. Using the development of immune synapses (IS) in the allogeneic, human physiological Th:DC interaction, we demonstrate that Th-Notch1 receptor and DC-Notch ligands (Delta-like1, Jagged1) cluster in their apposed central-supramolecular-activation-clusters (cSMAC), whereas DC-Notch1 receptor and Th-Notch ligands cluster in their apposed peripheral-SMAC (pSMAC). Numb, a negative regulator of Notch, is excluded from the IS-microdomains where Notch1 receptor accumulates. This antiparallel arrangement across the partnering halves of the IS supports reciprocal Notch signal propagation in the DC-to-Th direction via the cSMAC and Th-to-DC direction via the pSMAC. As a result, processed Notch1 receptor (Notch-intracellular-domain, NICD1) and its ligands, as well as their downstream targets, HES-1 and phosphorylated-STAT3, accumulate in the nuclei of both cell-types. There is also enhancement of GLUT1 expression in both cell-types, as well as increased production of Th-IFN-gamma. Significantly, neutralizing Notch1R Ab inhibits NICD1 and HES-1 nuclear translocation, and production of IFN-gamma. In contrast, the IS formed during Ag-nonspecific, autologous Th:DC interaction is immature, resulting in failure of Notch1 receptor segregation and subsequent nuclear translocation of NICD1. Our results provide the first evidence for the asymmetric recruitment of Notch components in the Th:DC immunological synapse, which regulates the bidirectional Notch signal propagation.


Subject(s)
Dendritic Cells/immunology , Lymphocyte Activation/immunology , Receptors, Notch/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Antigen Presentation , Cell Communication/immunology , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Imaging, Three-Dimensional , Receptors, Notch/metabolism , T-Lymphocytes/metabolism , Transplantation, Homologous
19.
Proc Natl Acad Sci U S A ; 103(20): 7777-82, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16682645

ABSTRACT

Statins, best known for their lipid-lowering actions, also possess immunomodulatory properties. Recent studies have shown a Th2-biasing effect of statins, although the underlying mechanism has not been identified. In this study, we investigated whether simvastatin can exercise a Th2-promoting effect through modulation of function of dendritic cells (DCs) without direct interaction with CD4+ T cells. Exposure of DCs to simvastatin induced the differentiation of a distinct subset of DCs characterized by a high expression of B220. These simvastatin-conditioned DCs up-regulated GATA-3 expression and down-regulated T-bet expression in cocultured CD4+ T cells in the absence of additional simvastatin added to the coculture. The Th2-biased transcription factor profile induced by simvastatin-treated DCs also was accompanied by increased Th2 (IL-4, IL-5, and IL-13) and decreased Th1 (IFN-gamma) cytokine secretion from the T cells. The Th2-promoting effect of simvastatin was found to depend on the chitinase family member Ym1, known to be a lectin. Anti-Ym1 antibody abolished the Th2-promoting effect of simvastatin-treated DCs. Also, simvastatin was unable to augment Ym1 expression in DCs developed from STAT6-/- or IL-4R alpha-/- mice. Thus, modulation of Ym1 production by DCs identifies a previously undescribed mechanism of Th2 polarization by statin.


Subject(s)
Dendritic Cells/immunology , Hypolipidemic Agents/pharmacology , Lectins/metabolism , Simvastatin/pharmacology , Th2 Cells , beta-N-Acetylhexosaminidases/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Shape , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , GATA3 Transcription Factor/metabolism , Humans , Interferon-gamma/metabolism , Lectins/genetics , Leukocyte Common Antigens/metabolism , Lymphocyte Subsets , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Phenotype , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Th1 Cells/drug effects , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/metabolism , beta-N-Acetylhexosaminidases/genetics
20.
Cancer ; 104(10): 2214-21, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16206297

ABSTRACT

BACKGROUND: Overall survival after recurrence of osteosarcoma (OS) is < 30%. The authors reported their experience treating recurrent OS at the time of first recurrence (R1). METHODS: Patients with high-grade OS who achieved complete disease remission (CR) after primary surgery and chemotherapy, and patients who were treated at R1 at Memorial Sloan-Kettering Cancer Center (New York, NY) after 1990 were analyzed by retrospective chart review. RESULTS: For 43 eligible patients, the median time to R1 from initial diagnosis was 21.7 months (range, 4.6-135.7 mos). The lungs were the most common sites of disease recurrence (n = 33 of 43). With a median follow-up of 15.2 months (range, 0.7-158.3 mos) after R1, 15 of 43 (35%) patients were alive. Four of 43 patients were treated with surgery alone (3 patients were alive and 1 had died of progressive disease at the time of last follow-up). Due to unresectable disease, eight patients received only chemotherapy, none of whom survived. For patients with disease recurrence treated with chemotherapy and surgery (n = 31), 22 patients achieved a second CR (CR2). Nine patients were alive and in disease remission (29%) at the time of last follow-up. Twenty-three patients received ifosfamide as part of their retrieval regimen. Of the 18 who achieved a CR2, 8 experienced disease recurrence, 7 remain alive in CR2, and 3 died due to toxicity. Eight patients did not receive ifosfamide. Of these, 4 achieved a CR2 but 3 subsequently experienced disease recurrence. CONCLUSIONS: At R1, 22 of 31 patients achieved a CR2 with aggressive surgery and chemotherapy. The majority of these patients subsequently developed a disease recurrence. Patients appeared to benefit from the addition of ifosfamide to their retrieval regimens. In the end, the role of chemotherapy in recurrent OS continues to remain undefined.


Subject(s)
Bone Neoplasms/therapy , Neoplasm Recurrence, Local/therapy , Osteosarcoma/therapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/mortality , Child , Child, Preschool , Female , Humans , Male , Neoplasm Recurrence, Local/mortality , Orthopedic Procedures , Osteosarcoma/mortality , Retrospective Studies , Survival Analysis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL