Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 174
Filter
1.
Croat Med J ; 65(3): 232-238, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38868969

ABSTRACT

AIM: To determine variations in allele and genotype frequencies between keratoacanthoma (KA) and common warts (CW), compared with the control group, in three single nucleotide polymorphisms (SNPs) within the TLR2, TLR3, and TLR9 genes. METHODS: This case-control study involved samples from 161 patients with KA, 152 patients with CW, and 469 controls. DNA was isolated from formalin-fixed paraffin-embedded tissue sections. Three SNPs - rs4696480 in TLR2, rs7657186 in TLR9, and rs35213 in TLR3 - were genotyped with TaqMan Genotyping Assays on the 7500 Real-Time PCR System. RESULTS: TLR2 rs4696480 and TLR3 rs7657186 were significantly overrepresented in KA and CW compared with controls (P<0.001). The association was stronger for CW than for KA, as evidenced by higher frequencies of the A allele and AA genotype for rs4696480. Both KA and CW patients had higher frequencies of the G allele and GG genotype for rs7657186 than controls. rs7657186 was moderately associated with KA and CW, with the G allele and GG genotype being more prevalent in CW cases, where no AA homozygotes were found. CONCLUSION: Genetic variants in TLR2 (rs4696480) and TLR3 (rs7657186) genes may affect KA and CW development, influencing immune responses and susceptibility to these skin lesions. Further research is required to elucidate TLR expression patterns and their role in KA development.


Subject(s)
Keratoacanthoma , Polymorphism, Single Nucleotide , Toll-Like Receptor 2 , Toll-Like Receptor 3 , Warts , Adolescent , Adult , Female , Humans , Male , Middle Aged , Young Adult , Alleles , Case-Control Studies , Gene Frequency , Genetic Predisposition to Disease , Genotype , Keratoacanthoma/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 3/genetics , Warts/genetics
4.
J Clin Periodontol ; 51(4): 464-473, 2024 04.
Article in English | MEDLINE | ID: mdl-38185798

ABSTRACT

AIM: WHIM (warts, hypogammaglobulinaemia, infections and myelokathexis) syndrome is a rare combined primary immunodeficiency disease caused by gain-of-function (GOF) mutations in the chemokine receptor CXCR4 and includes severe neutropenia as a common feature. Neutropenia is a known risk factor for periodontitis; however, a detailed periodontal evaluation of a WHIM syndrome cohort is lacking. This study aimed to establish the evidence base for the periodontal status of patients with WHIM syndrome. MATERIALS AND METHODS: Twenty-two adult WHIM syndrome patients and 22 age- and gender-matched healthy volunteers (HVs) were evaluated through a comprehensive medical and periodontal examination. A mouse model of WHIM syndrome was assessed for susceptibility to naturally progressing or inducible periodontitis. RESULTS: Fourteen patients with WHIM syndrome (63.6%) and one HV (4.5%) were diagnosed with Stage III/IV periodontitis. No WHIM patient presented with the early onset, dramatic clinical phenotypes typically associated with genetic forms of neutropenia. Age, but not the specific CXCR4 mutation or absolute neutrophil count, was associated with periodontitis severity in the WHIM cohort. Mice with a Cxcr4 GOF mutation did not exhibit increased alveolar bone loss in spontaneous or ligature-induced periodontitis. CONCLUSIONS: Overall, WHIM syndrome patients presented with an increased severity of periodontitis despite past and ongoing neutrophil mobilization treatments. GOF mutations in CXCR4 may be a risk factor for periodontitis in humans.


Subject(s)
Immunologic Deficiency Syndromes , Neutropenia , Periodontal Diseases , Periodontitis , Primary Immunodeficiency Diseases , Warts , Adult , Humans , Animals , Mice , Immunologic Deficiency Syndromes/complications , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/diagnosis , Primary Immunodeficiency Diseases/complications , Primary Immunodeficiency Diseases/genetics , Warts/genetics , Warts/therapy , Neutropenia/complications , Neutropenia/genetics , Periodontal Diseases/complications , Periodontal Diseases/genetics , Periodontitis/complications , Periodontitis/genetics
6.
Genet Med ; 26(2): 101028, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37978863

ABSTRACT

PURPOSE: Persistent human papillomavirus infection (PHPVI) causes cutaneous, anogenital, and mucosal warts. Cutaneous warts include common warts, Treeman syndrome, and epidermodysplasia verruciformis, among others. Although more reports of monogenic predisposition to PHPVI have been published with the development of genomic technologies, genetic testing is rarely incorporated into clinical assessments. To encourage broader molecular testing, we compiled a list of the various monogenic etiologies of PHPVI. METHODS: We conducted a systematic literature review to determine the genetic, immunological, and clinical characteristics of patients with PHPVI. RESULTS: The inclusion criteria were met by 261 of 40,687 articles. In 842 patients, 83 PHPVI-associated genes were identified, including 42, 6, and 35 genes with strong, moderate, and weak evidence for causality, respectively. Autosomal recessive inheritance predominated (69%). PHPVI onset age was 10.8 ± 8.6 years, with an interquartile range of 5 to 14 years. GATA2,IL2RG,DOCK8, CXCR4, TMC6, TMC8, and CIB1 are the most frequently reported PHPVI-associated genes with strong causality. Most genes (74 out of 83) belong to a catalog of 485 inborn errors of immunity-related genes, and 40 genes (54%) are represented in the nonsyndromic and syndromic combined immunodeficiency categories. CONCLUSION: PHPVI has at least 83 monogenic etiologies and a genetic diagnosis is essential for effective management.


Subject(s)
Epidermodysplasia Verruciformis , Papillomavirus Infections , Warts , Humans , Child, Preschool , Child , Adolescent , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Warts/genetics , Warts/complications , Epidermodysplasia Verruciformis/genetics , Epidermodysplasia Verruciformis/complications , Skin , Syndrome , Membrane Proteins/genetics , Guanine Nucleotide Exchange Factors
7.
J Clin Invest ; 133(19)2023 10 02.
Article in English | MEDLINE | ID: mdl-37561579

ABSTRACT

BACKGROUNDWarts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome is a primary immunodeficiency disorder caused by heterozygous gain-of-function CXCR4 mutations. Myelokathexis is a kind of neutropenia caused by neutrophil retention in bone marrow and in WHIM syndrome is associated with lymphopenia and monocytopenia. The CXCR4 antagonist plerixafor mobilizes leukocytes to the blood; however, its safety and efficacy in WHIM syndrome are undefined.METHODSIn this investigator-initiated, single-center, quadruple-masked phase III crossover trial, we compared the total infection severity score (TISS) as the primary endpoint in an intent-to-treat manner in 19 patients with WHIM who each received 12 months treatment with plerixafor and 12 months treatment with granulocyte CSF (G-CSF, the standard of care for severe congenital neutropenia). The treatment order was randomized for each patient.RESULTSPlerixafor was nonsuperior to G-CSF for TISS (P = 0.54). In exploratory endpoints, plerixafor was noninferior to G-CSF for maintaining neutrophil counts of more than 500 cells/µL (P = 0.023) and was superior to G-CSF for maintaining lymphocyte counts above 1,000 cells/µL (P < 0.0001). Complete regression of a subset of large wart areas occurred on plerixafor in 5 of 7 patients with major wart burdens at baseline. Transient rash occurred on plerixafor, and bone pain was more common on G-CSF. There were no significant differences in drug preference or quality of life or the incidence of drug failure or serious adverse events.CONCLUSIONPlerixafor was not superior to G-CSF in patients with WHIM for TISS, the primary endpoint. Together with wart regression and hematologic improvement, the infection severity results support continued study of plerixafor as a potential treatment for WHIM syndrome.TRIAL REGISTRATIONClinicaltrials.gov NCT02231879.FUNDINGThis study was funded by the Division of Intramural Research, National Institute of Allergy and Infectious Diseases.


Subject(s)
Heterocyclic Compounds , Immunologic Deficiency Syndromes , Primary Immunodeficiency Diseases , Warts , Humans , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/genetics , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Mobilization/adverse effects , Cross-Over Studies , Quality of Life , Heterocyclic Compounds/adverse effects , Primary Immunodeficiency Diseases/drug therapy , Primary Immunodeficiency Diseases/genetics , Warts/drug therapy , Warts/genetics , Receptors, CXCR4/genetics
9.
JCI Insight ; 8(5)2023 03 08.
Article in English | MEDLINE | ID: mdl-36883568

ABSTRACT

WHIM syndrome is an inherited immune disorder caused by an autosomal dominant heterozygous mutation in CXCR4. The disease is characterized by neutropenia/leukopenia (secondary to retention of mature neutrophils in bone marrow), recurrent bacterial infections, treatment-refractory warts, and hypogammaglobulinemia. All mutations reported in WHIM patients lead to the truncations in the C-terminal domain of CXCR4, R334X being the most frequent. This defect prevents receptor internalization and enhances both calcium mobilization and ERK phosphorylation, resulting in increased chemotaxis in response to the unique ligand CXCL12. Here, we describe 3 patients presenting neutropenia and myelokathexis, but normal lymphocyte count and immunoglobulin levels, carrying what we believe to be a novel Leu317fsX3 mutation in CXCR4, leading to a complete truncation of its intracellular tail. The analysis of the L317fsX3 mutation in cells derived from patients and in vitro cellular models reveals unique signaling features in comparison with R334X mutation. The L317fsX3 mutation impairs CXCR4 downregulation and ß-arrestin recruitment in response to CXCL12 and reduces other signaling events - including ERK1/2 phosphorylation, calcium mobilization, and chemotaxis - all processes that are typically enhanced in cells carrying the R334X mutation. Our findings suggest that, overall, the L317fsX3 mutation may be causative of a form of WHIM syndrome not associated with an augmented CXCR4 response to CXCL12.


Subject(s)
GTP-Binding Proteins , Primary Immunodeficiency Diseases , beta-Arrestins , Humans , beta-Arrestin 1/genetics , beta-Arrestin 1/immunology , beta-Arrestins/genetics , beta-Arrestins/immunology , Calcium/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Mutation , Neutropenia/genetics , Neutropenia/immunology , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Signal Transduction/genetics , Signal Transduction/physiology , Warts/genetics , Warts/immunology
10.
Blood ; 142(1): 23-32, 2023 07 06.
Article in English | MEDLINE | ID: mdl-36928087

ABSTRACT

WHIM syndrome is an autosomal dominant immunodeficiency disorder caused by gain-of-function mutations in chemokine receptor CXCR4 that promote severe panleukopenia because of retention of mature leukocytes in the bone marrow (BM). We previously reported that Cxcr4-haploinsufficient (Cxcr4+/o) hematopoietic stem cells (HSCs) have a strong selective advantage for durable hematopoietic reconstitution over wild-type (Cxcr4+/+) and WHIM (Cxcr4+/w) HSCs and that a patient with WHIM was spontaneously cured by chromothriptic deletion of the disease allele in an HSC, suggesting that WHIM allele inactivation through gene editing may be a safe genetic cure strategy for the disease. We have developed a 2-step preclinical protocol of autologous hematopoietic stem and progenitor cell (HSPC) transplantation to achieve this goal. First, 1 copy of Cxcr4 in HSPCs was inactivated in vitro by CRISPR/Cas9 editing with a single guide RNA (sgRNA) that does not discriminate between Cxcr4+/w and Cxcr4+/+ alleles. Then, through in vivo natural selection, WHIM allele-inactivated cells were enriched over wild-type allele-inactivated cells. The WHIM allele-inactivated HSCs retained long-term pluripotency and selective hematopoietic reconstitution advantages. To our knowledge, this is the first example of gene therapy for an autosomal dominant gain-of-function disease using a disease allele inactivation strategy in place of the less efficient disease allele repair approach.


Subject(s)
Immunologic Deficiency Syndromes , Warts , Mice , Animals , Alleles , CRISPR-Cas Systems , RNA, Guide, CRISPR-Cas Systems , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Warts/genetics , Warts/therapy , Genetic Therapy , Receptors, CXCR4/genetics
11.
Med Sci (Paris) ; 39(1): 23-30, 2023 Jan.
Article in French | MEDLINE | ID: mdl-36692314

ABSTRACT

CXCR4 is a chemokine receptor that plays a central role in cell migration but also in other essential processes such as the development of the immune system. Together with its ligand, the chemokine CXCL12, this signalling axis plays an important role in B lymphocyte biology from their early differentiation in the bone marrow to their activation and differentiation into antibody secreting cells, also called plasma cells. Gain-of-function mutations of CXCR4 are found in a rare immunodeficiency, the WHIM Syndrome. These mutations affect the desensitization of the receptor and lead to a gain of function in response to CXCL12. This review summarizes the role of CXCR4 in the humoral immune responses and using the WHIM Syndrome as a paradigm, highlights the critical regulatory role of CXCR4 desensitization in these processes. Indeed, recent works report that fine-tuning of CXCR4 signalling is essential to limit the extra-follicular immune response and support long term antibody-mediated protection.


Title: La signalisation de CXCR4, un rhéostat de la réponse immunitaire à médiation humorale. Abstract: CXCR4 est un récepteur de chimiokine qui joue un rôle central dans la migration cellulaire mais également dans d'autres mécanismes essentiels, tels que le développement du système immunitaire. De concert avec son ligand naturel, la chimiokine CXCL12, cet axe de signalisation joue un rôle important dans la biologie des lymphocytes B, des stades précoces de différenciation dans la moelle osseuse à leur activation et différenciation en cellules sécrétrices d'anticorps, aussi appelées plasmocytes. Des mutations gain de fonction de CXCR4 sont retrouvées dans une immunodéficience rare, le Syndrome WHIM. Ces mutations affectent le mécanisme de désensibilisation du récepteur et entraînent un gain de fonction en réponse à CXCL12. Cette revue résume le rôle de CXCR4 dans la réponse immune humorale et, à travers l'étude du Syndrome WHIM, souligne le rôle régulateur essentiel de la désensibilisation de CXCR4 dans ces processus. Des travaux récents rapportent en effet qu'une signalisation correcte de CXCR4 est essentielle pour limiter la réponse immune dite « extra-folliculaire ¼ et pour permettre une protection au long terme assurée par les anticorps.


Subject(s)
Immunologic Deficiency Syndromes , Primary Immunodeficiency Diseases , Warts , Humans , Primary Immunodeficiency Diseases/genetics , Immunologic Deficiency Syndromes/genetics , Warts/genetics , Signal Transduction/genetics , Chemokine CXCL12/genetics , Receptors, CXCR4/genetics
12.
J Med Genet ; 60(1): 84-90, 2023 01.
Article in English | MEDLINE | ID: mdl-34916230

ABSTRACT

BACKGROUND: Primary lymphoedema (PL) syndromes are increasingly recognised as presentations of complex genetic disease, with at least 20 identified causative genes. Recognition of clinical patterns is key to diagnosis, research and therapeutics. The defining criteria for one such clinical syndrome, 'WILD syndrome' (Warts, Immunodeficiency, Lymphoedema and anogenital Dysplasia), have previously depended on a single case report. METHODS AND RESULTS: We present 21 patients (including the first described case) with similar clinical and immunological phenotypes. All had PL affecting multiple segments, with systemic involvement (intestinal lymphangiectasia/pleural or pericardial effusions) in 70% (n=14/20). Most (n=20, 95%) had a distinctive cutaneous lymphovascular malformation on the upper anterior chest wall. Some (n=10, 48%) also had hyperpigmented lesions resembling epidermal naevi (but probably lymphatic in origin). Warts were common (n=17, 81%) and often refractory. In contrast to the previous case report, anogenital dysplasia was uncommon-only found in two further cases (total n=3, 14%). Low CD4 counts and CD4:CD8 ratios typified the syndrome (17 of 19, 89%), but monocyte counts were universally normal, unlike GATA2 deficiency. CONCLUSION: WILD syndrome is a previously unrecognised, underdiagnosed generalised PL syndrome. Based on this case series, we redefine WILD as 'Warts, Immunodeficiency, andLymphatic Dysplasia' and suggest specific diagnostic criteria. The essential criterion is congenital multisegmental PL in a 'mosaic' distribution. The major diagnostic features are recurrent warts, cutaneous lymphovascular malformations, systemic involvement (lymphatic dysplasia), genital swelling and CD4 lymphopaenia with normal monocyte counts. The absence of family history suggests a sporadic condition, and the random distribution of swelling implicates mosaic postzygotic mutation as the cause.


Subject(s)
Immunologic Deficiency Syndromes , Lymphedema , Warts , Humans , Warts/diagnosis , Warts/genetics , Lymphedema/diagnosis , Lymphedema/genetics
13.
Genes Immun ; 23(6): 196-204, 2022 09.
Article in English | MEDLINE | ID: mdl-36089616

ABSTRACT

Warts, hypogammaglobulinemia, infections, myelokathexis (WHIM) syndrome is a rare primary immunodeficiency predominantly caused by heterozygous gain-of-function mutations in CXCR4 C-terminus. We assessed genotype-phenotype correlations for known pathogenic CXCR4 variants and in vitro response of each variant to mavorixafor, an investigational CXCR4 antagonist. We used cell-based assays to analyze CXCL12-induced receptor trafficking and downstream signaling of 14 pathogenic CXCR4 variants previously identified in patients with WHIM syndrome. All CXCR4 variants displayed impaired receptor trafficking, hyperactive downstream signaling, and enhanced chemotaxis in response to CXCL12. Mavorixafor inhibited CXCL12-dependent signaling and hyperactivation in cells harboring CXCR4WHIM mutations. A strong correlation was found between CXCR4 internalization defect and severity of blood leukocytopenias and infection susceptibility, and between AKT activation and immunoglobulin A level and CD4+ T-cell counts. This study is the first to show WHIM syndrome clinical phenotype variability as a function of both CXCR4WHIM genotype diversity and associated functional dysregulation. Our findings suggest that CXCR4 internalization may be used to assess the pathogenicity of CXCR4 variants in vitro and also as a potential WHIM-related disease biomarker. The investigational CXCR4 antagonist mavorixafor inhibited CXCL12-dependent signaling in all tested CXCR4-variant cell lines at clinically relevant concentrations.


Subject(s)
Agammaglobulinemia , Immunologic Deficiency Syndromes , Neutropenia , Warts , Agammaglobulinemia/genetics , Aminoquinolines , Benzimidazoles , Biomarkers , Butylamines , Genetic Association Studies , Humans , Immunoglobulin A/genetics , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Neutropenia/genetics , Neutropenia/metabolism , Primary Immunodeficiency Diseases , Proto-Oncogene Proteins c-akt/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Warts/genetics , Warts/metabolism , Warts/pathology
14.
J Clin Immunol ; 42(8): 1748-1765, 2022 11.
Article in English | MEDLINE | ID: mdl-35947323

ABSTRACT

Warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome (WS) is a combined immunodeficiency caused by gain-of-function mutations in the C-X-C chemokine receptor type 4 (CXCR4) gene. We characterize a unique international cohort of 66 patients, including 57 (86%) cases previously unreported, with variable clinical phenotypes. Of 17 distinct CXCR4 genetic variants within our cohort, 11 were novel pathogenic variants affecting 15 individuals (23%). All variants affect the same CXCR4 region and impair CXCR4 internalization resulting in hyperactive signaling. The median age of diagnosis in our cohort (5.5 years) indicates WHIM syndrome can commonly present in childhood, although some patients are not diagnosed until adulthood. The prevalence and mean age of recognition and/or onset of clinical manifestations within our cohort were infections 88%/1.6 years, neutropenia 98%/3.8 years, lymphopenia 88%/5.0 years, and warts 40%/12.1 years. However, we report greater prevalence and variety of autoimmune complications of WHIM syndrome (21.2%) than reported previously. Patients with versus without family history of WHIM syndrome were diagnosed earlier (22%, average age 1.3 years versus 78%, average age 5 years, respectively). Patients with a family history of WHIM syndrome also received earlier treatment, experienced less hospitalization, and had less end-organ damage. This observation reinforces previous reports that early treatment for WHIM syndrome improves outcomes. Only one patient died; death was attributed to complications of hematopoietic stem cell transplantation. The variable expressivity of WHIM syndrome in pediatric patients delays their diagnosis and therapy. Early-onset bacterial infections with severe neutropenia and/or lymphopenia should prompt genetic testing for WHIM syndrome, even in the absence of warts.


Subject(s)
Agammaglobulinemia , Immunologic Deficiency Syndromes , Lymphopenia , Neutropenia , Warts , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/epidemiology , Immunologic Deficiency Syndromes/genetics , Warts/diagnosis , Warts/epidemiology , Warts/genetics , Agammaglobulinemia/genetics , Receptors, CXCR4/genetics , Neutropenia/genetics , Lymphopenia/complications , Disease Progression
15.
Immunohorizons ; 6(7): 543-558, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35882421

ABSTRACT

Warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome immunodeficiency is caused by autosomal dominant gain-of-function CXCR4 mutations that promote severe panleukopenia caused by bone marrow retention of mature leukocytes. Consequently, WHIM patients develop recurrent bacterial infections; however, sepsis is uncommon. To study this clinical dichotomy, we challenged WHIM model mice with LPS. The LD50 was similar in WHIM and wild-type (WT) mice, and LPS induced acute lymphopenia in WT mice that was Cxcr4 independent. In contrast, in WHIM mice, LPS did not affect circulating T cell levels, but the B cell levels anomalously increased because of selective, cell-intrinsic, and Cxcr4 WHIM allele-dependent emergence of Cxcr4high late pre-B cells, a pattern that was phenocopied by Escherichia coli infection. In both WT and WHIM mice, the CXCR4 antagonist AMD3100 rapidly increased circulating lymphocyte levels that then rapidly contracted after subsequent LPS treatment. Thus, LPS-induced lymphopenia is CXCR4 independent, and a WHIM mutation does not increase clinical LPS sensitivity. Anomalous WT Cxcr4-independent, but Cxcr4 WHIM-dependent, promobilizing effects of LPS on late pre-B cell mobilization reveal a distinct signaling pathway for the variant receptor.


Subject(s)
Agammaglobulinemia , Lymphopenia , Neutropenia , Warts , Agammaglobulinemia/genetics , Animals , Endotoxins/therapeutic use , Lipopolysaccharides , Mice , Neutropenia/genetics , Primary Immunodeficiency Diseases , Warts/drug therapy , Warts/genetics
16.
Proc Natl Acad Sci U S A ; 119(21): e2119483119, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35588454

ABSTRACT

Chemokine receptor nanoscale organization at the cell membrane is orchestrated by the actin cytoskeleton and influences cell responses. Using single-particle tracking analysis we show that CXCR4R334X, a truncated mutant chemokine receptor linked to WHIM syndrome (warts, hypogammaglobulinemia, infections, myelokathexis), fails to nanoclusterize after CXCL12 stimulation, and alters the lateral mobility and spatial organization of CXCR4 when coexpressed. These findings correlate with multiple phalloidin-positive protrusions in cells expressing CXCR4R334X, and their inability to correctly sense chemokine gradients. The underlying mechanisms involve inappropriate actin cytoskeleton remodeling due to the inadequate ß-arrestin1 activation by CXCR4R334X, which disrupts the equilibrium between activated and deactivated cofilin. Overall, we provide insights into the molecular mechanisms governing CXCR4 nanoclustering, signaling and cell function, and highlight the essential scaffold role of ß-arrestin1 to support CXCL12-mediated actin reorganization and receptor clustering. These defects associated with CXCR4R334X expression might contribute to the severe immunological symptoms associated with WHIM syndrome.


Subject(s)
Primary Immunodeficiency Diseases , Receptors, CXCR4 , Warts , Actin Depolymerizing Factors/metabolism , Cell Membrane/metabolism , Cell Movement , Humans , Mutation , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Single Molecule Imaging , Warts/genetics , Warts/metabolism
17.
J Invest Dermatol ; 142(9): 2435-2445, 2022 09.
Article in English | MEDLINE | ID: mdl-35276224

ABSTRACT

Recalcitrant warts, caused by human papillomaviruses (HPVs), can be a cutaneous manifestation of inborn error of immunity. This study investigated the clinical manifestations, immunodeficiency, single-gene susceptibility, and HPV repertoire in a consanguineous family with severe sinopulmonary infections and recalcitrant warts. Clinical and immunologic evaluations, including FACS and lymphocyte transformation test, provided evidence for immunodeficiency. Combined whole-exome sequencing and genome-wide homozygosity mapping were utilized to disclose candidate sequence variants. Whole-transcriptome sequencing was used to concomitantly investigate the HPV genotypes and the consequences of detected sequence variants in the host. The proband, a male aged 41 years, was found to be homozygous for the c.6delG, p.Lys2Asnfs∗17 variant in ICOS, encoding the inducible T-cell costimulator. This variant was located inside the 5 megabase of runs of homozygosity on 2q33.2. RNA sequencing confirmed the deleteriousness of the ICOS variant in three skin biopsies revealing significant downregulation of ICOS and its ligand, ICOSLG. Reads unaligned to the human genome were applied to 926 different viruses, and α-HPV57, ß-HPV107, ß-HPV14, and ß-HPV17 were detected. Collectively, we describe a previously unrecognized inborn error of T-cell immunity to HPVs, indicating that autosomal recessive ICOS deficiency can underlie recalcitrant warts, emphasizing the immunologic underpinnings of recalcitrant warts at the nexus of human and viral genomic variation.


Subject(s)
Inducible T-Cell Co-Stimulator Protein , Papillomavirus Infections , Warts , Adult , Humans , Inducible T-Cell Co-Stimulator Protein/genetics , Male , Papillomaviridae , Papillomavirus Infections/genetics , Skin/pathology , Warts/genetics , Warts/pathology , Exome Sequencing
18.
J Biol Chem ; 298(2): 101551, 2022 02.
Article in English | MEDLINE | ID: mdl-34973340

ABSTRACT

WHIM syndrome is a rare immunodeficiency disorder that is characterized by warts, hypogammaglobulinemia, infections, and myelokathexis. While several gain-of-function mutations that lead to C-terminal truncations, frame shifts and point mutations in the chemokine receptor CXCR4 have been identified in WHIM syndrome patients, the functional effect of these mutations are not fully understood. Here, we report on a new WHIM syndrome mutation that results in a frame shift within the codon for Ser339 (S339fs5) and compare the properties of S339fs5 with wild-type CXCR4 and a previously identified WHIM syndrome mutant, R334X. The S339fs5 and R334X mutants exhibited significantly increased signaling compared to wild-type CXCR4 including agonist-promoted calcium flux and extracellular-signal-regulated kinase activation. This increase is at least partially due to a significant decrease in agonist-promoted phosphorylation, ß-arrestin binding, and endocytosis of S339fs5 and R334X compared with wild-type CXCR4. Interestingly, there were also significant differences in receptor degradation, with S339fs5 having a very high basal level of degradation compared with that of R334X and wild-type CXCR4. In contrast to wild-type CXCR4, both R334X and S339fs5 were largely insensitive to CXCL12-promoted degradation. Moreover, while basal and agonist-promoted degradation of wild-type CXCR4 was effectively inhibited by the CXCR4 antagonist TE-14016, this had no effect on the degradation of the WHIM mutants. Taken together, these studies identify a new WHIM syndrome mutant, CXCR4-S339fs5, which promotes enhanced signaling, reduced phosphorylation, ß-arrestin binding and endocytosis, and a very high basal rate of degradation that is not protected by antagonist treatment.


Subject(s)
Primary Immunodeficiency Diseases , Receptors, CXCR4 , Warts , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Humans , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Warts/genetics , Warts/metabolism , beta-Arrestin 1/metabolism , beta-Arrestins/metabolism
19.
J Invest Dermatol ; 142(5): 1265-1269, 2022 05.
Article in English | MEDLINE | ID: mdl-34843682

ABSTRACT

Human papillomavirus (HPV) infections can cause common warts, which usually resolve spontaneously or become recalcitrant, resistant to multiple treatments. In rare cases, they transform into cutaneous giant horns resulting in the tree-man syndrome (TMS). Defective ß-HPVs can cause flat warts in epidermodysplasia verruciformis (EV), a genetic disorder. In typical EV, limited to the skin, the mutated genes are critical for keratinocyte-intrinsic immunity, whereas atypical, syndromic EV involves genes controlling T cells. Inborn errors of immunity due to mutations in distinct genes underlying recalcitrant warts and the α-HPV2‒driven TMS have been identified, all disrupting T-cell immunity. Collectively, these observations attest to the wide phenotypic spectrum of cutaneous infections caused by different HPV types at the intersection of the genetic diversity of the viral and human genomes.


Subject(s)
Epidermodysplasia Verruciformis , Papillomavirus Infections , Warts , DNA, Viral/genetics , Epidermodysplasia Verruciformis/genetics , Genome, Human , Humans , Papillomaviridae/genetics , Papillomavirus Infections/genetics , Syndrome , Warts/genetics
20.
J Clin Immunol ; 42(1): 171-182, 2022 01.
Article in English | MEDLINE | ID: mdl-34697698

ABSTRACT

PURPOSE: WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome is a rare disease, caused by CXCR4 gene mutations, which incorporates features of combined immunodeficiency, congenital neutropenia, and a predisposition to human papillomavirus infection. Established conventional treatment for WHIM syndrome does not fully prevent infectious complications in these patients. Only single case reports of hematopoietic stem cell transplantation (HSCT) efficacy in WHIM have been published. METHODS: To summarize current information on HSCT efficacy in disease treatment, seven pediatric patients with WHIM syndrome who underwent allogeneic HSCT were identified in five centers worldwide. RESULTS: All patients presented early after birth with neutropenia. Two of seven patients exhibited severe disease complications: poorly controlled autoimmunity (arthritis and anemia) in one and progressive myelofibrosis with recurrent infections in the other. The remaining patients received HSCT to correct milder disease symptoms (recurrent respiratory infections, progressing thrombocytopenia) and/or to preclude severe disease course in older age. All seven patients engrafted but one developed graft rejection and died of infectious complications after third HSCT. Three other patients experienced severe viral infections after HSCT (including post-transplant lymphoproliferative disease in one) which completely resolved with therapy. At last follow-up (median 6.7 years), all six surviving patients were alive with full donor chimerism. One patient 1.4 years after HSCT had moderate thrombocytopenia and delayed immune recovery; the others had adequate immune recovery and were free of prior disease symptoms. CONCLUSION: HSCT in WHIM syndrome corrects neutropenia and immunodeficiency, and leads to resolution of autoimmunity and recurrent infections, including warts.


Subject(s)
Hematopoietic Stem Cell Transplantation , Neutropenia , Warts , Child , Humans , Neutropenia/complications , Neutropenia/therapy , Primary Immunodeficiency Diseases , Receptors, CXCR4/genetics , Warts/diagnosis , Warts/genetics , Warts/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...