Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
J Clin Invest ; 133(19)2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37561579

RESUMO

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.


Assuntos
Compostos Heterocíclicos , Síndromes de Imunodeficiência , Doenças da Imunodeficiência Primária , Verrugas , Humanos , Síndromes de Imunodeficiência/tratamento farmacológico , Síndromes de Imunodeficiência/genética , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Mobilização de Células-Tronco Hematopoéticas/efeitos adversos , Estudos Cross-Over , Qualidade de Vida , Compostos Heterocíclicos/efeitos adversos , Doenças da Imunodeficiência Primária/tratamento farmacológico , Doenças da Imunodeficiência Primária/genética , Verrugas/tratamento farmacológico , Verrugas/genética , Receptores CXCR4/genética
2.
J Immunol ; 210(12): 1913-1924, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37133343

RESUMO

Warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome is an ultra-rare combined primary immunodeficiency disease caused by heterozygous gain-of-function mutations in the chemokine receptor CXCR4. WHIM patients typically present with recurrent acute infections associated with myelokathexis (severe neutropenia due to bone marrow retention of mature neutrophils). Severe lymphopenia is also common, but the only associated chronic opportunistic pathogen is human papillomavirus and mechanisms are not clearly defined. In this study, we show that WHIM mutations cause more severe CD8 than CD4 lymphopenia in WHIM patients and WHIM model mice. Mechanistic studies in mice revealed selective and WHIM allele dose-dependent accumulation of mature CD8 single-positive cells in thymus in a cell-intrinsic manner due to prolonged intrathymic residence, associated with increased CD8 single-positive thymocyte chemotactic responses in vitro toward the CXCR4 ligand CXCL12. In addition, mature WHIM CD8+ T cells preferentially home to and are retained in the bone marrow in mice in a cell-intrinsic manner. Administration of the specific CXCR4 antagonist AMD3100 (plerixafor) in mice rapidly and transiently corrected T cell lymphopenia and the CD4/CD8 ratio. After lymphocytic choriomeningitis virus infection, we found no difference in memory CD8+ T cell differentiation or viral load between wild-type and WHIM model mice. Thus, lymphopenia in WHIM syndrome may involve severe CXCR4-dependent CD8+ T cell deficiency resulting in part from sequestration in the primary lymphoid organs, thymus, and bone marrow.


Assuntos
Agamaglobulinemia , Compostos Heterocíclicos , Síndromes de Imunodeficiência , Linfopenia , Neutropenia , Humanos , Animais , Camundongos , Síndromes de Imunodeficiência/genética , Mobilização de Células-Tronco Hematopoéticas/efeitos adversos , Agamaglobulinemia/complicações , Agamaglobulinemia/genética , Neutropenia/genética , Linfócitos T CD8-Positivos , Receptores CXCR4/genética
3.
Blood ; 142(1): 23-32, 2023 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-36928087

RESUMO

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.


Assuntos
Síndromes de Imunodeficiência , Verrugas , Camundongos , Animais , Alelos , Sistemas CRISPR-Cas , RNA Guia de Sistemas CRISPR-Cas , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/terapia , Verrugas/genética , Verrugas/terapia , Terapia Genética , Receptores CXCR4/genética
4.
PLoS Pathog ; 18(11): e1010502, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36318581

RESUMO

The atypical IκB family member Bcl3 associates with p50/NF-κB1 or p52/NF-κB2 homodimers in the nucleus, and positively or negatively modulates transcription in a context-dependent manner. In mice lacking Bcl3 globally or specifically in CD11c+ cells, we previously reported that Toxoplasma gondii infection is uniformly fatal and is associated with an impaired Th1 immune response. Since Bcl3 expression in dendritic cells (DC) is pivotal for antigen presentation and since classical DCs (cDC) are major antigen presenting cells, we investigated the role of Bcl3 specifically in cDCs in vivo by crossing Zbtb46 cre mice with Bcl3flx/flx mice. Bcl3flx/flx Zbtb46 cre mice were as susceptible to lethal T. gondii infection as total Bcl3-/- mice and generated poor Th1 immune responses. Consistent with this, compared to wildtype controls, splenic Xcr1+ Bcl3-deficient cDC1 cells were defective in presenting Ova antigen to OT-I cells both for Ova257-264 peptide and after infection with Ovalbumin-expressing T. gondii. Moreover, splenic CD4+ and CD8+ T cells from infected Bcl3flx/flx Zbtb46 cre mice exhibited decreased T. gondii-specific priming as revealed by both reduced cytokine production and reduced T. gondii-specific tetramer staining. In vitro differentiation of cDCs from bone marrow progenitors also revealed Bcl3-dependent cDC-specific antigen-presentation activity. Consistent with this, splenocyte single cell RNA seq (scRNAseq) in infected mice revealed Bcl3-dependent expression of genes involved in antigen processing in cDCs. We also identified by scRNAseq, a unique Bcl3-dependent hybrid subpopulation of Zbtb46+ DCs co-expressing the monocyte/macrophage transcription factor Lysozyme M. This subpopulation exhibited Bcl3-dependent expansion after infection. Likewise, by flow cytometry we identified two T. gondii-induced hybrid subpopulations of Bcl3-dependent cDC1 and cDC2 cells both expressing monocyte/macrophage markers, designated as icDC1 and icDC2. Together, our results indicate that Bcl3 in classical DCs is a major determinant of protective T cell responses and survival in T. gondii-infection.


Assuntos
Proteína 3 do Linfoma de Células B , Toxoplasma , Toxoplasmose , Animais , Camundongos , Linfócitos T CD8-Positivos , Células Dendríticas , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Toxoplasma/metabolismo , Toxoplasmose/metabolismo , Proteína 3 do Linfoma de Células B/metabolismo
5.
Sci Rep ; 11(1): 9123, 2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33907292

RESUMO

PAX2 is a transcription factor essential for kidney development and the main causative gene for renal coloboma syndrome (RCS). The mechanisms of PAX2 action during kidney development have been evaluated in mice but not in humans. This is a critical gap in knowledge since important differences have been reported in kidney development in the two species. In the present study, we hypothesized that key human PAX2-dependent kidney development genes are differentially expressed in nephron progenitor cells from induced pluripotent stem cells (iPSCs) in patients with RCS relative to healthy individuals. Cap analysis of gene expression revealed 189 candidate promoters and 71 candidate enhancers that were differentially activated by PAX2 in this system in three patients with RCS with PAX2 mutations. By comparing this list with the list of candidate Pax2-regulated mouse kidney development genes obtained from the Functional Annotation of the Mouse/Mammalian (FANTOM) database, we prioritized 17 genes. Furthermore, we ranked three genes-PBX1, POSTN, and ITGA9-as the top candidates based on closely aligned expression kinetics with PAX2 in the iPSC culture system and susceptibility to suppression by a Pax2 inhibitor in cultured mouse embryonic kidney explants. Identification of these genes may provide important information to clarify the pathogenesis of RCS, human kidney development, and kidney regeneration.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Rim/crescimento & desenvolvimento , Fator de Transcrição PAX2/genética , Adulto , Animais , Moléculas de Adesão Celular/genética , Linhagem da Célula , Coloboma/patologia , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas , Integrinas/genética , Rim/citologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Pessoa de Meia-Idade , Néfrons/citologia , Néfrons/fisiologia , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , Insuficiência Renal/patologia
6.
Immunol Cell Biol ; 99(6): 586-595, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33525048

RESUMO

Regulatory T cells (Tregs) exert inhibitory function under various physiological conditions and adopt diverse characteristics following environmental cues. Multiple subsets of Tregs expressing master transcription factors of helper T cells such as RORγt, T-bet, Gata3 and PPARγ have been characterized, but the molecular mechanism governing the differentiation of these subsets remains largely unknown. Here we report that the atypical IκB protein family member Bcl-3 suppresses RORγt+ Treg accumulation. The suppressive effect of Bcl-3 was particularly evident in the mouse immune tolerance model of anti-CD3 therapy. Using conditional knockout mice, we illustrate that loss of Bcl-3 specifically in Tregs was sufficient to boost RORγt+ Treg formation and resistance of mice to dextran sulfate sodium-induced colitis. We further demonstrate the suppressive effect of Bcl-3 on RORγt+ Treg differentiation in vitro. Our results reveal a novel role of nuclear factor-kappa B signaling pathways in Treg subset differentiation that may have clinical implications in immunotherapy.


Assuntos
Colite , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Animais , Proteína 3 do Linfoma de Células B , Diferenciação Celular , Colite/induzido quimicamente , Fatores de Transcrição Forkhead , Camundongos , Camundongos Endogâmicos C57BL , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Linfócitos T Reguladores , Células Th17
7.
Blood ; 137(22): 3050-3063, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-33512437

RESUMO

The extrafollicular immune response is essential to generate a rapid but transient wave of protective antibodies during infection. Despite its importance, the molecular mechanisms controlling this first response are poorly understood. Here, we demonstrate that enhanced Cxcr4 signaling caused by defective receptor desensitization leads to exacerbated extrafollicular B-cell response. Using a mouse model bearing a gain-of-function mutation of Cxcr4 described in 2 human hematologic disorders, warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome and Waldenström macroglobulinemia, we demonstrated that mutant B cells exhibited enhanced mechanistic target of rapamycin signaling, cycled more, and differentiated more potently into plasma cells than wild-type B cells after Toll-like receptor (TLR) stimulation. Moreover, Cxcr4 gain of function promoted enhanced homing and persistence of immature plasma cells in the bone marrow, a phenomenon recapitulated in WHIM syndrome patient samples. This translated in increased and more sustained production of antibodies after T-independent immunization in Cxcr4 mutant mice. Thus, our results establish that fine-tuning of Cxcr4 signaling is essential to limit the strength and length of the extrafollicular immune response.


Assuntos
Mutação com Ganho de Função , Doenças Hematológicas/imunologia , Plasmócitos/imunologia , Receptores CXCR4/imunologia , Transdução de Sinais/imunologia , Animais , Doenças Hematológicas/genética , Humanos , Camundongos , Camundongos Transgênicos , Receptores CXCR4/genética , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia
8.
PLoS Pathog ; 17(1): e1009249, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33508001

RESUMO

Bcl-3 is an atypical member of the IκB family that acts in the nucleus to modulate transcription of many NF-κB targets in a highly context-dependent manner. Accordingly, complete Bcl-3-/- mice have diverse defects in both innate and adaptive immune responses; however, direct effects of Bcl-3 action in individual immune cell types have not been clearly defined. Here, we document a cell-autonomous role for Bcl-3 in CD8+ T cell differentiation during the response to lymphocytic choriomeningitis virus infection. Single-cell RNA-seq and flow cytometric analysis of virus-specific Bcl3-/- CD8+ T cells revealed that differentiation was skewed towards terminal effector cells at the expense of memory precursor effector cells (MPECs). Accordingly, Bcl3-/- CD8+ T cells exhibited reduced memory cell formation and a defective recall response. Conversely, Bcl-3-overexpression in transgenic CD8+ T cells enhanced MPEC formation but reduced effector cell differentiation. Together, our results establish Bcl-3 as an autonomous determinant of memory/terminal effector cell balance during CD8+ T cell differentiation in response to acute viral infection. Our results provide proof-of-principle for targeting Bcl-3 pharmacologically to optimize adaptive immune responses to infectious agents, cancer cells, vaccines and other stimuli that induce CD8+ T cell differentiation.


Assuntos
Proteína 3 do Linfoma de Células B/metabolismo , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , NF-kappa B/imunologia , Animais , Proteína 3 do Linfoma de Células B/genética , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Transgênicos , Análise de Sequência de RNA , Análise de Célula Única
9.
J Immunol ; 204(9): 2464-2473, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32221037

RESUMO

Formyl peptide receptors (FPRs, mouse Fprs) belong to the G protein-coupled receptor superfamily and mediate phagocyte migration in response to bacteria- and host-derived chemoattractants; however, knowledge about their in vivo roles in bacterial pathogenesis is limited. In this study, we investigated the role of Fpr1 and Fpr2 in host defense against Escherichia coli infection. In vitro, we found that supernatants from E. coli cultures induced chemotaxis of wild-type (WT) mouse bone marrow-derived neutrophils and that the activity was significantly reduced in cells genetically deficient in either Fpr1 or Fpr2 and was almost absent in cells lacking both receptors. Consistent with this, E. coli supernatants induced chemotaxis and MAPK phosphorylation in HEK293 cells expressing either recombinant Fpr1 or Fpr2 but not untransfected parental cells. WT bone marrow -derived neutrophils could actively phagocytose and kill E. coli, whereas both activities were diminished in cells lacking Fpr1 or Fpr2; again, an additive effect was observed in cells lacking both receptors. In vivo, Fpr1 and Fpr2 deficiency resulted in reduced recruitment of neutrophils in the liver and peritoneal cavity of mice infected with inactivated E. coli Moreover, Fpr1-/- and Fpr2-/- mice had significantly increased mortality compared with WT mice after i.p. challenge with a virulent E. coli clinical isolate. These results indicate a critical role of Fprs in host defense against E. coli infection.


Assuntos
Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/metabolismo , Escherichia coli/imunologia , Receptores de Formil Peptídeo/imunologia , Receptores de Formil Peptídeo/metabolismo , Animais , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/microbiologia , Células Cultivadas , Quimiotaxia/imunologia , Células HEK293 , Humanos , Fígado/imunologia , Fígado/metabolismo , Fígado/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Cavidade Peritoneal/microbiologia , Fagocitose/imunologia , Fosforilação/imunologia
10.
JCI Insight ; 4(24)2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31687976

RESUMO

Warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome immunodeficiency is caused by autosomal dominant gain-of-function mutations in chemokine receptor CXCR4. Patient WHIM-09 was spontaneously cured by chromothriptic deletion of 1 copy of 164 genes, including the CXCR4WHIM allele, presumably in a single hematopoietic stem cell (HSC) that repopulated HSCs and the myeloid lineage. Testing the specific contribution of CXCR4 hemizygosity to her cure, we previously demonstrated enhanced engraftment of Cxcr4+/o HSCs after transplantation in WHIM (Cxcr4+/w) model mice, but the potency was not quantitated. We now report graded-dose competitive transplantation experiments using lethally irradiated Cxcr4+/+ recipients in which mixed BM cells containing approximately 5 Cxcr4+/o HSCs and a 100-fold excess of Cxcr4+/w HSCs achieved durable 50% Cxcr4+/o myeloid and B cell chimerism in blood and approximately 20% Cxcr4+/o HSC chimerism in BM. In Cxcr4+/o/Cxcr4+/w parabiotic mice, we observed 80%-100% Cxcr4+/o myeloid and lymphoid chimerism in the blood and 15% Cxcr4+/o HSC chimerism in BM from the Cxcr4+/w parabiont, which was durable after separation from the Cxcr4+/o parabiont. Thus, CXCR4 haploinsufficiency likely significantly contributed to the selective repopulation of HSCs and the myeloid lineage from a single chromothriptic HSC in WHIM-09. Moreover, the results suggest that WHIM allele silencing of patient HSCs is a viable gene therapy strategy.


Assuntos
Haploinsuficiência , Transplante de Células-Tronco Hematopoéticas , Leucopenia/terapia , Doenças da Imunodeficiência Primária/terapia , Receptores CXCR4/genética , Verrugas/terapia , Animais , Cromotripsia , Modelos Animais de Doenças , Feminino , Mutação com Ganho de Função , Terapia Genética/métodos , Humanos , Leucopenia/genética , Masculino , Camundongos , Doenças da Imunodeficiência Primária/complicações , Doenças da Imunodeficiência Primária/genética , Quimeras de Transplante , Verrugas/complicações , Verrugas/genética
11.
J Clin Immunol ; 39(6): 532-556, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31313072

RESUMO

WHIM syndrome is a rare combined primary immunodeficiency disease named by acronym for the diagnostic tetrad of warts, hypogammaglobulinemia, infections, and myelokathexis. Myelokathexis is a unique form of non-cyclic severe congenital neutropenia caused by accumulation of mature and degenerating neutrophils in the bone marrow; monocytopenia and lymphopenia, especially B lymphopenia, also commonly occur. WHIM syndrome is usually caused by autosomal dominant mutations in the G protein-coupled chemokine receptor CXCR4 that impair desensitization, resulting in enhanced and prolonged G protein- and ß-arrestin-dependent responses. Accordingly, CXCR4 antagonists have shown promise as mechanism-based treatments in phase 1 clinical trials. This review is based on analysis of all 105 published cases of WHIM syndrome and covers current concepts, recent advances, unresolved enigmas and controversies, and promising future research directions.


Assuntos
Doenças da Imunodeficiência Primária/diagnóstico , Doenças da Imunodeficiência Primária/etiologia , Doenças da Imunodeficiência Primária/terapia , Verrugas/diagnóstico , Verrugas/etiologia , Verrugas/terapia , Imunidade Adaptativa , Alelos , Terapia Combinada , Diagnóstico Diferencial , Gerenciamento Clínico , Suscetibilidade a Doenças/imunologia , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Imunidade Inata , Mutação , Fenótipo , Medicina de Precisão/métodos , Doenças da Imunodeficiência Primária/epidemiologia , Verrugas/epidemiologia
13.
Nat Commun ; 10(1): 616, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30728353

RESUMO

Hematopoietic chimerism after allogeneic bone marrow transplantation may establish a state of donor antigen-specific tolerance. However, current allotransplantation protocols involve genotoxic conditioning which has harmful side-effects and predisposes to infection and cancer. Here we describe a non-genotoxic conditioning protocol for fully MHC-mismatched bone marrow allotransplantation in mice involving transient immunosuppression and selective depletion of recipient hematopoietic stem cells with a CD117-antibody-drug-conjugate (ADC). This protocol resulted in multilineage, high level (up to 50%), durable, donor-derived hematopoietic chimerism after transplantation of 20 million total bone marrow cells, compared with ≤ 2.1% hematopoietic chimerism from 50 million total bone marrow cells without conditioning. Moreover, long-term survival of bone marrow donor-type but not third party skin allografts is achieved in CD117-ADC-conditioned chimeric mice without chronic immunosuppression. The only observed adverse event is transient elevation of liver enzymes in the first week after conditioning. These results provide proof-of-principle for CD117-ADC as a non-genotoxic, highly-targeted conditioning agent in allotransplantation and tolerance protocols.


Assuntos
Transplante de Medula Óssea/métodos , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas/métodos , Imunoconjugados/farmacologia , Proteínas Proto-Oncogênicas c-kit/imunologia , Tolerância ao Transplante/efeitos dos fármacos , Animais , Células da Medula Óssea/efeitos dos fármacos , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Células-Tronco Hematopoéticas , Tolerância Imunológica , Terapia de Imunossupressão/métodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Animais , Pele/patologia , Transplante de Pele/métodos , Quimeras de Transplante , Transplante Homólogo
14.
N Engl J Med ; 380(2): 163-170, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30625055

RESUMO

WHIM syndrome (warts, hypogammaglobulinemia, infections, and myelokathexis), a primary immunodeficiency disorder involving panleukopenia, is caused by autosomal dominant gain-of-function mutations in CXC chemokine receptor 4 (CXCR4). Myelokathexis is neutropenia caused by neutrophil retention in bone marrow. Patients with WHIM syndrome are often treated with granulocyte colony-stimulating factor (G-CSF), which can increase neutrophil counts but does not affect cytopenias other than neutropenia. In this investigator-initiated, open-label study, three severely affected patients with WHIM syndrome who could not receive G-CSF were treated with low-dose plerixafor, a CXCR4 antagonist, for 19 to 52 months. Myelofibrosis, panleukopenia, anemia, and thrombocytopenia were ameliorated, the wart burden and frequency of infection declined, human papillomavirus-associated oropharyngeal squamous-cell carcinoma stabilized, and quality of life improved markedly. Adverse events were mainly infections attributable to the underlying immunodeficiency. One patient died from complications of elective reconstructive surgery. (Funded by the National Institutes of Health.).


Assuntos
Medula Óssea/patologia , Compostos Heterocíclicos/uso terapêutico , Síndromes de Imunodeficiência/tratamento farmacológico , Receptores CXCR4/antagonistas & inibidores , Verrugas/tratamento farmacológico , Benzilaminas , Exame de Medula Óssea , Ciclamos , Evolução Fatal , Humanos , Síndromes de Imunodeficiência/patologia , Masculino , Pessoa de Meia-Idade , Neoplasias de Células Escamosas/tratamento farmacológico , Neoplasias de Células Escamosas/genética , Fenótipo , Doenças da Imunodeficiência Primária , Mielofibrose Primária/tratamento farmacológico , Mielofibrose Primária/patologia , Receptores CXCR4/genética , Verrugas/patologia
15.
Immunol Rev ; 287(1): 91-102, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30565238

RESUMO

WHIM syndrome is a rare, autosomal dominant immunodeficiency which is named for the four key manifestations: Warts, Hypogammaglobulinemia, Infections, and Myelokathexis. It results from heterozygous gain-of-function mutations in the chemokine receptor CXCR4 which is widely expressed on leukocytes and has profound influences on immune system homeostasis and organogenesis. New treatments for the disease using drugs to reduce CXCR4 function are excellent examples of precision medicine. Since CXCR4 and its ligand CXCL12 play an important role in a variety of infectious, inflammatory, autoimmune, and malignant diseases, the study of WHIM syndrome provides important insights into both the physiologic and disease roles of these molecules.


Assuntos
Síndromes de Imunodeficiência/imunologia , Papillomaviridae/fisiologia , Infecções por Papillomavirus/imunologia , Receptores CXCR4/genética , Verrugas/imunologia , Agamaglobulinemia , Animais , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/terapia , Infecções , Leucopenia , Terapia de Alvo Molecular , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/terapia , Medicina de Precisão , Doenças da Imunodeficiência Primária , Verrugas/genética , Verrugas/terapia
16.
Viruses ; 12(1)2019 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-31892128

RESUMO

Many viruses initiate interaction with target cells by binding to cell surface glycosaminoglycans (GAGs). Heparan sulfate (HS) appears to be particularly important in fibroblasts, epithelial cells and endothelial cells, where it represents the dominant GAG. How GAGs influence viral infectivity in HS-poor target cells such as macrophages has not been clearly defined. Here, we show that mouse cytomegalovirus (MCMV) targets HS in susceptible fibroblasts and cultured salivary gland acinar cells (SGACs), but not in macrophage cell lines and primary bone marrow-derived macrophages, where chondroitin sulfate was the dominant virus-binding GAG. MCK-2, an MCMV-encoded GAG-binding chemokine that promotes infection of macrophages as part of a gH/gL/MCK-2 entry complex, was dispensable for MCMV attachment to the cell surface and for direct infection of SGACs. Thus, MCMV tropism for target cells is markedly influenced by differential GAG expression, suggesting that the specificity of anti-GAG peptides now under development as HCMV therapeutics may need to be broadened for effective application as anti-viral agents.


Assuntos
Quimiocinas CC/imunologia , Sulfatos de Condroitina/metabolismo , Interações entre Hospedeiro e Microrganismos , Macrófagos/virologia , Muromegalovirus/fisiologia , Proteínas Virais/imunologia , Internalização do Vírus , Animais , Linhagem Celular , Quimiocinas CC/genética , Fibroblastos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Células RAW 264.7 , Proteínas Virais/genética , Proteínas Virais/metabolismo , Tropismo Viral
17.
Int J Mol Sci ; 20(1)2018 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-30577453

RESUMO

Cysteine-X-cysteine chemokine receptor 4 (CXCR4) is a broadly expressed and multifunctional G protein-coupled chemokine receptor critical for organogenesis, hematopoiesis, and antimicrobial host defense. In the hematopoietic system, the binding of CXCR4 to its cognate chemokine ligand, CXCL12, mediates leukocyte trafficking, distribution, survival, activation, and proliferation. Warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome is a rare, autosomal dominant, combined immunodeficiency disorder caused by mutations in the C-terminus of CXCR4 that prevent receptor downregulation and therefore result in pathologically increased signaling. The "M" in the acronym WHIM refers to myelokathexis, the retention of neutrophils in the bone marrow resulting in neutropenia, which explains in part the increased susceptibility to bacterial infection. However, WHIM patients also present with B and T lymphopenia, which may explain the susceptibility to human papillomavirus (HPV), the cause of warts. The impact of WHIM mutations on lymphocytes and adaptive immunity has received less attention than myelokathexis and is the focus of this review.


Assuntos
Imunidade Adaptativa , Síndromes de Imunodeficiência/diagnóstico , Síndromes de Imunodeficiência/etiologia , Verrugas/diagnóstico , Verrugas/etiologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Biomarcadores , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Gerenciamento Clínico , Humanos , Síndromes de Imunodeficiência/metabolismo , Síndromes de Imunodeficiência/terapia , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/virologia , Doenças da Imunodeficiência Primária , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Verrugas/metabolismo , Verrugas/terapia
18.
mSphere ; 3(6)2018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30541782

RESUMO

Several immunodeficiencies are associated with high susceptibility to persistent and progressive human papillomavirus (HPV) infection leading to a wide range of cutaneous and mucosal lesions. However, the HPV types most commonly associated with such clinical manifestations in these patients have not been systematically defined. Here, we used virion enrichment, rolling circle amplification, and deep sequencing to identify circular DNA viruses present in skin swabs and/or wart biopsy samples from 48 patients with rare genetic immunodeficiencies, including patients with warts, hypogammaglobulinemia, infections, myelokathexis (WHIM) syndrome, or epidermodysplasia verruciformis (EV). Their profiles were compared with the profiles of swabs from 14 healthy adults and warts from 6 immunologically normal children. Individual patients were typically infected with multiple HPV types; up to 26 different types were isolated from a single patient (multiple anatomical sites, one time point). Among these, we identified the complete genomes of 83 previously unknown HPV types and 35 incomplete genomes representing possible additional new types. HPV types in the genus Gammapapillomavirus were common in WHIM patients, whereas EV patients mainly shed HPVs from the genus Betapapillomavirus. Preliminary evidence based on three WHIM patients treated with plerixafor, a leukocyte mobilizing agent, suggest that longer-term therapy may correlate with decreased HPV diversity and increased predominance of HPV types associated with childhood skin warts.IMPORTANCE Although some members of the viral family Papillomaviridae cause benign skin warts (papillomas), many human papillomavirus (HPV) infections are not associated with visible symptoms. For example, most healthy adults chronically shed Gammapapillomavirus (Gamma) virions from apparently healthy skin surfaces. To further explore the diversity of papillomaviruses, we performed viromic surveys on immunodeficient individuals suffering from florid skin warts. Our results nearly double the number of known Gamma HPV types and suggest that WHIM syndrome patients are uniquely susceptible to Gamma HPV-associated skin warts. Preliminary results suggest that treatment with the drug plerixafor may promote resolution of the unusual Gamma HPV skin warts observed in WHIM patients.


Assuntos
DNA Viral/genética , Síndromes de Imunodeficiência/complicações , Papillomaviridae/classificação , Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/virologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , DNA Viral/química , Feminino , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Metagenômica , Pessoa de Meia-Idade , Mucosa/virologia , Técnicas de Amplificação de Ácido Nucleico , Papillomaviridae/genética , Pele/virologia , Adulto Jovem
19.
J Clin Invest ; 128(8): 3312-3318, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29715199

RESUMO

For gene therapy of gain-of-function autosomal dominant diseases, either correcting or deleting the disease allele is potentially curative. To test whether there may be an advantage of one approach over the other for WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome - a primary immunodeficiency disorder caused by gain-of-function autosomal dominant mutations in chemokine receptor CXCR4 - we performed competitive transplantation experiments using both lethally irradiated WT (Cxcr4+/+) and unconditioned WHIM (Cxcr4+/w) recipient mice. In both models, hematopoietic reconstitution was markedly superior using BM cells from donors hemizygous for Cxcr4 (Cxcr4+/o) compared with BM cells from Cxcr4+/+ donors. Remarkably, only approximately 6% Cxcr4+/o hematopoietic stem cell (HSC) chimerism after transplantation in unconditioned Cxcr4+/w recipient BM supported more than 70% long-term donor myeloid chimerism in blood and corrected myeloid cell deficiency in blood. Donor Cxcr4+/o HSCs differentiated normally and did not undergo exhaustion as late as 465 days after transplantation. Thus, disease allele deletion resulting in Cxcr4 haploinsufficiency was superior to disease allele repair in a mouse model of gene therapy for WHIM syndrome, allowing correction of leukopenia without recipient conditioning.


Assuntos
Transplante de Medula Óssea , Haploinsuficiência , Síndromes de Imunodeficiência , Leucopenia , Receptores CXCR4 , Quimeras de Transplante , Verrugas , Aloenxertos , Animais , Modelos Animais de Doenças , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/metabolismo , Síndromes de Imunodeficiência/patologia , Síndromes de Imunodeficiência/terapia , Leucopenia/genética , Leucopenia/metabolismo , Leucopenia/patologia , Leucopenia/terapia , Camundongos , Camundongos Mutantes , Doenças da Imunodeficiência Primária , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Quimeras de Transplante/genética , Quimeras de Transplante/metabolismo , Verrugas/genética , Verrugas/metabolismo , Verrugas/patologia , Verrugas/terapia
20.
Cytokine ; 109: 2-10, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29398278

RESUMO

Chemokines are named and best known for their chemotactic cytokine activity in the hematopoietic system; however, their importance extends far beyond leukocytes, cell movement and immunoregulation. CXCL12, the most protean of chemokines, regulates development in multiple systems, including the hematopoietic, cardiovascular and nervous systems, and regulates diverse cell functions, including differentiation, distribution, activation, immune synapse formation, effector function, proliferation and survival in the immune system alone. The broad importance of CXCL12 is revealed by the complex lethal developmental phenotypes in mice lacking either Cxcl12 or either one of its two known 7-transmembrane domain receptors Cxcr4 and Ackr3, as well as by gain-of-function mutations in human CXCR4, which cause WHIM syndrome, a multisystem and combined immunodeficiency disease and the only Mendelian condition caused by a chemokine system mutation. In addition, wild type CXCR4 is important in the pathogenesis of HIV/AIDS and cancer. Thus, CXCL12 and its receptors CXCR4 and ACKR3 provide extraordinary examples of multisystem multitasking in the chemokine system in both health and disease.


Assuntos
Movimento Celular/imunologia , Quimiocina CXCL12/metabolismo , Leucócitos/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Síndrome da Imunodeficiência Adquirida/patologia , Animais , HIV-1/genética , HIV-1/patogenicidade , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/patologia , Camundongos , Doenças da Imunodeficiência Primária , Transdução de Sinais/imunologia , Verrugas/genética , Verrugas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA