Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
J Clin Periodontol ; 51(4): 464-473, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38185798

RESUMEN

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.


Asunto(s)
Síndromes de Inmunodeficiencia , Neutropenia , Enfermedades Periodontales , Periodontitis , Enfermedades de Inmunodeficiencia Primaria , Verrugas , Adulto , Humanos , Animales , Ratones , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/complicaciones , Enfermedades de Inmunodeficiencia Primaria/genética , Verrugas/genética , Verrugas/terapia , Neutropenia/complicaciones , Neutropenia/genética , Enfermedades Periodontales/complicaciones , Enfermedades Periodontales/genética , Periodontitis/complicaciones , Periodontitis/genética
2.
J Immunol ; 210(12): 1913-1924, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37133343

RESUMEN

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.


Asunto(s)
Agammaglobulinemia , Compuestos Heterocíclicos , Síndromes de Inmunodeficiencia , Linfopenia , Neutropenia , Humanos , Animales , Ratones , Síndromes de Inmunodeficiencia/genética , Movilización de Célula Madre Hematopoyética/efectos adversos , Agammaglobulinemia/complicaciones , Agammaglobulinemia/genética , Neutropenia/genética , Linfocitos T CD8-positivos , Receptores CXCR4/genética
3.
Blood ; 142(1): 23-32, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-36928087

RESUMEN

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.


Asunto(s)
Síndromes de Inmunodeficiencia , Verrugas , Ratones , Animales , Alelos , Sistemas CRISPR-Cas , ARN Guía de Sistemas CRISPR-Cas , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/terapia , Verrugas/genética , Verrugas/terapia , Terapia Genética , Receptores CXCR4/genética
4.
Immunohorizons ; 6(7): 543-558, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35882421

RESUMEN

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.


Asunto(s)
Agammaglobulinemia , Linfopenia , Neutropenia , Verrugas , Agammaglobulinemia/genética , Animales , Endotoxinas/uso terapéutico , Lipopolisacáridos , Ratones , Neutropenia/genética , Enfermedades de Inmunodeficiencia Primaria , Verrugas/tratamiento farmacológico , Verrugas/genética
5.
Dev Biol ; 477: 70-84, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34015362

RESUMEN

The C-X-C chemokine receptor CXCR4 and its ligand CXCL12 play an important role in organ-specific vascular branching morphogenesis. CXCR4 is preferentially expressed by arterial endothelial cells, and local secretion of CXCL12 determines the organotypic pattern of CXCR4+ arterial branching. Previous loss-of-function studies clearly demonstrated that CXCL12-CXCR4 signaling is necessary for proper arterial branching in the developing organs such as the skin and heart. To further understand the role of CXCL12-CXCR4 signaling in organ-specific vascular development, we generated a mouse model carrying the Cre recombinase-inducible Cxcr4 transgene. Endothelial cell-specific Cxcr4 gain-of-function embryos exhibited defective vascular remodeling and formation of a hierarchical vascular branching network in the developing skin and heart. Ectopic expression of CXCR4 in venous endothelial cells, but not in lymphatic endothelial cells, caused blood-filled, enlarged lymphatic vascular phenotypes, accompanied by edema. These data suggest that CXCR4 expression is tightly regulated in endothelial cells for appropriate vascular development in an organ-specific manner.


Asunto(s)
Vasos Sanguíneos/embriología , Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Receptores CXCR4/fisiología , Animales , Vasos Sanguíneos/anatomía & histología , Células Endoteliales/metabolismo , Mutación con Ganancia de Función , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Receptores CXCR4/biosíntesis , Remodelación Vascular/fisiología
6.
FASEB J ; 35(2): e21315, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33538366

RESUMEN

Cataracts are a common consequence of aging; however, pathogenesis remains poorly understood. Here, we observed that after 3 months of age mice lacking the G protein-coupled leukocyte chemotactic receptor Fpr1 (N-formyl peptide receptor 1) began to develop bilateral posterior subcapsular cataracts that progressed to lens rupture and severe degeneration, without evidence of either systemic or local ocular infection or inflammation. Consistent with this, Fpr1 was detected in both mouse and human lens in primary lens epithelial cells (LECs), the only cell type present in the lens; however, expression was confined to subcapsular LECs located along the anterior hemispheric surface. To maximize translucency, LECs at the equator proliferate and migrate posteriorly, then differentiate into lens fiber cells by nonclassical apoptotic signaling, which results in loss of nuclei and other organelles, including mitochondria which are a rich source of endogenous N-formyl peptides. In this regard, denucleation and posterior migration of LECs were abnormal in lenses from Fpr1-/- mice, and direct stimulation of LECs with the prototypic N-formyl peptide agonist fMLF promoted apoptosis. Thus, Fpr1 is repurposed beyond its immunoregulatory role in leukocytes to protect against cataract formation and lens degeneration during aging.


Asunto(s)
Envejecimiento/metabolismo , Envejecimiento/patología , Catarata/metabolismo , Receptores de Formil Péptido/metabolismo , Animales , Catarata/patología , Diferenciación Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Citometría de Flujo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Receptores de Formil Péptido/genética , Ultrasonografía
7.
J Immunol ; 204(9): 2464-2473, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32221037

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/metabolismo , Escherichia coli/inmunología , Receptores de Formil Péptido/inmunología , Receptores de Formil Péptido/metabolismo , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/microbiología , Células Cultivadas , Quimiotaxis/inmunología , Células HEK293 , Humanos , Hígado/inmunología , Hígado/metabolismo , Hígado/microbiología , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/inmunología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Cavidad Peritoneal/microbiología , Fagocitosis/inmunología , Fosforilación/inmunología
8.
Crit Care Med ; 48(2): e123-e132, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31939811

RESUMEN

OBJECTIVES: Trauma predisposes to systemic sterile inflammation (systemic inflammatory response syndrome) as well as infection, but the mechanisms linking injury to infection are poorly understood. Mitochondrial debris contains formyl peptides. These bind formyl peptide receptor-1, trafficking neutrophils to wounds, initiating systemic inflammatory response syndrome, and wound healing. Bacterial formyl peptides, however, also attract neutrophils via formyl peptide receptor-1. Thus, mitochondrial formyl peptides might suppress neutrophils antimicrobial function. Also, formyl peptide receptor-1 blockade used to mitigate systemic inflammatory response syndrome might predispose to sepsis. We examined how mitochondrial formyl peptides impact neutrophils functions contributing to antimicrobial responses and how formyl peptide receptor-1 antagonists affect those functions. DESIGN: Prospective study of human and murine neutrophils and clinical cohort analysis. SETTING: University research laboratory and level 1 trauma center. PATIENTS: Trauma patients, volunteer controls. ANIMAL SUBJECTS: C57Bl/6, formyl peptide receptor-1, and formyl peptide receptor-2 knockout mice. INTERVENTIONS: Human and murine neutrophils functions were activated with autologous mitochondrial debris, mitochondrial formyl peptides, or bacterial formyl peptides followed by chemokines or leukotrienes. The experiments were repeated using formyl peptide receptor-1 antagonist cyclosporin H, "designer" human formyl peptide receptor-1 antagonists (POL7178 and POL7200), or anti-formyl peptide receptor-1 antibodies. Mouse injury/lung infection model was used to evaluate effect of formyl peptide receptor-1 inhibition. MEASUREMENTS AND MAIN RESULTS: Human neutrophils cytosolic calcium, chemotaxis, reactive oxygen species production, and phagocytosis were studied before and after exposure to mitochondrial debris, mitochondrial formyl peptides, and bacterial formyl peptides. Mitochondrial formyl peptide and bacterial formyl peptides had similar effects on neutrophils. Responses to chemokines and leukotrienes were suppressed by prior exposure to formyl peptides. POL7200 and POL7178 were specific antagonists of human formyl peptide receptor-1 and more effective than cyclosporin H or anti-formyl peptide receptor-1 antibodies. Formyl peptides inhibited mouse neutrophils responses to chemokines only if formyl peptide receptor-1 was present. Formyl peptide receptor-1 blockade did not inhibit neutrophils bacterial phagocytosis or reactive oxygen species production. Cyclosporin H increased bacterial clearance in lungs after injury. CONCLUSIONS: Formyl peptides both activate and desensitize neutrophils. Formyl peptide receptor-1 blockade prevents desensitization, potentially both diminishing systemic inflammatory response syndrome and protecting the host against secondary infection after tissue trauma or primary infection.


Asunto(s)
Proteínas Mitocondriales/inmunología , Activación Neutrófila/inmunología , Receptores de Formil Péptido/antagonistas & inhibidores , Animales , Ciclosporina/farmacología , Humanos , Lesión Pulmonar/fisiopatología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Infecciones del Sistema Respiratorio/fisiopatología
9.
JCI Insight ; 4(24)2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31687976

RESUMEN

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.


Asunto(s)
Haploinsuficiencia , Trasplante de Células Madre Hematopoyéticas , Leucopenia/terapia , Enfermedades de Inmunodeficiencia Primaria/terapia , Receptores CXCR4/genética , Verrugas/terapia , Animales , Cromotripsis , Modelos Animales de Enfermedad , Femenino , Mutación con Ganancia de Función , Terapia Genética/métodos , Humanos , Leucopenia/genética , Masculino , Ratones , Enfermedades de Inmunodeficiencia Primaria/complicaciones , Enfermedades de Inmunodeficiencia Primaria/genética , Quimera por Trasplante , Verrugas/complicaciones , Verrugas/genética
10.
J Clin Immunol ; 39(6): 532-556, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31313072

RESUMEN

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.


Asunto(s)
Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Enfermedades de Inmunodeficiencia Primaria/etiología , Enfermedades de Inmunodeficiencia Primaria/terapia , Verrugas/diagnóstico , Verrugas/etiología , Verrugas/terapia , Inmunidad Adaptativa , Alelos , Terapia Combinada , Diagnóstico Diferencial , Manejo de la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Inmunidad Innata , Mutación , Fenotipo , Medicina de Precisión/métodos , Enfermedades de Inmunodeficiencia Primaria/epidemiología , Verrugas/epidemiología
11.
J Clin Invest ; 128(8): 3312-3318, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29715199

RESUMEN

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.


Asunto(s)
Trasplante de Médula Ósea , Haploinsuficiencia , Síndromes de Inmunodeficiencia , Leucopenia , Receptores CXCR4 , Quimera por Trasplante , Verrugas , Aloinjertos , Animales , Modelos Animales de Enfermedad , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Síndromes de Inmunodeficiencia/terapia , Leucopenia/genética , Leucopenia/metabolismo , Leucopenia/patología , Leucopenia/terapia , Ratones , Ratones Mutantes , Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Quimera por Trasplante/genética , Quimera por Trasplante/metabolismo , Verrugas/genética , Verrugas/metabolismo , Verrugas/patología , Verrugas/terapia
12.
J Clin Immunol ; 38(1): 77-87, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29177911

RESUMEN

WHIM-09 is the first patient described with WHIM syndrome, an autosomal dominant form of neutropenia related to bone marrow retention of neutrophils. Originally diagnosed incorrectly with autoimmune neutropenia, the patient underwent splenectomy at age 9, but the absolute neutrophil count (ANC) did not rise. Subsequently, she was spontaneously cured by chromothripsis (chromosome shattering), which deleted the disease allele CXCR4 R334X , and 163 other genes, on chromosome 2 in a single hematopoietic stem cell (HSC). Chromothriptic CXCR4 +/o HSCs replaced CXCR4 +/R334X WHIM HSCs, and the ANC rose to a new sustained and benign baseline ~ 2-3-fold above normal that had remained unexplained. Here, we show that splenectomized Cxcr4 +/o mice had sustained and benign neutrophilia, phenocopying neutrophilia in WHIM-09. In addition, WHIM-09's granulocyte-macrophage precursor cells possessed increased granulocyte colony-forming activity ex vivo. Thus, WHIM-09's neutrophilia may be multifactorial, involving neutrophil-extrinsic factors (splenectomy), as well as CXCR4 haploinsufficiency-dependent neutrophil-intrinsic factors (increased myeloid precursor cell differentiation). The strong bone marrow retention signal for neutrophils conferred by the WHIM mutation may have prevented neutrophilia after splenectomy until the mutation was deleted by chromothripsis.


Asunto(s)
Cromotripsis , Células Madre Hematopoyéticas/fisiología , Síndromes de Inmunodeficiencia/diagnóstico , Mutación/genética , Neutrófilos/fisiología , Receptores CXCR4/genética , Verrugas/diagnóstico , Alelos , Animales , Diferenciación Celular/genética , Niño , Modelos Animales de Enfermedad , Femenino , Haploinsuficiencia , Humanos , Síndromes de Inmunodeficiencia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades de Inmunodeficiencia Primaria , Esplenectomía , Verrugas/genética
13.
Expert Opin Orphan Drugs ; 5(10): 813-825, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29057173

RESUMEN

21 INTRODUCTION: WHIM syndrome is a rare combined primary immunodeficiency disorder caused by autosomal dominant gain-of-function mutations in the chemokine receptor CXCR4. It is the only Mendelian condition known to be caused by mutation of a chemokine or chemokine receptor. As such, it provides a scientific opportunity to understand chemokine-dependent immunoregulation in humans and a medical opportunity to develop mechanism-based treatment and cure strategies. 22 AREAS COVERED: This review covers the clinical features, genetics, immunopathogenesis and clinical management of WHIM syndrome. Clinical trials of targeted therapeutic agents and potential cure strategies are also included. 23 EXPERT OPINION: WHIM syndrome may be particularly amenable to mechanism-based therapeutics for three reasons: 1) CXCR4 has been validated as the molecular target in the disease by Mendelian genetics; 2) the biochemical abnormality is excessive CXCR4 signaling; and 3) antagonists selective for CXCR4 have been developed. Plerixafor is FDA-approved for hematopoietic stem cell (HSC) mobilization and has shown preliminary safety and efficacy in phase I clinical trials in WHIM syndrome. Gene editing may represent a viable cure strategy, since chromothriptic deletion of the disease allele in HSCs resulted in clinical cure of a patient and because CXCR4 haploinsufficiency enhances engraftment of transplanted HSCs in mice.

14.
Sci Rep ; 7: 44816, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28317879

RESUMEN

Agonists that target the A1, A2A, A2B and A3 adenosine receptors have potential to be potent treatment options for a number of diseases, including autoimmune diseases, cardiovascular disease and cancer. Because each of these adenosine receptors plays a distinct role throughout the body, obtaining highly specific receptor agonists is essential. Of these receptors, the adenosine A2AR and A2BR share many sequence and structural similarities but highly differ in their responses to inflammatory stimuli. Our laboratory, using a combination of specially developed cell lines and calcium release analysis hardware, has created a new and faster method for determining specificity of synthetic adenosine agonist compounds for the A2A and A2B receptors in human cells. A2A receptor expression was effectively removed from K562 cells, resulting in the development of a distinct null line. Using HIV-lentivector and plasmid DNA transfection, we also developed A2A and A2B receptor over-expressing lines. As adenosine is known to cause changes in intracellular calcium levels upon addition to cell culture, calcium release can be determined in these cell lines upon compound addition, providing a functional readout of receptor activation and allowing us to isolate the most specific adenosine agonist compounds.


Asunto(s)
Descubrimiento de Drogas/métodos , Agonistas del Receptor Purinérgico P1/química , Agonistas del Receptor Purinérgico P1/farmacología , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Adenosina/metabolismo , Sistemas CRISPR-Cas , Calcio/metabolismo , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Citometría de Flujo , Expresión Génica , Técnicas de Inactivación de Genes , Marcación de Gen , Humanos , Células K562 , Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/clasificación
15.
Sci Transl Med ; 8(322): 322ra10, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26791948

RESUMEN

Systemic Candida albicans infection causes high morbidity and mortality and is now the leading cause of nosocomial bloodstream infection in the United States. Neutropenia is a major risk factor for poor outcome in infected patients; however, the molecular factors that mediate neutrophil trafficking and effector function during infection are poorly defined. Using a mouse model of systemic candidiasis, we found that the neutrophil-selective CXC chemokine receptor Cxcr1 and its ligand, Cxcl5, are highly induced in the Candida-infected kidney, the target organ in the model. To investigate the role of Cxcr1 in antifungal host defense in vivo, we generated Cxcr1(-/-) mice and analyzed their immune response to Candida. Mice lacking Cxcr1 exhibited decreased survival with enhanced Candida growth in the kidney and renal failure. Increased susceptibility of Cxcr1(-/-) mice to systemic candidiasis was not due to impaired neutrophil trafficking from the blood into the infected kidney but was the result of defective killing of the fungus by neutrophils that exhibited a cell-intrinsic decrease in degranulation. In humans, the mutant CXCR1 allele CXCR1-T276 results in impaired neutrophil degranulation and fungal killing and was associated with increased risk of disseminated candidiasis in infected patients. Together, our data demonstrate a biological function for mouse Cxcr1 in vivo and indicate that CXCR1-dependent neutrophil effector function is a critical innate protective mechanism of fungal clearance and host survival in systemic candidiasis.


Asunto(s)
Candida/fisiología , Candidiasis/microbiología , Degranulación de la Célula , Interacciones Huésped-Patógeno , Viabilidad Microbiana , Neutrófilos/fisiología , Receptores de Interleucina-8A/metabolismo , Alelos , Animales , Candida/crecimiento & desarrollo , Candidiasis/sangre , Candidiasis/inmunología , Candidiasis/patología , Quimiocina CXCL5/metabolismo , Modelos Animales de Enfermedad , Humanos , Hifa/fisiología , Riñón/microbiología , Riñón/patología , Ligandos , Ratones , Proteínas Mutantes/metabolismo , Polimorfismo de Nucleótido Simple/genética , Receptores de Interleucina-8A/deficiencia , Receptores de Interleucina-8A/genética , Análisis de Supervivencia , Donantes de Tejidos
16.
Rare Dis ; 3(1): e1073430, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26459672

RESUMEN

We recently reported a 59 year old female, designated WHIM-09, who was born with the rare immunodeficiency disease WHIM syndrome but underwent spontaneous phenotypic reversion as an adult. The causative WHIM mutation CXCR4 (R334X) was absent in her myeloid and erythroid lineage, but present in her lymphoid lineage and in epithelial cells, defining her as a somatic genetic mosaic. Genomic and hematologic analysis revealed chromothripsis (chromosome shattering) on one copy of chromosome 2, which deleted 164 genes including CXCR4 (R334X) in a single haematopoietic stem cell (HSC) (Fig. 1). Experiments in mice indicated that deleting one copy of Cxcr4 is sufficient to confer a selective advantage for engraftment of transplanted HSCs, suggesting a mechanism for clinical cure in WHIM-09. Genome editing may allow autologous transplantation of HSCs lacking one copy of CXCR4 without bone marrow conditioning as a general cure strategy in WHIM syndrome, safely recapitulating the outcome in patient WHIM-09. Figure 1.Chromothripsis (chromosomal shattering) resulted in clinical cure of a patient with a rare immunodeficiency (WHIM syndrome) by deleting the mutant copy of CXCR4.

17.
Eur J Immunol ; 45(6): 1855-67, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25801950

RESUMEN

AMD3100 (plerixafor), is a specific CXCR4 antagonist approved by the FDA for mobilizing hematopoietic stem cells from bone marrow to blood for transplantation in cancer. AMD3100 also mobilizes most mature leukocyte subsets to blood; however, their source and trafficking potential have not been fully delineated. Here, we show that a single injection of AMD3100 10 mg/kg into C57Bl/6 mice rapidly mobilizes (peak ∼ 2.5 h) the same leukocyte subsets to blood as in humans. Using this model, we found that AMD3100 mobilization of neutrophils, lymphocytes, and monocytes to blood is not reduced by splenectomy or by blockade of lymphocyte egress from lymph node with FTY720, but is coupled to (i) reduced content of each of these cell types in the bone marrow; (ii) reduced T-cell numbers in thymuses; (iii) increased lymphocytes in lymph nodes; and (iv) increased neutrophil and monocyte content in the lung. Direct intrathymic labeling showed that AMD3100 selectively mobilizes naïve thymic CD4(+) and CD8(+) T cells to blood. Finally, AMD3100-induced neutrophil mobilization to blood did not reduce neutrophil trafficking to thioglycollate-inflamed peritoneum. Thus, AMD3100 redistributes lymphocytes, monocytes, and neutrophils from primary immune organs to secondary immune organs, peripheral tissues, and blood, without compromising neutrophil trafficking to inflamed sites.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Animales , Bencilaminas , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Movimiento Celular/inmunología , Ciclamas , Femenino , Inmunofenotipificación , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Recuento de Leucocitos , Leucocitos/inmunología , Pulmón/inmunología , Pulmón/patología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ratones , Fenotipo , Bazo/efectos de los fármacos , Bazo/inmunología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Timo/efectos de los fármacos , Timo/inmunología
18.
Cell ; 160(4): 686-699, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25662009

RESUMEN

Chromothripsis is a catastrophic cellular event recently described in cancer in which chromosomes undergo massive deletion and rearrangement. Here, we report a case in which chromothripsis spontaneously cured a patient with WHIM syndrome, an autosomal dominant combined immunodeficiency disease caused by gain-of-function mutation of the chemokine receptor CXCR4. In this patient, deletion of the disease allele, CXCR4(R334X), as well as 163 other genes from one copy of chromosome 2 occurred in a hematopoietic stem cell (HSC) that repopulated the myeloid but not the lymphoid lineage. In competitive mouse bone marrow (BM) transplantation experiments, Cxcr4 haploinsufficiency was sufficient to confer a strong long-term engraftment advantage of donor BM over BM from either wild-type or WHIM syndrome model mice, suggesting a potential mechanism for the patient's cure. Our findings suggest that partial inactivation of CXCR4 may have general utility as a strategy to promote HSC engraftment in transplantation.


Asunto(s)
Inestabilidad Cromosómica , Síndromes de Inmunodeficiencia/genética , Verrugas/genética , Animales , Cromosomas Humanos , Modelos Animales de Enfermedad , Haploinsuficiencia , Células Madre Hematopoyéticas/metabolismo , Humanos , Linfocitos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Mosaicismo , Mutación , Células Mieloides/metabolismo , Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4/genética , Remisión Espontánea
19.
Zhongguo Zhong Yao Za Zhi ; 39(12): 2367-9, 2014 Jun.
Artículo en Chino | MEDLINE | ID: mdl-25244777

RESUMEN

Primary hepatic cancer is one of common malignant tumors. When being diagnosed, most patients were in middle and advanced stage and missed opportunities for surgical treatment. Therefore, chemotherapy and Chinese medicines become the main therapies for advanced primary hepatic cancer. This study was designed to observe the efficacy of Ganfule prescription combined with chemotherapy in treating advanced primary hepatic cancer. In the study, 58 cases of advanced primary hepatic cancer were randomly divided into the treatment group (30 cases) and the control group (28 cases). The treatment group was administered with Ganfule prescription combining with chemotherapy, while the control group was given chemotherapy alone. The tumors progress, quality of life, serum AFP level were evaluated in every three treatment cycles; and the survival rate was followed up for one year. According to the results of this study, after the treatment, there was no statistical significance in the comparison between the two groups in terms of response rate (RR) and disease control rate (DCR) (30.0% vs 25.0%, P = 0.670; 66.7% vs 60.7%, P = 0.637). The improvement rate of KPS score in the treatment group was significantly higher than that of the control group (43.33% vs 21.43%, P < 0.05). The reduction of serum AFP level in the treatment group was more significant than that of the control group (P < 0.05). During the one-year follow-up visit, the survival rate of the treatment group was 26.67%, and the control group was 25.00%, which indicated no statistical significance. This study drew the following conclusion that the oral administration of Ganfule prescription could improve the quality of life of patients of primary hepatic cancer, decrease the serum AFP level and maintain the disease control rate and the one-year survival rate.


Asunto(s)
Medicamentos Herbarios Chinos/administración & dosificación , Neoplasias Hepáticas/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Femenino , Humanos , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Resultado del Tratamiento
20.
Behav Genet ; 44(5): 498-515, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24997773

RESUMEN

Atypical Chemokine Receptor 1 (ACKR1), previously known as Duffy Antigen Receptor for Chemokines, stands out among chemokine receptors for high selective expression on cerebellar Purkinje neurons. Although ACKR1 ligands activate Purkinje cells in vitro, evidence for ACKR1 regulation of brain function in vivo is lacking. Here we demonstrate that Ackr1 (-/-) mice have markedly impaired balance and ataxia on a rotating rod and increased tremor when injected with harmaline, which induces whole-body tremor by activating Purkinje cells. Ackr1 (-/-) mice also exhibited impaired exploratory behavior, increased anxiety-like behavior and frequent episodes of marked hypoactivity under low-stress conditions. Surprisingly, Ackr1 (+/-) had similar behavioral abnormalities, indicating pronounced haploinsufficiency. The behavioral phenotype of Ackr1 (-/-) mice was the opposite of mouse models of cerebellar degeneration, and the defects persisted when Ackr1 was deficient only on non-hematopoietic cells. Together, the results suggest that normal motor function and behavior may partly depend on negative regulation of Purkinje cell activity by Ackr1.


Asunto(s)
Sistema del Grupo Sanguíneo Duffy , Actividad Motora , Células de Purkinje , Receptores de Superficie Celular , Animales , Femenino , Masculino , Ratones , Sistema del Grupo Sanguíneo Duffy/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Células de Purkinje/metabolismo , Receptores de Superficie Celular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA