Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Hamostaseologie ; 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37918839

RESUMEN

Inherited platelet disorders (IPDs) represent a heterogeneous group of disorders that include both quantitative (thrombocytopenia or thrombocytosis) and qualitative (thrombocytopathy) defects. To gain better knowledge about the prevalence, pathogenesis, and clinical consequences of specific diseases, to improve diagnosis and treatment of patients with IPD, and to support translational research on a genetic, molecular, and physiological basis, the THROMKIDplus study group currently comprising 24 sites in Germany, Austria, and Switzerland decided to establish a patient registry with associated biomaterial banking for children. This registry is designed as a retrospective-prospective, multicenter observational study and supposed to launch in the second half of 2023. Blood smears, plasma, platelet pellets, and DNA of patients will be stored in certified biomaterial banks for future translational research projects. The main inclusion criteria are (1) diagnosis of or highly suspected IPD after assessment of a THROMKIDplus competence center and (2) patients aged 0 to 17 years. Initial and follow-up data on patient history, laboratory parameters, standardized documentation of bleeding tendency, and congenital defects are collected according to good clinical practice and current data protection acts by using the MARVIN platform, a broadly used data management system supported by the German Society for Pediatric Oncology Hematology (GPOH). The THROMKIDplus study group intends to enroll ∼200 patients retrospectively and an annual amount of ∼50 patients prospectively.

2.
Best Pract Res Clin Haematol ; 34(2): 101286, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34404532

RESUMEN

Congenital amegakaryocytic thrombocytopenia (CAMT) is a rare inherited bone marrow failure syndrome (IBMFS) that is characterized by severe thrombocytopenia at birth due to ineffective megakaryopoiesis and development towards aplastic anemia during the first years of life. CAMT is not a single monogenetic disorder; rather, many descriptions of CAMT include different entities with different etiologies. CAMT in a narrow sense, which is primarily restricted to the hematopoietic system, is caused mainly by mutations in the gene for the thrombopoietin receptor (MPL), sometimes in the gene for its ligand (THPO). CAMT in association with radio-ulnar synostosis, which is not always clinically apparent, is mostly caused by mutations in MECOM, rarely in HOXA11. Patients affected by other IBMFS - especially Fanconi anemia or dyskeratosis congenita - may be misdiagnosed as having CAMT when they lack typical disease features of these syndromes or have only mild symptoms. This article reviews scientific and clinical aspects of the various disorders associated with the term "CAMT" with a main focus on the disease caused by mutations in the MPL gene.


Asunto(s)
Trombocitopenia , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Humanos , Recién Nacido , Mutación , Trombocitopenia/diagnóstico , Trombocitopenia/genética
3.
Elife ; 102021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34002692

RESUMEN

Expression levels of CX3CR1 (C-X3-C motif chemokine receptor 1) on immune cells have significant importance in maintaining tissue homeostasis under physiological and pathological conditions. The factors implicated in the regulation of CX3CR1 and its specific ligand CX3CL1 (fractalkine) expression remain largely unknown. Recent studies provide evidence that host's misfolded proteins occurring in the forms of polymers or amyloid fibrils can regulate CX3CR1 expression. Herein, a novel example demonstrates that polymers of human ZZ alpha-1 antitrypsin (Z-AAT) protein, resulting from its conformational misfolding due to the Z (Glu342Lys) mutation in SERPINA1 gene, strongly lower CX3CR1 mRNA expression in human peripheral blood mononuclear cells (PBMCs). This parallels with increase of intracellular levels of CX3CR1 and Z-AAT proteins. Presented data indicate the involvement of the CX3CR1 pathway in the Z-AAT-related disorders and further support the role of misfolded proteins in CX3CR1 regulation.


Proteins can lose their structure and form polymers because of mutations or changes in their immediate environment which can lead to cell damage and disease. Interestingly, polymers formed by a variety of proteins can reduce the levels of CX3C chemokine receptor 1 (CX3CR1 for short) that controls the behaviour of immune cells and is implicated in a range of illnesses. Inherited ZZ alpha-1 antitrypsin deficiency is a rare genetic condition that highly increases the risk of liver and lung diseases. This disorder is characterised by mutant alpha-1 antitrypsin proteins (AAT for short) reacting together to form polymers; yet it remains unclear how the polymers affect different cells or organs, and lead to diseases. To investigate this question, Tumpara et al. examined whether polymers of mutant AAT influence the level of the CX3CR1 protein in specific classes of immune cells. Experiments revealed that in people with AAT deficiency, certain blood immune cells express lower levels of CX3CR1. Regardless of age, clinical diagnosis, or treatment regimen, all individuals with ZZ alpha-1 antitrypsin deficiency had AAT polymers circulating in their blood: the higher the levels of polymers measured, the lower the expression of CX3CR1 recorded in the specific immune cells. When Tumpara et al. added polymers of mutant AAT to the immune cells of healthy donors, the expression of CX3CR1 dropped in a manner dependent on the polymer concentration. According to microscopy data, AAT polymers occurred inside cells alongside the CX3CR1 protein, suggesting that the two molecular actors interact. In the future, new drugs that remove these polymers, either from inside cells or as they circulate in the body, could help patients suffering from conditions associated with this abnormal protein aggregation.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Mutación , Polímeros , alfa 1-Antitripsina/genética , Adulto , Receptor 1 de Quimiocinas CX3C/genética , Quimiocina CX3CL1/metabolismo , Femenino , Humanos , Leucocitos Mononucleares , Masculino , Persona de Mediana Edad , Conformación Proteica , ARN Mensajero/metabolismo , alfa 1-Antitripsina/química
5.
Haematologica ; 106(9): 2439-2448, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32703794

RESUMEN

Congenital amegakaryocytic thrombocytopenia caused by deleterious homozygous or compound heterozygous mutations in MPL (CAMT-MPL) is a rare inherited bone marrow failure syndrome presenting as an isolated thrombocytopenia at birth progressing to pancytopenia due to exhaustion of hematopoietic progenitors. The analysis of samples and clinical data from a large cohort of 56 patients with CAMT-MPL resulted in a detailed description of the clinical picture and reliable genotype-phenotype correlations for this rare disease. We extended the spectrum of CAMT causing MPL mutations regarding number (17 novel mutations) and impact. The clinical courses showed a great variability with respect to the severity of thrombocytopenia, the development of pancytopenia and the consequences from bleedings. The most severe clinical problems were (1) intracranial bleedings pre- and perinatally and the resulting long-term consequences, and (2) the development of aplastic anemia in the later course of the disease. An important and new finding was that thrombocytopenia was not detected at birth in a quarter of the patients. The rate of non-hematological abnormalities in CAMT-MPL was higher than described so far. Most of the anomalies were related to the head region (brain anomalies, ocular and orbital anomalies) and consequences of intracranial bleedings. The present study demonstrates a higher variability of clinical courses than described so far and has important implications on diagnosis and therapy. The diagnosis CAMT-MPL has to be considered even for those patients who are inconspicuous in the first months of life or show somatic anomalies typical for other inherited bone marrow failure syndromes.


Asunto(s)
Receptores de Trombopoyetina , Trombocitopenia , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Humanos , Megacariocitos , Mutación , Receptores de Trombopoyetina/genética , Trombocitopenia/genética
6.
Front Psychiatry ; 11: 571, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32636772

RESUMEN

BACKGROUND: Major depressive disorder (MDD) represents a tremendous health threat to the world's population. Electroconvulsive therapy (ECT) is the most effective treatment option for refractory MDD patients. Ample evidence suggests brain-derived neurotrophic factor (BDNF) to play a crucial role in ECT's mode of action. Tissue-type plasminogen activator (t-PA) and plasminogen activator inhibitor-1 (PAI-1) are involved in BDNF production. HYPOTHESIS: The DNA methylation of gene regions encoding for t-PA and PAI-1 might be a suitable biomarker for ECT response prediction. METHODS: We withdrew blood from two cohorts of treatment-resistant MDD patients receiving ECT. In the first cohort (n = 59), blood was collected at baseline only. To evaluate DNA methylation changes throughout the treatment course, we acquired a second group (n = 28) and took blood samples at multiple time points. DNA isolated from whole blood and defined immune cell subtypes (B cells, monocytes, natural killer cells, and T cells) served for epigenetic analyses. RESULTS: Mixed linear models (corrected for multiple testing by Sidak's post-hoc test) revealed (1) no detectable baseline blood DNA methylation differences between ECT remitters (n = 33) and non-remitters (n = 53) in the regions analyzed, but (2) a significant difference in t-PA's DNA methylation between the investigated immune cell subtypes instead (p < 0.00001). This difference remained stable throughout the treatment course, showed no acute changes after ECT, and was independent of clinical remission. CONCLUSION: DNA methylation of both proteins seems to play a minor role in ECT's mechanisms. Generally, we recommend using defined immune cell subtypes (instead of whole blood only) for DNA methylation analyses.

7.
PLoS Pathog ; 16(7): e1008560, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32667948

RESUMEN

Human cytomegalovirus (HCMV) causes serious complications to immune compromised hosts. Dendritic cells (iDCgB) expressing granulocyte-macrophage colony-stimulating factor, interferon-alpha and HCMV-gB were developed to promote de novo antiviral adaptive responses. Mice reconstituted with a human immune system (HIS) were immunized with iDCgB and challenged with HCMV, resulting into 93% protection. Immunization stimulated the expansion of functional effector memory CD8+ and CD4+ T cells recognizing gB. Machine learning analyses confirmed bone marrow T/CD4+, liver B/IgA+ and spleen B/IgG+ cells as predictive biomarkers of immunization (≈87% accuracy). CD8+ and CD4+ T cell responses against gB were validated. Splenic gB-binding IgM-/IgG+ B cells were sorted and analyzed at a single cell level. iDCgB immunizations elicited human-like IgG responses with a broad usage of various IgG heavy chain V gene segments harboring variable levels of somatic hypermutation. From this search, two gB-binding human monoclonal IgGs were generated that neutralized HCMV infection in vitro. Passive immunization with these antibodies provided proof-of-concept evidence of protection against HCMV infection. This HIS/HCMV in vivo model system supported the validation of novel active and passive immune therapies for future clinical translation.


Asunto(s)
Anticuerpos Antivirales/inmunología , Infecciones por Citomegalovirus/inmunología , Vacunas contra Citomegalovirus/inmunología , Inmunización Pasiva , Inmunoglobulina G/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Antígenos Virales/inmunología , Citomegalovirus/inmunología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina G/farmacología , Ratones
8.
Eur Neuropsychopharmacol ; 36: 18-28, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32446707

RESUMEN

Major Depressive Disorder (MDD) is a major contributor to the global burden of disease. Approximately 30-50% of depressed patients fail to reach remission with standard treatment approaches. Electroconvulsive therapy (ECT) is one of the most effective options for these patients. Its exact therapeutic mechanism remains elusive, and reliable predictors of response are absent in the routine clinical practice. To characterize its mode of action and to facilitate treatment decision-making, we analyzed ECT's acute and chronic effects on various immune cell subsets. For this purpose, blood was withdrawn from depressed patients (n=21) directly before and 15 min after the first and last ECT session, respectively. After isolating peripheral blood mononuclear cells, we investigated defined populations of immune cells and their proportional changes upon ECT treatment using flow cytometry. By these means, we found ECT remitters (R; n=10) and non-remitters (NR; n=11) to differ in their relative proportion of putative immunoregulatory CD56highCD16-/dim and cytotoxic CD56dimCD16+ natural killer (NK) cells (CD56highCD16-/dim/CD56dimCD16+: R=0.064(±0.005), NR=0.047(±0.005), p<0.05; linear mixed models) and thus in their NK cell cytotoxicity. NK cell cytotoxicity was further increased after a single ECT session (before=0.066(±0.005), after=0.045(±0.005), p<0.001) and was associated with ECT quality parameters (maximum sustained coherence: r2=0.389, ß=-0.656, p<0.001) and long-term BDI-II rating changes (r2=0.459, ß=-0.726, p<0.05; both linear regression analysis). To conclude, particular NK cell subsets seem to be involved in ECT's acute effect and its clinical outcome. Due to the limited number of patients participating in our pilot study, future approaches are required to replicate our findings.


Asunto(s)
Trastorno Depresivo Mayor/sangre , Trastorno Depresivo Mayor/terapia , Terapia Electroconvulsiva/tendencias , Inmunidad Celular/fisiología , Células Asesinas Naturales/metabolismo , Convulsiones/sangre , Adulto , Anciano , Trastorno Depresivo Mayor/inmunología , Terapia Electroconvulsiva/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Convulsiones/inmunología , Resultado del Tratamiento , Adulto Joven
9.
Eur J Immunol ; 50(7): 1078-1080, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32181500

RESUMEN

AMPK (adenosine monophosphate-activated protein kinase) is phosphorylated (AMPK-P) in response to low energy through allosteric activation by Adenosine mono- or diphosphate (AMP/ADP). Folliculin (FLCN) and the FLCN-interacting proteins 1 and 2 (FNIP1, 2) modulate AMPK. FNIP1 deficiency patients have a AMPK-P gain of function phenotype with hypertrophic cardiomyopathy, Wolff-Parkinson-White pre-excitation syndrome, myopathy of skeletal muscles and combined immunodeficiency.


Asunto(s)
Cardiomiopatías , Proteínas Portadoras , Genes Recesivos , Síndromes de Inmunodeficiencia , Mutación , Síndromes de Preexcitación , Cardiomiopatías/genética , Cardiomiopatías/inmunología , Cardiomiopatías/patología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Femenino , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Masculino , Síndromes de Preexcitación/genética , Síndromes de Preexcitación/inmunología , Síndromes de Preexcitación/patología
10.
Theranostics ; 9(1): 152-166, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30662559

RESUMEN

Ischemia triggers a complex tissue response involving vascular, metabolic and inflammatory changes. METHODS: We combined hybrid SPECT/CT or PET/CT nuclear imaging studies of perfusion, metabolism and inflammation with multicolor flow cytometry-based cell population analysis to comprehensively analyze the ischemic tissue response and to elucidate the cellular substrate of noninvasive molecular imaging techniques in a mouse model of hind limb ischemia. RESULTS: Comparative analysis of tissue perfusion with [99mTc]-Sestamibi and arterial influx with [99mTc]-labeled albumin microspheres by SPECT/CT revealed a distinct pattern of response to vascular occlusion: an early ischemic period of matched suppression of tissue perfusion and arterial influx, a subacute ischemic period of normalized arterial influx but impaired tissue perfusion, and a protracted post-ischemic period of hyperdynamic arterial and normalized tissue perfusion, indicating coordination of macrovascular and microvascular responses. In addition, the subacute period showed increased glucose uptake by [18F]-FDG PET/CT scanning as the metabolic response of viable tissue to hypoperfusion. This was associated with robust macrophage infiltration by flow cytometry, and glucose uptake studies identified macrophages as major contributors to glucose utilization in ischemic tissue. Furthermore, imaging with the TSPO ligand [18F]-GE180 showed a peaked response during the subacute phase due to preferential labeling of monocytes and macrophages, while imaging with [68Ga]-RGD, an integrin ligand, showed prolonged post-ischemic upregulation, which was attributed to labeling of macrophages and endothelial cells by flow cytometry. CONCLUSION: Combined nuclear imaging and cell population analysis reveals distinct components of the ischemic tissue response and associated cell subsets, which could be targeted for therapeutic interventions.


Asunto(s)
Extremidades/patología , Isquemia/patología , Isquemia/fisiopatología , Animales , Arterias/patología , Modelos Animales de Enfermedad , Inflamación/patología , Metabolismo , Ratones , Imagen Óptica/métodos , Tomografía Computarizada por Tomografía de Emisión de Positrones , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único
11.
Blood Adv ; 2(6): 586-596, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29540340

RESUMEN

Heterozygous mutations in MECOM (MDS1 and EVI1 complex locus) have been reported to be causative of a rare association of congenital amegakaryocytic thrombocytopenia and radioulnar synostosis. Here we report on 12 patients with congenital hypomegakaryocytic thrombocytopenia caused by MECOM mutations (including 10 novel mutations). The mutations affected different functional domains of the EVI1 protein. The spectrum of phenotypes was much broader than initially reported for the first 3 patients; we found familial as well as sporadic cases, and the clinical spectrum ranged from isolated radioulnar synostosis with no or mild hematological involvement to severe bone marrow failure without obvious skeletal abnormality. The clinical picture included radioulnar synostosis, bone marrow failure, clinodactyly, cardiac and renal malformations, B-cell deficiency, and presenile hearing loss. No single clinical manifestation was detected in all patients affected by MECOM mutations. Radioulnar synostosis and B-cell deficiency were observed only in patients with mutations affecting a short region in the C-terminal zinc finger domain of EVI1. We propose the term MECOM-associated syndrome for this heterogeneous hereditary disease and inclusion of MECOM sequencing in the diagnostic workup of congenital bone marrow failure.


Asunto(s)
Anemia Aplásica/genética , Enfermedades de la Médula Ósea/genética , Estudios de Asociación Genética , Heterogeneidad Genética , Predisposición Genética a la Enfermedad , Hemoglobinuria Paroxística/genética , Proteína del Locus del Complejo MDS1 y EV11/genética , Trombocitopenia/genética , Anemia Aplásica/diagnóstico , Enfermedades de la Médula Ósea/diagnóstico , Trastornos de Fallo de la Médula Ósea , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Proteínas de Unión al ADN/sangre , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hemoglobinuria Paroxística/diagnóstico , Humanos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Masculino , Mutación , Linaje , Trombocitopenia/diagnóstico , Factores de Transcripción/sangre
12.
Eur J Med Genet ; 61(11): 715-722, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29545013

RESUMEN

Inherited thrombocytopenias (ITs) are a heterogeneous group of disorders characterized by low platelet count resulting in impaired hemostasis. Patients can have spontaneous hemorrhages and/or excessive bleedings provoked by hemostatic challenges as trauma or surgery. To date, ITs encompass 32 different rare monogenic disorders caused by mutations of 30 genes. This review will focus on the major discoveries that have been made in the last years on the diagnosis, treatment and molecular mechanisms of ANKRD26-Related Thrombocytopenia and MYH9-Related Diseases. Furthermore, we will discuss the use a Thrombopoietin mimetic as a novel approach to treat the thrombocytopenia in these patients. We will propose the use of a new 3D bone marrow model to study the mechanisms of action of these drugs and to test their efficacy and safety in patients. The overall purpose of this review is to point out that important progresses have been made in understanding the pathogenesis of ANKRD26-Related Thrombocytopenia and MYH9-Related Diseases and new therapeutic approaches have been proposed and tested. Future advancement in this research will rely in the development of more physiological models to study the regulation of human platelet biogenesis, disease mechanisms and specific pharmacologic targets.


Asunto(s)
Predisposición Genética a la Enfermedad , Proteínas Motoras Moleculares/genética , Cadenas Pesadas de Miosina/genética , Proteínas Nucleares/genética , Trombocitopenia/tratamiento farmacológico , Plaquetas/metabolismo , Plaquetas/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular , Mutación , Trombocitopenia/genética , Trombocitopenia/patología
13.
Blood ; 127(23): 2791-803, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27084890

RESUMEN

Inherited bleeding, thrombotic, and platelet disorders (BPDs) are diseases that affect ∼300 individuals per million births. With the exception of hemophilia and von Willebrand disease patients, a molecular analysis for patients with a BPD is often unavailable. Many specialized tests are usually required to reach a putative diagnosis and they are typically performed in a step-wise manner to control costs. This approach causes delays and a conclusive molecular diagnosis is often never reached, which can compromise treatment and impede rapid identification of affected relatives. To address this unmet diagnostic need, we designed a high-throughput sequencing platform targeting 63 genes relevant for BPDs. The platform can call single nucleotide variants, short insertions/deletions, and large copy number variants (though not inversions) which are subjected to automated filtering for diagnostic prioritization, resulting in an average of 5.34 candidate variants per individual. We sequenced 159 and 137 samples, respectively, from cases with and without previously known causal variants. Among the latter group, 61 cases had clinical and laboratory phenotypes indicative of a particular molecular etiology, whereas the remainder had an a priori highly uncertain etiology. All previously detected variants were recapitulated and, when the etiology was suspected but unknown or uncertain, a molecular diagnosis was reached in 56 of 61 and only 8 of 76 cases, respectively. The latter category highlights the need for further research into novel causes of BPDs. The ThromboGenomics platform thus provides an affordable DNA-based test to diagnose patients suspected of having a known inherited BPD.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/genética , Predisposición Genética a la Enfermedad , Hemorragia/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Trombosis/genética , Estudios de Casos y Controles , Variaciones en el Número de Copia de ADN , Femenino , Estudios de Asociación Genética/métodos , Humanos , Masculino , Mutación , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN/métodos
15.
Cytometry A ; 87(5): 405-18, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25728583

RESUMEN

Lentiviral and gammaretroviral vectors are state-of-the-art tools for transgene expression within target cells. The integration of these vectors can be deliberately suppressed to derive a transient gene expression system based on extrachromosomal circular episomes with intact coding regions. These episomes can be used to deliver DNA templates and to express RNA or protein. Importantly, transient gene transfer avoids the genotoxic side effects of integrating vectors. Restricting their applicability, episomes are rapidly lost upon dilution in dividing target cells. Addressing this limitation, we could establish comparably stable percentages of transgene-positive cells over prolonged time periods in proliferating cells by repeated transductions. Flow cytometry was applied for kinetic analyses to decipher the impact of individual parameters on the kinetics of fluoroprotein expression after episomal retransduction and to visualize sequential and simultaneous transfer of heterologous fluoroproteins. Expression windows could be exactly timed by the number of transduction steps. The kinetics of signal loss was affected by the cell proliferation rate. The transfer of genes encoding fluoroproteins with different half-lives revealed a major impact of protein stability on temporal signal distribution and accumulation, determining optimal retransduction intervals. In addition, sequential transductions proved broad applicability in different cell types and using different envelope pseudotypes without receptor overload. Stable percentages of cells coexpressing multiple transgenes could be generated upon repeated coadministration of different episomal vectors. Alternatively, defined patterns of transgene expression could be recapitulated by sequential transductions. Altogether, we established a methodology to control and adjust a temporally defined window of transgene expression using retroviral episomal vectors. Combined with the highly efficient cell entry of these vectors while avoiding integration, the developed technology is of great significance for a broad panel of applications, including transcription-factor-based induced cell fate conversion and controlled transfer of genetically encoded RNA- or protein-based drugs.


Asunto(s)
Expresión Génica , Vectores Genéticos , Transducción Genética/métodos , Humanos , Cinética , Lentivirus/genética , Plásmidos/genética , Transgenes/genética
16.
Eur J Immunol ; 44(11): 3368-79, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25100508

RESUMEN

Monocytes are known to engage in reciprocal crosstalk with NK cells but their influence on NK-cell-associated antibody-dependent cellular cytotoxicity (ADCC) is not well understood. We demonstrate that in humans FcγRIII (CD16)-dependent ADCC by NK cells is considerably enhanced by monocytes, and that this effect is regulated by FcγRII (CD32) crosslinking in healthy individuals. It is known that during HIV-1 infection, NK cells are known to express low levels of CD16 and exhibit reduced ADCC. We show that immune regulation of CD16-mediated NK-cell cytotoxicity by monocytes through CD32 engagement is substantially disturbed in chronic progressive HIV-1 infection. Expression of activating isoform of CD32 represented a compensatory mechanism for reduced expression of CD16 on NK cells during HIV-1 infection. As a result, the regulation of NK-cell-associated ADCC by monocytes is skewed and eventually constitutes a novel factor that contributes to HIV-1-associated immune deficiency, dysregulation and pathogenesis. Our data therefore provide evidence, for the first time, that in humans monocytes act as a rheostat for FcγRIII-mediated NK-cell functions maintaining a well-balanced immune response.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Infecciones por VIH/inmunología , Células Asesinas Naturales/inmunología , Monocitos/inmunología , Receptores de IgG/inmunología , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/inmunología , VIH-1/inmunología , Humanos , Receptores de IgG/biosíntesis , Receptores de IgG/genética
17.
J Virol ; 88(21): 12397-408, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25122796

RESUMEN

UNLABELLED: Natural killer (NK) cells are effector and regulatory innate immune cells and play a critical role in the first line of defense against various viral infections. Although previous reports have indicated the vital contributions of NK cells to HIV-1 immune control, nongenetic NK cell parameters directly associated with slower disease progression have not been defined yet. In a longitudinal, retrospective study of 117 untreated HIV-infected subjects, we show that higher frequencies as well as the absolute numbers of CD8(+) CD3(-) lymphocytes are linked to delayed HIV-1 disease progression. We show that the majority of these cells are well-described blood NK cells. In a subsequent cross-sectional study, we demonstrate a significant loss of CD8(+) NK cells in untreated HIV-infected individuals, which correlated with HIV loads and inversely correlated with CD4(+) T cell counts. CD8(+) NK cells had modestly higher frequencies of CD57-expressing cells than CD8(-) cells, but CD8(+) and CD8(-) NK cells showed no differences in the expression of a number of activating and inhibiting NK cell receptors. However, CD8(+) NK cells exhibited a more functional profile, as detected by cytokine production and degranulation. IMPORTANCE: We demonstrate that the frequency of highly functional CD8(+) NK cells is inversely associated with HIV-related disease markers and linked with delayed disease progression. These results thus indicate that CD8(+) NK cells represent a novel NK cell-derived, innate immune correlate with an improved clinical outcome in HIV infection.


Asunto(s)
Antígenos CD8/análisis , Infecciones por VIH/inmunología , VIH-1/inmunología , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/inmunología , Complejo CD3/análisis , Estudios de Cohortes , Estudios Transversales , Progresión de la Enfermedad , Infecciones por VIH/virología , Humanos , Células Asesinas Naturales/química , Estudios Longitudinales , Subgrupos Linfocitarios/química , Estudios Retrospectivos
18.
Hum Mutat ; 35(2): 236-47, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24186861

RESUMEN

MYH9-related disease (MYH9-RD) is a rare autosomal-dominant disorder caused by mutations in the gene for nonmuscle myosin heavy chain IIA (NMMHC-IIA). MYH9-RD is characterized by a considerable variability in clinical evolution: patients present at birth with only thrombocytopenia, but some of them subsequently develop sensorineural deafness, cataract, and/or nephropathy often leading to end-stage renal disease (ESRD). We searched for genotype-phenotype correlations in the largest series of consecutive MYH9-RD patients collected so far (255 cases from 121 families). Association of genotypes with noncongenital features was assessed by a generalized linear regression model. The analysis defined disease evolution associated to seven different MYH9 genotypes that are responsible for 85% of MYH9-RD cases. Mutations hitting residue R702 demonstrated a complete penetrance for early-onset ESRD and deafness. The p.D1424H substitution associated with high risk of developing all the noncongenital manifestations of disease. Mutations hitting a distinct hydrophobic seam in the NMMHC-IIA head domain or substitutions at R1165 associated with high risk of deafness but low risk of nephropathy or cataract. Patients with p.E1841K, p.D1424N, and C-terminal deletions had low risk of noncongenital defects. These findings are essential to patients' clinical management and genetic counseling and are discussed in view of molecular pathogenesis of MYH9-RD.


Asunto(s)
Catarata/genética , Estudios de Asociación Genética , Pérdida Auditiva Sensorineural/genética , Proteínas Motoras Moleculares/genética , Cadenas Pesadas de Miosina/genética , Trombocitopenia/congénito , Adulto , Edad de Inicio , Sustitución de Aminoácidos , Femenino , Genotipo , Pérdida Auditiva Sensorineural/complicaciones , Pérdida Auditiva Sensorineural/diagnóstico , Humanos , Italia , Modelos Lineales , Masculino , Mutación , Fenotipo , Factores de Riesgo , Trombocitopenia/complicaciones , Trombocitopenia/diagnóstico , Trombocitopenia/genética
19.
J Hepatol ; 59(3): 528-35, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23665041

RESUMEN

BACKGROUND & AIMS: Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of cells associated with the suppression of immunity. However, little is known about how or where MDSCs are induced and from which cells they originate. The liver is known for its immune regulatory functions. Here, we investigated the capacity of human hepatic stellate cells (HSCs) to transform peripheral blood monocytes into MDSCs. METHODS: We cultured freshly isolated human monocytes from healthy donors on primary human HSCs or an HSC cell-line and characterized the phenotype and function of resulting CD14(+)HLA-DR(-/low) monocytes by flow cytometry, quantitative PCR, and functional assays. We analyzed the molecular mechanisms underlying the induction and function of the CD14(+)HLA-DR(-/low) cells by using blocking antibodies or knock-down technology. RESULTS: Mature peripheral blood monocytes co-cultured with HSCs downregulated HLA-DR and developed a phenotypic and functional profile similar to MDSCs. Only activated but not freshly isolated HSCs were capable of inducing CD14(+)HLA-DR(-/low) cells. Such CD14(+)HLA-DR(-/low) monocyte-derived MDSCs suppressed T-cell proliferation in an arginase-1 dependent fashion. HSC-induced development of CD14(+)HLA-DR(-/low) monocyte-derived MDSCs was not mediated by soluble factors, but required physical interaction and was abrogated by blocking CD44. CONCLUSIONS: Our study shows that activated human HSCs convert mature peripheral blood monocytes into MDSCs. As HSCs are activated during chronic inflammation, the subsequent local induction of MDSCs may prevent ensuing excessive liver injury. HSC-induced MDSCs functionally and phenotypically resemble those isolated from liver cancer patients. Thus, our data suggest that local generation of MDSCs by liver-resident HSCs may contribute to immune suppression during inflammation and cancer in the liver.


Asunto(s)
Células Estrelladas Hepáticas/inmunología , Receptores de Hialuranos/metabolismo , Monocitos/citología , Monocitos/inmunología , Células Mieloides/citología , Células Mieloides/inmunología , Arginasa/antagonistas & inhibidores , Arginasa/metabolismo , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Línea Celular , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Regulación hacia Abajo , Antígenos HLA-DR/metabolismo , Humanos , Tolerancia Inmunológica , Receptores de Lipopolisacáridos/metabolismo , Activación de Linfocitos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
20.
Proc Natl Acad Sci U S A ; 110(18): 7407-12, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23589855

RESUMEN

T-cell receptor (TCR) signal strength determines selection and lineage fate at the CD4(+)CD8(+) double-positive stage of intrathymic T-cell development. Members of the miR-181 family constitute the most abundantly expressed microRNA at this stage of T-cell development. Here we show that deletion of miR-181a/b-1 reduced the responsiveness of double-positive thymocytes to TCR signals and virtually abrogated early invariant natural killer T (iNKT) cell development, resulting in a dramatic reduction in iNKT cell numbers in thymus as well as in the periphery. Increased concentrations of agonist ligand rescued iNKT cell development in miR-181a/b-1(-/-) mice. Our results define a critical role of miR-181a/b-1 in early iNKT cell development and show that miR-181a/b-1 sets a TCR signaling threshold for agonist selection.


Asunto(s)
Selección Clonal Mediada por Antígenos/inmunología , MicroARNs/metabolismo , Células T Asesinas Naturales/inmunología , Animales , Proliferación Celular , Ligandos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Células T Asesinas Naturales/citología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...