Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Glia ; 68(6): 1165-1181, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31859421

RESUMEN

Distal axonopathy is a recognized pathological feature of amyotrophic lateral sclerosis (ALS). In the peripheral nerves of ALS patients, motor axon loss elicits a Wallerian-like degeneration characterized by denervated Schwann cells (SCs) together with immune cell infiltration. However, the pathogenic significance of denervated SCs accumulating following impaired axonal growth in ALS remains unclear. Here, we analyze SC phenotypes in sciatic nerves of ALS patients and paralytic SOD1G93A rats, and identify remarkably similar and specific reactive SC phenotypes based on the pattern of S100ß, GFAP, isolectin and/or p75NTR immunoreactivity. Different subsets of reactive SCs expressed colony-stimulating factor-1 (CSF1) and Interleukin-34 (IL-34) and closely interacted with numerous endoneurial CSF-1R-expressing monocyte/macrophages, suggesting a paracrine mechanism of myeloid cell expansion and activation. SCs bearing phagocytic phenotypes as well as endoneurial macrophages expressed stem cell factor (SCF), a trophic factor that attracts and activates mast cells through the c-Kit receptor. Notably, a subpopulation of Ki67+ SCs expressed c-Kit in the sciatic nerves of SOD1G93A rats, suggesting a signaling pathway that fuels SC proliferation in ALS. c-Kit+ mast cells were also abundant in the sciatic nerve from ALS donors but not in controls. Pharmacological inhibition of CSF-1R and c-Kit with masitinib in SOD1G93A rats potently reduced SC reactivity and immune cell infiltration in the sciatic nerve and ventral roots, suggesting a mechanism by which the drug ameliorates peripheral nerve pathology. These findings provide strong evidence for a previously unknown inflammatory mechanism triggered by SCs in ALS peripheral nerves that has broad application in developing novel therapies.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Inflamación/metabolismo , Interleucinas/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Células de Schwann/metabolismo , Factor de Células Madre/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Axones/patología , Modelos Animales de Enfermedad , Humanos , Masculino , Neuronas Motoras/patología , Neuroglía/metabolismo , Ratas Transgénicas
2.
Nat Immunol ; 9(10): 1148-56, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18758465

RESUMEN

Dendritic cell (DC) maturation and migration are events critical for the initiation of immune responses. After encountering pathogens, DCs upregulate the expression of costimulatory molecules and subsequently migrate to secondary lymphoid organs. Calcium (Ca(2+)) entry governs the functions of many hematopoietic cell types, but the role of Ca(2+) entry in DC biology remains unclear. Here we report that the Ca(2+)-activated nonselective cation channel TRPM4 was expressed in and controlled the Ca(2+) homeostasis of mouse DCs. The absence of TRPM4, which elicited Ca(2+) overload, did not influence DC maturation but did considerably impair chemokine-dependent DC migration. Our results establish TRPM4-regulated Ca(2+) homeostasis as crucial for DC mobility but not maturation and emphasize that DC maturation and migration are independently regulated.


Asunto(s)
Señalización del Calcio/inmunología , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Células Dendríticas/citología , Canales Catiónicos TRPM/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Expresión Génica/inmunología , Homeostasis/inmunología , Immunoblotting , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo
3.
Haematologica ; 105(9): 2240-2249, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33054049

RESUMEN

ß-thalassemia major (ß-TM) is an inherited hemoglobinopathy caused by a quantitative defect in the synthesis of ß-globin chains of hemoglobin, leading to the accumulation of free a-globin chains that aggregate and cause ineffective erythropoiesis. We have previously demonstrated that terminal erythroid maturation requires a transient activation of caspase-3 and that the chaperone Heat Shock Protein 70 (HSP70) accumulates in the nucleus to protect GATA-1 transcription factor from caspase-3 cleavage. This nuclear accumulation of HSP70 is inhibited in human ß-TM erythroblasts due to HSP70 sequestration in the cytoplasm by free a-globin chains, resulting in maturation arrest and apoptosis. Likewise, terminal maturation can be restored by transduction of a nuclear-targeted HSP70 mutant. Here we demonstrate that in normal erythroid progenitors, HSP70 localization is regulated by the exportin-1 (XPO1), and that treatment of ß-thalassemic erythroblasts with an XPO1 inhibitor increased the amount of nuclear HSP70, rescued GATA-1 expression and improved terminal differentiation, thus representing a new therapeutic option to ameliorate ineffective erythropoiesis of ß-TM.


Asunto(s)
Carioferinas , Receptores Citoplasmáticos y Nucleares , Talasemia beta , Diferenciación Celular , Eritroblastos , Eritropoyesis , Humanos , Carioferinas/genética , Receptores Citoplasmáticos y Nucleares/genética , Talasemia beta/tratamiento farmacológico , Talasemia beta/genética , Proteína Exportina 1
4.
Int J Mol Sci ; 20(16)2019 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-31395804

RESUMEN

Amyotrophic lateral sclerosis (ALS) is characterized by degeneration of upper and lower motor neurons accompanied by proliferation of reactive microglia in affected regions. However, it is unknown whether the hematopoietic marker CD34 can identify a subpopulation of proliferating microglial cells in the ALS degenerating spinal cord. Immunohistochemistry for CD34 and microglia markers was performed in lumbar spinal cords of ALS rats bearing the SOD1G93A mutation and autopsied ALS and control human subjects. Characterization of CD34-positive cells was also performed in primary cell cultures of the rat spinal cords. CD34 was expressed in a large number of cells that closely interacted with degenerating lumbar spinal cord motor neurons in symptomatic SOD1G93A rats, but not in controls. Most CD34+ cells co-expressed the myeloid marker CD11b, while only a subpopulation was stained for Iba1 or CD68. Notably, CD34+ cells actively proliferated and formed clusters adjacent to damaged motor neurons bearing misfolded SOD1. CD34+ cells were identified in the proximity of motor neurons in autopsied spinal cord from sporadic ALS subjects but not in controls. Cell culture of symptomatic SOD1G93A rat spinal cords yielded a large number of CD34+ cells exclusively in the non-adherent phase, which generated microglia after successive passaging. A yet unrecognized CD34+ cells, expressing or not the microglial marker Iba1, proliferate and accumulate adjacent to degenerating spinal motor neurons, representing an intriguing cell target for approaching ALS pathogenesis and therapeutics.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Antígenos CD34/análisis , Microglía/patología , Neuronas Motoras/patología , Esclerosis Amiotrófica Lateral/genética , Animales , Proliferación Celular , Células Cultivadas , Humanos , Masculino , Microglía/citología , Mutación Puntual , Pliegue de Proteína , Ratas , Médula Espinal/patología , Superóxido Dismutasa-1/análisis , Superóxido Dismutasa-1/genética
5.
J Am Soc Nephrol ; 28(12): 3605-3615, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28784700

RESUMEN

Renal transplants remain a medical challenge, because the parameters governing allograft outcome are incompletely identified. Here, we investigated the role of serum iron in the sterile inflammation that follows kidney ischemia-reperfusion injury. In a retrospective cohort study of renal allograft recipients (n=169), increased baseline levels of serum ferritin reliably predicted a positive outcome for allografts, particularly in elderly patients. In mice, systemic iron overload protected against renal ischemia-reperfusion injury-associated sterile inflammation. Furthermore, chronic iron injection in mice prevented macrophage recruitment after inflammatory stimuli. Macrophages cultured in high-iron conditions had reduced responses to Toll-like receptor-2, -3, and -4 agonists, which associated with decreased reactive oxygen species production, increased nuclear localization of the NRF2 transcription factor, increased expression of the NRF2-related antioxidant response genes, and limited NF-κB and proinflammatory signaling. In macrophage-depleted animals, the infusion of macrophages cultured in high-iron conditions did not reconstitute AKI after ischemia-reperfusion, whereas macrophages cultured in physiologic iron conditions did. These findings identify serum iron as a critical protective factor in renal allograft outcome. Increasing serum iron levels in patients may thus improve prognosis of renal transplants.


Asunto(s)
Hierro/sangre , Riñón/patología , Daño por Reperfusión/prevención & control , Adulto , Aloinjertos , Animales , Antioxidantes/metabolismo , Femenino , Ferritinas/sangre , Tasa de Filtración Glomerular , Humanos , Inflamación , Hierro/química , Riñón/metabolismo , Trasplante de Riñón , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Monocitos/citología , Factor 2 Relacionado con NF-E2/metabolismo , Peritonitis/metabolismo , Pronóstico , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal
6.
Blood ; 126(17): 1974-5, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26494918

RESUMEN

In this issue of Blood, Kautz et al show that the ablation of the erythroid-derived factor erythroferrone (ERFE), which has been shown to be highly expressed in ß-thalassemic mice, restores hepcidin levels and corrects iron overload. However, correction of hepcidin levels in those mice does not improve anemia of ß-thalassemia.


Asunto(s)
Citocinas/fisiología , Modelos Animales de Enfermedad , Hepcidinas/metabolismo , Sobrecarga de Hierro/etiología , Proteínas Musculares/fisiología , Talasemia beta/complicaciones , Talasemia beta/patología , Animales , Femenino , Masculino
7.
Curr Opin Hematol ; 23(3): 181-8, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26779615

RESUMEN

PURPOSE OF REVIEW: The review provides an overview of recent data regarding the molecular players in ß-thalassemia dyserythropoiesis and the corresponding therapeutic implications. RECENT FINDINGS: ß-thalassemia dyserythropoiesis is characterized by four steps: expansion of erythroid progenitors, accelerated erythroid differentiation until the polychromatophilic stage, maturation arrest, and apoptosis at the polychromatophilic stage. Excess α-globin chains are the primary culprit in the disease, but the link between this excess and ineffective erythropoiesis has only recently been established. Important recent advances in understanding the molecular determinants involved in two critical steps of dyserythropoiesis are paving the way to new alternative targets for the treatment of this disease. SUMMARY: Growth differentiation factor 11 (GDF11) blockade increases the apoptosis of erythroblasts with excess α-chains by upregulating Fas-ligand in late basophilic and polychromatophilic erythroblasts, thereby decreasing cell expansion (step 1). Blocking GDF11 alleviates anemia in a mouse model of ß-thalassemia and also in humans, most likely by promoting cells of 'good' erythroblastic lineage containing an α-/non-α-globin chain ratio of close to 1. Maturation arrest at the polychromatophilic stage (step 3) is associated with the depletion of GATA binding protein 1 (GATA-1) from the nucleus, which results from cytoplasmic sequestration of heat shock protein 70 (HSP70) by α-globin chains. Small molecules disrupting the HSP70/α-globin complex in the cytoplasm or decreasing HSP70 nuclear export might increase the nuclear localization of HSP70, thereby protecting GATA-1 and alleviating anemia. Finally, increasing the serum levels of hepcidin or transferrin alleviates anemia and dyserythropoiesis by diminishing iron uptake by erythroblasts in mouse models.


Asunto(s)
Eritropoyesis , Talasemia beta/tratamiento farmacológico , Talasemia beta/metabolismo , Animales , Humanos
8.
J Neuroinflammation ; 13(1): 177, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27400786

RESUMEN

BACKGROUND: In the SOD1(G93A) mutant rat model of amyotrophic lateral sclerosis (ALS), neuronal death and rapid paralysis progression are associated with the emergence of activated aberrant glial cells that proliferate in the degenerating spinal cord. Whether pharmacological downregulation of such aberrant glial cells will decrease motor neuron death and prolong survival is unknown. We hypothesized that proliferation of aberrant glial cells is dependent on kinase receptor activation, and therefore, the tyrosine kinase inhibitor masitinib (AB1010) could potentially control neuroinflammation in the rat model of ALS. METHODS: The cellular effects of pharmacological inhibition of tyrosine kinases with masitinib were analyzed in cell cultures of microglia isolated from aged symptomatic SOD1(G93A) rats. To determine whether masitinib prevented the appearance of aberrant glial cells or modified post-paralysis survival, the drug was orally administered at 30 mg/kg/day starting after paralysis onset. RESULTS: We found that masitinib selectively inhibited the tyrosine kinase receptor colony-stimulating factor 1R (CSF-1R) at nanomolar concentrations. In microglia cultures from symptomatic SOD1(G93A) spinal cords, masitinib prevented CSF-induced proliferation, cell migration, and the expression of inflammatory mediators. Oral administration of masitinib to SOD1(G93A) rats starting after paralysis onset decreased the number of aberrant glial cells, microgliosis, and motor neuron pathology in the degenerating spinal cord, relative to vehicle-treated rats. Masitinib treatment initiated 7 days after paralysis onset prolonged post-paralysis survival by 40 %. CONCLUSIONS: These data show that masitinib is capable of controlling microgliosis and the emergence/expansion of aberrant glial cells, thus providing a strong biological rationale for its use to control neuroinflammation in ALS. Remarkably, masitinib significantly prolonged survival when delivered after paralysis onset, an unprecedented effect in preclinical models of ALS, and therefore appears well-suited for treating ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/complicaciones , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Parálisis/tratamiento farmacológico , Parálisis/etiología , Inhibidores de Proteínas Quinasas/uso terapéutico , Tiazoles/uso terapéutico , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/mortalidad , Animales , Benzamidas , Muerte Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Masculino , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Mutación/genética , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Piperidinas , Piridinas , Ratas , Ratas Transgénicas , Médula Espinal/patología , Superóxido Dismutasa/genética
9.
Curr Opin Hematol ; 22(3): 193-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25767952

RESUMEN

PURPOSE OF REVIEW: The type 1 transferrin receptor (TfR1) is well known as a key player in erythroid differentiation through its role in iron uptake. Recently, it has been demonstrated that TfR1 could also have signaling functions in erythroid cells. Moreover, the second transferrin receptor, TfR2, whose signaling functions in hepatic cells are well established, was recently shown to be a partner of the erythropoietin receptor (EpoR) and thereby likely to play a role in erythroid differentiation. RECENT FINDINGS: This review reports recent findings regarding the specificities of the regulation of TfR1 expression and iron uptake in erythroblasts. The newly discovered noncanonical actions of TfR1 and TfR2 in erythroid cells are also discussed. SUMMARY: Erythrocytes contain more than 60% of the iron of the body and each day, differentiating erythroid cells uptake around 20 mg of iron for heme synthesis. Accordingly, TfR1 is one of the most abundant membrane proteins of the erythroblasts and it is not surprising that specific regulations regarding both its expression and its mechanism of action operate in erythroblasts. The signaling functions of both TfR1 and TfR2 in erythroid cells were unexpected and these recent findings open a new field of research regarding the last steps of erythroid differentiation and their regulation.


Asunto(s)
Eritropoyesis/fisiología , Receptores de Transferrina/metabolismo , Diferenciación Celular , Humanos , Hierro/metabolismo
10.
J Immunol ; 191(5): 2335-43, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23926325

RESUMEN

IgA plays ambivalent roles in the immune system. The balance between inhibitory and activating responses relies on the multimerization status of IgA and interaction with their cognate receptors. In mucosal sites, secretory IgA (SIgA) protects the host through immune-exclusion mechanisms, but its function in the bloodstream remains unknown. Using bone marrow-derived dendritic cells, we found that both human and mouse SIgA induce tolerogenic dendritic cells (DCs) following binding to specific ICAM-3 grabbing nonintegrin receptor 1. This interaction was dependent on Ca(2+) and mannose residues. SIgA-primed DCs (SIgA-DCs) are resistant to TLR-dependent maturation. Although SIgA-DCs fail to induce efficient proliferation and Th1 differentiation of naive responder T cells, they generate the expansion of regulatory T cells through IL-10 production. SIgA-DCs are highly potent in inhibiting autoimmune responses in mouse models of type 1 diabetes and multiple sclerosis. This discovery may offer new insights about mucosal-derived DC immunoregulation through SIgA opening new therapeutic approaches to autoimmune diseases.


Asunto(s)
Autoinmunidad/inmunología , Moléculas de Adhesión Celular/inmunología , Células Dendríticas/inmunología , Inmunoglobulina A Secretora/inmunología , Lectinas Tipo C/inmunología , Receptores de Superficie Celular/inmunología , Animales , Células de la Médula Ósea/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/citología , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Humanos , Tolerancia Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL
11.
Lab Invest ; 92(4): 625-35, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22330344

RESUMEN

Coeliac disease (CD) is a malabsorptive enteropathy resulting from intolerance to gluten. Environmental factors and the microbiota are suggested to have critical roles in the onset of CD. The CD71 IgA receptor on epithelial cells is responsible for abnormal retrotranscytosis of IgA-gluten peptide complexes from the intestinal lumen into the lamina propria, inducing intestinal inflammation. However, understanding the role of gluten in the CD physiopathology has been hindered by the absence of relevant animal models. Here, we generated a mouse model for CD to study the factors controlling its pathogenesis as well as to investigate the influence of oral delivery of probiotics on disease development. Gluten sensitivity was established by feeding three generations of BALB/c mice a gluten-free diet (G-) followed by gluten challenge (G+) for 30 days. The G+ mice developed villous atrophy, crypt hyperplasia and infiltration of T cells and macrophages in the small intestine. Inflammation was associated with an overexpression of CD71 on the apical side of enterocytes and an increase of plasma cells producing IgA, which colocalised with the CD71. Moreover, IgA colocalised with the transglutaminase 2 (TG2), the production of which was increased in the lamina propria of G+ mice. These mice displayed increased production of cyclooxygenase-2 (COX-2), pro-inflammatory cytokines and IL-15, as well as anti-gliadin and anti-TG2 autoantibodies. The commensal flora-isolated presumptive probiotic Saccharomyces boulardii KK1 strain hydrolysed the 28-kDa α-gliadin fraction, and its oral delivery in G+ mice improved enteropathy development in association with decrease of epithelial cell CD71 expression and local cytokine production. In conclusion, the G+ BALB/c mouse represents a new mouse model for human CD based on histopathological features and expression of common biomarkers. The selected probiotic treatment reversing disease development will allow the study of the role of probiotics as a new therapeutic approach of CD.


Asunto(s)
Antígenos CD/metabolismo , Enfermedad Celíaca/etiología , Modelos Animales de Enfermedad , Proteínas de Unión al GTP/metabolismo , Glútenes/efectos adversos , Inmunoglobulina A/metabolismo , Receptores de Transferrina/metabolismo , Transglutaminasas/metabolismo , Inmunidad Adaptativa , Animales , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/prevención & control , Enterocitos/metabolismo , Femenino , Glútenes/inmunología , Inmunidad Innata , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Probióticos/uso terapéutico , Proteína Glutamina Gamma Glutamiltransferasa 2 , Saccharomyces
12.
Kidney Int ; 82(12): 1284-96, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22951891

RESUMEN

IgA nephropathy (IgAN), the most common primary glomerulonephritis worldwide, has significant morbidity and mortality as 20-40% of patients progress to end-stage renal disease within 20 years of onset. In order to gain insight into the molecular mechanisms involved in the progression of IgAN, we systematically evaluated renal biopsies from such patients. This showed that the MAPK/ERK signaling pathway was activated in the mesangium of patients presenting with over 1 g/day proteinuria and elevated blood pressure, but absent in biopsy specimens of patients with IgAN and modest proteinuria (<1 g/day). ERK activation was not associated with elevated galactose-deficient IgA1 or IgG specific for galactose-deficient IgA1 in the serum. In human mesangial cells in vitro, ERK activation through mesangial IgA1 receptor (CD71) controlled pro-inflammatory cytokine secretion and was induced by large-molecular-mass IgA1-containing circulating immune complexes purified from patient sera. Moreover, IgA1-dependent ERK activation required renin-angiotensin system as its blockade was efficient in reducing proteinuria in those patients exhibiting substantial mesangial activation of ERK. Thus, ERK activation alters mesangial cell-podocyte crosstalk, leading to renal dysfunction in IgAN. Assessment of MAPK/ERK activation in diagnostic renal biopsies may predict the therapeutic efficacy of renin-angiotensin system blockers in IgAN.


Asunto(s)
Comunicación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glomerulonefritis por IGA/inmunología , Inmunoglobulina A/metabolismo , Sistema de Señalización de MAP Quinasas , Células Mesangiales/inmunología , Podocitos/inmunología , Adulto , Anciano , Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Complejo Antígeno-Anticuerpo , Antígenos CD/metabolismo , Biopsia , Presión Sanguínea , Calcio/metabolismo , Comunicación Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Activación Enzimática , Femenino , Glomerulonefritis por IGA/enzimología , Glomerulonefritis por IGA/patología , Glomerulonefritis por IGA/fisiopatología , Humanos , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Células Mesangiales/efectos de los fármacos , Células Mesangiales/enzimología , Células Mesangiales/patología , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Podocitos/efectos de los fármacos , Podocitos/enzimología , Podocitos/patología , Proteinuria/enzimología , Proteinuria/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Transferrina/metabolismo , Sistema Renina-Angiotensina , Serina-Treonina Quinasas TOR/metabolismo , Factores de Tiempo , Adulto Joven
13.
PLoS Biol ; 7(3): e54, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19278296

RESUMEN

Mucosal-associated invariant T (MAIT) cells display two evolutionarily conserved features: an invariant T cell receptor (TCR)alpha (iTCRalpha) chain and restriction by the nonpolymorphic class Ib major histocompatibility complex (MHC) molecule, MHC-related molecule 1 (MR1). MR1 expression on thymus epithelial cells is not necessary for MAIT cell development but their accumulation in the gut requires MR1 expressing B cells and commensal flora. MAIT cell development is poorly known, as these cells have not been found in the thymus so far. Herein, complementary human and mouse experiments using an anti-humanValpha7.2 antibody and MAIT cell-specific iTCRalpha and TCRbeta transgenic mice in different genetic backgrounds show that MAIT cell development is a stepwise process, with an intra-thymic selection followed by peripheral expansion. Mouse MAIT cells are selected in an MR1-dependent manner both in fetal thymic organ culture and in double iTCRalpha and TCRbeta transgenic RAG knockout mice. In the latter mice, MAIT cells do not expand in the periphery unless B cells are added back by adoptive transfer, showing that B cells are not required for the initial thymic selection step but for the peripheral accumulation. In humans, contrary to natural killer T (NKT) cells, MAIT cells display a naïve phenotype in the thymus as well as in cord blood where they are in low numbers. After birth, MAIT cells acquire a memory phenotype and expand dramatically, up to 1%-4% of blood T cells. Finally, in contrast with NKT cells, human MAIT cell development is independent of the molecular adaptor SAP. Interestingly, mouse MAIT cells display a naïve phenotype and do not express the ZBTB16 transcription factor, which, in contrast, is expressed by NKT cells and the memory human MAIT cells found in the periphery after birth. In conclusion, MAIT cells are selected by MR1 in the thymus on a non-B non-T hematopoietic cell, and acquire a memory phenotype and expand in the periphery in a process dependent both upon B cells and the bacterial flora. Thus, their development follows a unique pattern at the crossroad of NKT and gammadelta T cells.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Inmunidad Mucosa/inmunología , Células T Asesinas Naturales/fisiología , Subgrupos de Linfocitos T/fisiología , Timo/inmunología , Animales , Linfocitos B/fisiología , Niño , Sangre Fetal/inmunología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Receptores de Antígenos de Linfocitos T alfa-beta , Linfocitos T , Timo/citología
14.
EBioMedicine ; 75: 103785, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34959131

RESUMEN

BACKGROUND: Erythrocytosis is a hematological disorder usually related to hematopoietic stem cell somatic mutations. However, unexplained erythrocytosis remains frequent. In this study, we evaluated the involvement of IgA1, a regulator of erythropoiesis also implicated in IgA nephropathy (IgAN) pathophysiology, in unexplained polycythemia/erythrocytosis (PE) of IgAN patients. METHODS: IgAN-PE patients' serum was collected, analyzed and used to study IgA1 effect on proliferation and differentiation of erythroid progenitors. Hematological parameters of transgenic mice for human alpha1 heavy chain were studied. Multicentric observational cohorts of chronic kidney disease (CKD) patients, including both native kidney diseases and renal transplants, were studied to analyze patient hemoglobin levels. FINDINGS: We retrospectively identified 6 patients with IgAN and unexplained PE. In large CKD cohorts, IgAN was associated with PE in 3.5% of patients (p<0.001 compared to other nephropathies). IgAN was an independent factor associated with higher hemoglobin levels (13.1g/dL vs 12.2 g/dL, p=0.01). During post-transplant anemia, anemia recovery was faster in IgAN patients. Elevated polymeric/monomeric IgA1 ratio as well as high Gd-IgA1 rate were observed in circulating IgA1 of the 6 IgAN-PE patients as compared with control or IgAN patients without PE. IgA1 from these patients increased the sensitivity of erythroid progenitors to Epo. In mice, we also observed an elevation of hematocrit in alpha1 knock-in mice compared to wild type controls. INTERPRETATION: These data identify a new etiology of erythrocytosis and demonstrate the role of pIgA1 in human erythropoiesis. This syndrome of IgA-related erythrocytosis should be investigated in case of unexplained erythrocytosis and renal disease. FUNDING: This work was supported by INSERM (French national institute for health and medical research), Labex GRex and Imagine Institute (Paris, France).


Asunto(s)
Glomerulonefritis por IGA , Policitemia , Animales , Biomarcadores , Galactosa , Glomerulonefritis por IGA/complicaciones , Glomerulonefritis por IGA/genética , Humanos , Inmunoglobulina A , Ratones , Policitemia/complicaciones , Policitemia/genética , Estudios Retrospectivos
15.
Kidney Int ; 80(12): 1352-63, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21866091

RESUMEN

Abnormalities of IgA arise in alcoholic cirrhosis, including mesangial IgA deposits with possible development of secondary IgA nephropathy (IgAN). Since little is known about circulating immune complexes in cases of secondary IgAN, we analyzed IgA-associated parameters in the serum of 32 patients with compensated or advanced alcoholic cirrhosis. Galactose deficiency and decreased sialylation of IgA1, as well as increased amounts of abnormally glycosylated polymeric IgA1, were detected in the serum of patients with advanced alcoholic cirrhosis. Moreover, aberrant IgA1 formed complexes with IgG and soluble CD89 in serum of patients with advanced alcoholic cirrhosis, similar to those found in primary IgAN. The IgA1 of alcoholic cirrhosis, however, had a modified N-glycosylation, not found in primary IgAN. In patients with alcoholic cirrhosis and IgAN, IgA deposits were associated with CD71 overexpression in mesangial areas, suggesting that CD71 might be involved in deposit formation. Although the IgA1 found in alcoholic cirrhosis bound more extensively to human mesangial cells than control IgA1, they differ from primary IgAN by not inducing mesangial cell proliferation. Thus, abnormally glycosylated IgA1 and soluble CD89-IgA and IgA-IgG complexes, features of primary IgAN, are also present in alcoholic cirrhosis. Hence, common mechanisms appear to be shared by diseases of distinct origins, indicating that common environmental factors may influence the development of IgAN.


Asunto(s)
Complejo Antígeno-Anticuerpo , Antígenos CD/sangre , Glomerulonefritis por IGA/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Riñón/inmunología , Cirrosis Hepática Alcohólica/inmunología , Receptores Fc/sangre , Adulto , Anciano , Biopsia , Proliferación Celular , Células Cultivadas , Femenino , Galactosa/sangre , Galactosa/deficiencia , Glomerulonefritis por IGA/sangre , Glomerulonefritis por IGA/patología , Glicosilación , Humanos , Riñón/patología , Cirrosis Hepática Alcohólica/sangre , Cirrosis Hepática Alcohólica/complicaciones , Cirrosis Hepática Alcohólica/patología , Masculino , Células Mesangiales/inmunología , Células Mesangiales/patología , Persona de Mediana Edad , Paris
16.
Kidney Int ; 79(11): 1217-27, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21412216

RESUMEN

Focal and segmental glomerulosclerosis (FSGS) is one of the most important causes of end-stage renal failure. The bradykinin B1 receptor has been associated with tissue inflammation and renal fibrosis. To test for a role of the bradykinin B1 receptor in podocyte injury, we pharmacologically modulated its activity at different time points in an adriamycin-induced mouse model of FSGS. Estimated albuminuria and urinary protein to creatinine ratios correlated with podocytopathy. Adriamycin injection led to loss of body weight, proteinuria, and upregulation of B1 receptor mRNA. Early treatment with a B1 antagonist reduced albuminuria and glomerulosclerosis, and inhibited the adriamycin-induced downregulation of podocin, nephrin, and α-actinin-4 expression. Moreover, delayed treatment with antagonist also induced podocyte protection. Conversely, a B1 agonist aggravated renal dysfunction and even further suppressed the levels of podocyte-related molecules. Thus, we propose that kinin has a crucial role in the pathogenesis of FSGS operating through bradykinin B1 receptor signaling.


Asunto(s)
Bradiquinina/análogos & derivados , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Podocitos/efectos de los fármacos , Receptor de Bradiquinina B1/agonistas , Transducción de Señal/efectos de los fármacos , Actinina/metabolismo , Albuminuria/inducido químicamente , Albuminuria/metabolismo , Albuminuria/prevención & control , Animales , Bradiquinina/farmacología , Bradiquinina/toxicidad , Antagonistas del Receptor de Bradiquinina B1 , Modelos Animales de Enfermedad , Doxorrubicina , Regulación de la Expresión Génica/efectos de los fármacos , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Glomeruloesclerosis Focal y Segmentaria/prevención & control , Hemo-Oxigenasa 1/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Podocitos/metabolismo , Podocitos/patología , ARN Mensajero/metabolismo , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B1/metabolismo , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
17.
Cell Rep ; 30(3): 739-754.e4, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31968250

RESUMEN

Vitamin D (VD) is a known differentiating agent, but the role of VD receptor (VDR) is still incompletely described in acute myeloid leukemia (AML), whose treatment is based mostly on antimitotic chemotherapy. Here, we present an unexpected role of VDR in normal hematopoiesis and in leukemogenesis. Limited VDR expression is associated with impaired myeloid progenitor differentiation and is a new prognostic factor in AML. In mice, the lack of Vdr results in increased numbers of hematopoietic and leukemia stem cells and quiescent hematopoietic stem cells. In addition, malignant transformation of Vdr-/- cells results in myeloid differentiation block and increases self-renewal. Vdr promoter is methylated in AML as in CD34+ cells, and demethylating agents induce VDR expression. Association of VDR agonists with hypomethylating agents promotes leukemia stem cell exhaustion and decreases tumor burden in AML mouse models. Thus, Vdr functions as a regulator of stem cell homeostasis and leukemic propagation.


Asunto(s)
Médula Ósea/patología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Receptores de Calcitriol/metabolismo , Animales , Apoptosis/efectos de los fármacos , Azacitidina/farmacología , Médula Ósea/efectos de los fármacos , Recuento de Células , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Progresión de la Enfermedad , Femenino , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/patología , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Células Mieloides/patología , Células Madre Neoplásicas/efectos de los fármacos , Oncogenes , Regiones Promotoras Genéticas/genética , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Ensayo de Tumor de Célula Madre
18.
Cancer Res ; 67(3): 1145-54, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17283149

RESUMEN

Mantle cell lymphoma (MCL) is one of the most frequent of the newly recognized non-Hodgkin's lymphomas. The major problem of MCL therapy is the occurrence of relapse and subsequent resistance to chemotherapy and immunotherapy in virtually all cases. Here, we show that one injection of anti-human transferrin receptor (TfR) monoclonal antibody A24 totally prevented xenografted MCL tumor establishment in nude mice. It also delayed and inhibited tumor progression of established tumors, prolonging mice survival. In vitro, A24 induced up to 85% reduction of MCL cell proliferation (IC(50) = 3.75 nmol/L) independently of antibody aggregation, complement-dependent or antibody-dependent cell-mediated cytotoxicity. A24 induced MCL cell apoptosis through caspase-3 and caspase-9 activation, either alone or synergistically with chemotherapeutic agents. A24 induced TfR endocytosis via the clathrin adaptor protein-2 complex pathway followed by transport to lysosomal compartments. Therefore, A24-based therapies alone or in association with classic chemotherapies could provide a new alternative strategy against MCL, particularly in relapsing cases.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Inmunización Pasiva/métodos , Linfoma de Células del Manto/prevención & control , Lisosomas/metabolismo , Receptores de Transferrina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Endocitosis/efectos de los fármacos , Femenino , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Linfoma de Células del Manto/metabolismo , Linfoma de Células del Manto/patología , Ratones , Ratones Desnudos , Receptores de Transferrina/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Semin Nephrol ; 28(1): 88-95, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18222350

RESUMEN

Compelling evidence points to a role for IgA receptors in the pathogenesis of IgA nephropathy. The soluble form of the type I IgA receptor (FcalphaRI or CD89) forms complexes with IgA that can be found in patients' serum and that initiate the disease in CD89 transgenic mice. A nonclassic IgA receptor, identified as the transferrin receptor (TfR), is highly expressed in patients' mesangium and colocalizes with IgA deposits. TfR preferentially binds polymeric IgA1 complexes, but not monomeric IgA1 or IgA2. The TfR-IgA1 interaction is dependent on carbohydrate moieties because hypoglycosylated IgA1 has superior binding to TfR than normally glycosylated IgA1. Polymeric IgA1 binding enhances mesangial cell TfR expression and results in cell proliferation and inflammatory and profibrogenic cytokine and chemokine production, suggesting a pivotal role in mesangial cell proliferation, matrix expansion, and recruitment of inflammatory cells. We propose that, as a second event, activation of the classic, FcRgamma-associated transmembrane FcalphaRI expressed on circulating myeloid leukocytes takes place. FcalphaRI/gamma2 cross-linking in human FcalphaRI transgenic animals promotes disease progression by enhancing leukocyte chemotaxis and cytokine production, and IgA immune complexes from IgA nephropathy patients induce FcalphaRI-dependent cell activation. This review therefore details the functional consequences of IgA/receptor interactions and discusses proposed mechanisms to explain the development and chronicity of the disease.


Asunto(s)
Glomerulonefritis por IGA/fisiopatología , Inmunoglobulina A/inmunología , Células Mesangiales/inmunología , Animales , Complejo Antígeno-Anticuerpo/efectos adversos , Antígenos CD/inmunología , Modelos Animales de Enfermedad , Glomerulonefritis por IGA/inmunología , Humanos , Inflamación/fisiopatología , Células Mesangiales/patología , Ratones , Receptores Fc/inmunología , Receptores de Transferrina/inmunología
20.
Oncoimmunology ; 7(11): e1470735, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30377560

RESUMEN

Despite increasing evidence for a protective role of invariant (i) NKT cells in the control of graft-versus-host disease (GVHD), the mechanisms underpinning regulation of the allogeneic immune response in humans are not known. In this study, we evaluated the distinct effects of human in vitro expanded and flow-sorted human CD4+ and CD4- iNKT subsets on human T cell activation in a pre-clinical humanized NSG mouse model of xenogeneic GVHD. We demonstrate that human CD4- but not CD4+ iNKT cells could control xenogeneic GVHD, allowing significantly prolonged overall survival and reduced pathological GVHD scores without impairing human T cell engraftment. Human CD4- iNKT cells reduced the activation of human T cells and their Th1 and Th17 differentiation in vivo. CD4- and CD4+ iNKT cells had distinct effects upon DC maturation and survival. Compared to their CD4+ counterparts, in co-culture experiments in vitro, human CD4- iNKT cells had a higher ability to make contacts and degranulate in the presence of mouse bone marrow-derived DCs, inducing their apoptosis. In vivo we observed that infusion of PBMC and CD4- iNKT cells was associated with decreased numbers of splenic mouse CD11c+ DCs. Similar differential effects of the iNKT cell subsets were observed on the maturation and in the induction of apoptosis of human monocyte-derived dendritic cells in vitro. These results highlight the increased immunosuppressive functions of CD4-versus CD4+ human iNKT cells in the context of alloreactivity, and provide a rationale for CD4- iNKT selective expansion or transfer to prevent GVHD in clinical trials.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA