Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22
Filtrer
1.
Nat Commun ; 15(1): 4182, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38755157

RÉSUMÉ

Bone marrow plasma cells (BMPC) are the correlate of humoral immunity, consistently releasing antibodies into the bloodstream. It remains unclear if BMPC reflect different activation environments or maturation of their precursors. Here we define human BMPC heterogeneity and track the recruitment of antibody-secreting cells (ASC) from SARS-CoV-2 vaccine immune reactions to the bone marrow (BM). Trajectories based on single-cell transcriptomes and repertoires of peripheral and BM ASC reveal sequential colonisation of BMPC compartments. In activated B cells, IL-21 suppresses CD19 expression, indicating that CD19low-BMPC are derived from follicular, while CD19high-BMPC originate from extrafollicular immune reactions. In primary immune reactions, both CD19low- and CD19high-BMPC compartments are populated. In secondary immune reactions, most BMPC are recruited to CD19high-BMPC compartments, reflecting their origin from extrafollicular reactivations of memory B cells. A pattern also observable in vaccinated-convalescent individuals and upon diphtheria/tetanus/pertussis recall-vaccination. Thus, BMPC diversity reflects the evolution of a given humoral immune response.


Sujet(s)
Antigènes CD19 , Moelle osseuse , Interleukines , Plasmocytes , Humains , Plasmocytes/immunologie , Interleukines/immunologie , Interleukines/métabolisme , Moelle osseuse/immunologie , Antigènes CD19/immunologie , Antigènes CD19/métabolisme , Immunité humorale/immunologie , COVID-19/immunologie , COVID-19/virologie , SARS-CoV-2/immunologie , Cellules de la moelle osseuse/immunologie , Cellules de la moelle osseuse/cytologie , Analyse sur cellule unique , Adulte , Lymphocytes B/immunologie , Cellules productrices d'anticorps/immunologie , Femelle , Mâle , Vaccination , Adulte d'âge moyen , Vaccin diphtérie-tétanos-coqueluche/immunologie
2.
Front Med (Lausanne) ; 9: 928516, 2022.
Article de Anglais | MEDLINE | ID: mdl-35911418

RÉSUMÉ

Early detection of kidney transplant (KT) rejection remains a challenge in patient care. Non-invasive biomarkers hold high potential to detect rejection, adjust immunosuppression, and monitor KT patients. So far, no approach has fully satisfied requirements to innovate routine monitoring of KT patients. In this two-center study we analyzed a total of 380 urine samples. T cells and tubular epithelial cells were quantified in KT patients with graft deterioration using flow cytometry. Epigenetic urine cell quantification was used to confirm flow cytometric results. Moreover, a cohort of KT patients was followed up during the first year after transplantation, tracking cell subsets over time. Abundance of urinary cell counts differed in patients with and without rejection. Most strikingly, various T cell subsets were enriched in patients with T cell-mediated rejection (TCMR) compared to patients without TCMR. Among T cell subsets, CD8+HLA-DR+ T cells were most distinctive (AUC = 0.91, Spec.: 95.9%, Sens.: 76.5%). Epigenetic analysis confirmed T cell and tubular epithelial cell quantities as determined by flow cytometry. Urinary T cell abundance in new KT patients decreased during their first year after transplantation. In conclusion urinary T cells reflect intrarenal inflammation in TCMR. T cell subsets yield high potential to monitor KT patients and detect rejection. Hereby we present a promising biomarker to non-invasively diagnose TCMR.

3.
Transplant Direct ; 8(5): e1316, 2022 May.
Article de Anglais | MEDLINE | ID: mdl-35434282

RÉSUMÉ

Timely recognition and treatment of acute kidney graft rejection is important to prevent premature graft failure. A predefined urinary marker set for acute T cell-mediated rejection (TCMR) containing 14 peptides was tested for this purpose in a multicenter in-place validation study. Methods: Three hundred twenty-nine prospectively collected and 306 archived urine samples from 11 transplant centers in Germany, France, and Belgium were examined. Samples were taken immediately before a biopsy, performed for graft dysfunction within the first transplant year. Primary outcomes were sensitivity and specificity of the marker set for the diagnosis of biopsy-proven acute TCMR, with prespecified thresholds of 83% for sensitivity and 70% for specificity. Results: Eighty-two patients (13%) had acute TCMR grade I-III. In relation to the biopsy diagnosis of TCMR, the sensitivity of the urine test was 0.66 (95% confidence interval, 0.56-0.76) and the specificity 0.47 (95% confidence interval, 0.43-0.51), with an area under the curve (AUC) of 0.60. The different TCMR grades I-III were not reflected by the marker set, and borderline TCMR was not specifically detected. Secondary independent masked assessment of biopsies consented by 2 pathologists revealed an interobserver kappa value of 0.49 for diagnosing TCMR, compared with the local center's diagnosis. Using this consensus diagnosis, the AUC of the urine test was 0.63 (sensitivity 0.73, specificity 0.45). Post hoc optimization of the marker set improved the diagnostic performance in the study cohort (AUC 0.67) and in an independent patient cohort (AUC 0.69). Conclusions: This study illustrates the difficulty of proteomics-based diagnosis of TCMR and highlights the need for rigorous independent in-place validation and optimization of diagnostic biomarkers.

4.
Nat Commun ; 12(1): 1961, 2021 03 30.
Article de Anglais | MEDLINE | ID: mdl-33785765

RÉSUMÉ

The pathogenesis of severe COVID-19 reflects an inefficient immune reaction to SARS-CoV-2. Here we analyze, at the single cell level, plasmablasts egressed into the blood to study the dynamics of adaptive immune response in COVID-19 patients requiring intensive care. Before seroconversion in response to SARS-CoV-2 spike protein, peripheral plasmablasts display a type 1 interferon-induced gene expression signature; however, following seroconversion, plasmablasts lose this signature, express instead gene signatures induced by IL-21 and TGF-ß, and produce mostly IgG1 and IgA1. In the sustained immune reaction from COVID-19 patients, plasmablasts shift to the expression of IgA2, thereby reflecting an instruction by TGF-ß. Despite their continued presence in the blood, plasmablasts are not found in the lungs of deceased COVID-19 patients, nor does patient IgA2 binds to the dominant antigens of SARS-CoV-2. Our results thus suggest that, in severe COVID-19, SARS-CoV-2 triggers a chronic immune reaction that is instructed by TGF-ß, and is distracted from itself.


Sujet(s)
Anticorps antiviraux/immunologie , COVID-19/immunologie , SARS-CoV-2/immunologie , Facteur de croissance transformant bêta/immunologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , COVID-19/virologie , Femelle , Humains , Immunoglobuline A/immunologie , Immunoglobuline G/immunologie , Interleukines/immunologie , Mâle , Adulte d'âge moyen , Plasmocytes/immunologie , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/immunologie
5.
Eur J Immunol ; 49(9): 1372-1379, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31149730

RÉSUMÉ

Bone marrow (BM) stromal cells are important in the development and maintenance of cells of the immune system. Using single cell RNA sequencing, we here explore the functional and phenotypic heterogeneity of individual transcriptomes of 1167 murine BM mesenchymal stromal cells. These cells exhibit a tremendous heterogeneity of gene expression, which precludes the identification of defined subpopulations. However, according to the expression of 108 genes involved in the communication of stromal cells with hematopoietic cells, we have identified 14 non-overlapping subpopulations, with distinct cytokine or chemokine gene expression signatures. With respect to the maintenance of subsets of immune memory cells by stromal cells, we identified distinct subpopulations expressing Il7, Il15 and Tnfsf13b. Together, this study provides a comprehensive dissection of the BM stromal heterogeneity at the single cell transcriptome level and provides a basis to understand their lifestyle and their role as organizers of niches for the long-term maintenance of immune cells.


Sujet(s)
Cellules de la moelle osseuse/cytologie , Moelle osseuse/physiologie , Cellules stromales/cytologie , Transcriptome/génétique , Animaux , Facteur d'activation des lymphocytes B/génétique , Cellules cultivées , Cytokines/génétique , Cellules souches hématopoïétiques/cytologie , Interleukine-15/génétique , Interleukine-7/génétique , Cellules souches mésenchymateuses/cytologie , Souris , Souris de lignée C57BL , Analyse de séquence d'ARN/méthodes
6.
Clin Transplant ; 32(12): e13429, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30341925

RÉSUMÉ

CONTEXT: Antibody-mediated rejection (ABMR) after kidney transplantation (KTx) remains the crucial obstacle to successful long-term graft function. The identification of gene signatures involved in ABMR could grant the basis for better prevention and treatment strategies. OBJECTIVE: The identification of gene signatures in whole blood cells specific for ABMR after KTx. MATERIALS AND METHODS: Total RNA from blood cells of 16 kidney-transplanted patients with ABMR, stable graft function (SGF), and with T-cell-mediated rejection (TCMR) was isolated. Gene expression was determined by high-throughput sequencing followed by validation and analyses of differentially expressed candidates on mRNA level and on protein level in a large patient cohort (n = 185) in patients with SGF, urinary tract infection (UTI), borderline rejection (BL), TCMR, ABMR, and interstitial fibrosis and tubular atrophy. RESULTS: From the 570 genes detected, 111 discriminated ABMR from SGF and TCMR. A distinct enrichment of interferon (IFN) type I and type II signature gene set was observed. The expression of candidate genes IFIT1, ETV7, and RSAD2 distinguished ABMR patients from patients with SGF and also TCMR, whereas ETV7 and RSAD2 differentiated ABMR also from BL. CONCLUSION: The IFN-inducible genes ETV7 and RSAD2 represent specific biomarkers for ABMR episodes after KTx.


Sujet(s)
Marqueurs biologiques/analyse , Rejet du greffon/diagnostic , Alloanticorps/effets indésirables , Transplantation rénale/effets indésirables , Complications postopératoires/diagnostic , Protéines/génétique , Protéines proto-oncogènes c-ets/génétique , Études cas-témoins , Études de cohortes , Études de suivi , Analyse de profil d'expression de gènes , Rejet du greffon/étiologie , Rejet du greffon/génétique , Survie du greffon , Humains , Défaillance rénale chronique/chirurgie , Oxidoreductases acting on CH-CH group donors , Complications postopératoires/étiologie , Complications postopératoires/génétique , Pronostic , Protéines/métabolisme , Protéines proto-oncogènes c-ets/métabolisme , Facteurs de risque
7.
PLoS One ; 13(8): e0201925, 2018.
Article de Anglais | MEDLINE | ID: mdl-30102719

RÉSUMÉ

Interstitial fibrosis/tubular atrophy (IFTA) is associated with reduced allograft survival, whereas antibody-mediated rejection (ABMR) is the major cause for renal allograft failure. To identify specific microRNAs and their regulation involved in these processes, total RNA from blood cells of 16 kidney transplanted (KTx) patients with ABMR, stable graft function (SGF) and with T-cell mediated rejection (TCMR) was isolated. MicroRNA expression was determined by high-throughput sequencing. Differentially expressed candidate microRNAs were analyzed with RT-PCR in patients with SGF (n = 53), urinary tract infection (UTI) (n = 17), borderline rejection (BL) (n = 19), TCMR (n = 40), ABMR (n = 22) and IFTA (n = 30). From the 301 detected microRNAs, 64 were significantly regulated between the three cohorts. Selected candidate microRNAs miR-223-3p, miR-424-3p and miR-145-5p distinguished TCMR and ABMR from SGF, but not from other pathologies. Most importantly, miR-145-5p expression in IFTA patients was significantly downregulated and displayed a high diagnostic accuracy compared to SGF alone (AUC = 0.891) and compared to SGF, UTI, BL, TCMR and ABMR patients combined (AUC = 0.835), which was verified by cross-validation. The identification of miR-145-5p as IFTA specific marker in blood constitutes the basis for evaluating this potentially diagnostic microRNA as biomarker in studies including high numbers of patients and different pathologies and also the further analysis of fibrosis causing etiologies after kidney transplantation.


Sujet(s)
Cellules sanguines/métabolisme , Régulation de l'expression des gènes , Rejet du greffon/étiologie , Maladies du rein/étiologie , Transplantation rénale/effets indésirables , microARN/génétique , Atrophie , Femelle , Fibrose , Analyse de profil d'expression de gènes , Rejet du greffon/anatomopathologie , Séquençage nucléotidique à haut débit , Humains , Maladies du rein/anatomopathologie , Tubules rénaux/anatomopathologie , Mâle , Courbe ROC , Reproductibilité des résultats , Transcriptome , Transplantation homologue
8.
Transpl Immunol ; 39: 18-24, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27693310

RÉSUMÉ

Cellular and antibody-mediated rejection processes and also interstitial fibrosis/tubular atrophy (IFTA) lead to allograft dysfunction and loss. The search for accurate, specific and non-invasive diagnostic tools is still ongoing and essential for successful treatment of renal transplanted patients. Molecular markers in blood cells and serum may serve as diagnostic tools but studies with high patient numbers and differential groups are rare. We validated the potential value of several markers on mRNA level in blood cells and serum protein level in 166 samples from kidney transplanted patients under standard immunosuppressive therapy (steroids±mycophenolic acid±calcineurin inhibitor) with stable graft function, urinary tract infection (UTI), IFTA, antibody-mediated rejection (ABMR), and T-cell-mediated rejection (TCMR) applying RT-PCR and ELISA. The mRNA expression of RANTES, granulysin, granzyme-B, IP-10, Mic-A and Interferon-γ in blood cells did not distinguish specifically between the different pathologies. We furthermore discovered that the mRNA expression of the chemokine IL-8 is significantly lower in samples from IFTA patients than in samples from patients with stable graft function (p<0.001), ABMR (p<0.001), Borderline (BL) TCMR (p<0.001), tubulo-interstitial TCMR (p<0.001) and vascular TCMR (p<0.01), but not with UTI. Serum protein concentrations of granzyme-B, Interferon-γ and IL-8 did not differ between the patient groups, RANTES concentration was significantly different when comparing UTI and ABMR (p<0.01), whereas granulysin, Mic-A and IP-10 measurement differentiated ongoing rejection or IFTA processes from stable graft function but not from each other. The measurement of IL-8 mRNA in blood cells distinguishes clearly between IFTA and other complication after kidney transplantation and could easily be used as diagnostic tool in the clinic.


Sujet(s)
Cellules sanguines/immunologie , Rejet du greffon/diagnostic , Interleukine-8/métabolisme , Transplantation rénale , Rein/anatomopathologie , Lymphocytes T/immunologie , Infections urinaires/diagnostic , Adulte , Sujet âgé , Animaux , Atrophie , Marqueurs biologiques/métabolisme , Inhibiteurs de la calcineurine/usage thérapeutique , Diagnostic différentiel , Fibrose , Rejet du greffon/traitement médicamenteux , Humains , Tolérance immunitaire , Interleukine-8/génétique , Alloanticorps/métabolisme , Rein/immunologie , Souris , Adulte d'âge moyen , Acide mycophénolique/usage thérapeutique , Stéroïdes/usage thérapeutique , Infections urinaires/traitement médicamenteux
9.
Transpl Immunol ; 39: 52-59, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27663089

RÉSUMÉ

The potential diagnostic value of circulating free miRNAs in plasma compared to miRNA expression in blood cells for rejection processes after kidney transplantation is largely unknown, but offers the potential for better and timely diagnosis of acute rejection. Free microRNA expression of specific blood cell markers was measured in 160 plasma samples from kidney transplant patients under standard immunosuppressive therapy (steroids±mycophenolic acid±calcineurin inhibitor) with stable graft function, urinary tract infection, interstitial fibrosis and tubular atrophy, antibody-mediated rejection (ABMR), Borderline (Banff3), tubulo-interstitial (Banff4-I) and vascular rejection (Banff4-II/III) applying RT-PCR. The expression levels of specific microRNAs miR-15B, miR-103A and miR-106A discriminated patients with stable graft function significantly (p-values 0.001996, 0.0054 and 0.0019 resp.) from patients with T-cell mediated rejection (TCMR) and from patients with urinary tract infection (p-values 0.0001, <0.0001 and 0.0001, resp.). A combined measurement of several microRNAs after multivariate logistic regression improved the diagnostic value supported by subsequent cross-validation. In conclusion, the measurement of circulating microRNAs in plasma from patients with renal transplants distinguishes TCMR and urinary tract infection from stable graft function. In contrast to miRNA expression measurement in blood cells it does not allow a discrimination from ABMR or interstitial fibrosis and tubular atrophy.


Sujet(s)
Marqueurs biologiques/sang , Rejet du greffon/diagnostic , Transplantation rénale , Rein/anatomopathologie , microARN/sang , Lymphocytes T/immunologie , Infections urinaires/diagnostic , Adulte , Sujet âgé , Atrophie , Inhibiteurs de la calcineurine/usage thérapeutique , Diagnostic différentiel , Femelle , Fibrose , Rejet du greffon/traitement médicamenteux , Humains , Rein/immunologie , Mâle , Adulte d'âge moyen , Acide mycophénolique/usage thérapeutique , Valeur prédictive des tests , Pronostic , Stéroïdes/usage thérapeutique , Tolérance à la transplantation , Infections urinaires/traitement médicamenteux
10.
Transplantation ; 100(4): 898-907, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26444957

RÉSUMÉ

BACKGROUND: MicroRNAs (miRNAs, miR) hold important roles in the posttranscriptional regulation of gene expression. Their function has been correlated with kidney disease, and they might represent a new class of biomarkers for frequent evaluation of renal graft status. We analyzed their potential in identifying severe T cell-mediated vascular rejection (TCMVR) (Banff 4-II/III) in kidney transplanted patients. METHODS: Microarray experiments and semiquantitative real-time reverse transcription polymerase chain reaction were performed with total RNA isolated from blood cells of kidney graft recipients. Initial microarray analysis revealed 23 differentially expressed miRNAs distinguishing patients with TCMVR from patients with stable grafts. From these, we validated and further determined the expression of 6 differentially expressed miRNAs and 2 control miRNAs in 161 samples from patients with T cell-mediated rejection (Banff 3-Borderline, Banff 4-I/II/III), Banff-2 antibody-mediated rejection, Banff-5 interstitial fibrosis/tubular atrophy, in samples from stable patients and in samples from patients with urinary tract infection using real-time reverse transcription polymerase chain reaction. RESULTS: Expression levels of all 6 candidate miRNAs were significantly downregulated in blood of TCMVR patients compared to the other groups and displayed high sensitivities and specificities for diagnosing TCMVR. The combination of 5 miRNAs, identified by an unbiased multivariate logistic regression followed by cross-validation, enhanced the sensitivity and specificity for the diagnosis of TCMVR after renal transplantation. CONCLUSIONS: The combined measurement of miRNA-15B, miRNA-16, miRNA-103A, miRNA-106A, and miRNA-107 may help to better identify TCMVR after renal transplantation in a precise and clinically applicable way.


Sujet(s)
Rejet du greffon/sang , Rejet du greffon/génétique , Immunité cellulaire/génétique , Transplantation rénale/effets indésirables , microARN/sang , microARN/génétique , Lymphocytes T/immunologie , Aire sous la courbe , Analyse de regroupements , Régulation négative , Analyse de profil d'expression de gènes/méthodes , Marqueurs génétiques , Rejet du greffon/diagnostic , Rejet du greffon/immunologie , Humains , Modèles logistiques , Analyse multifactorielle , Séquençage par oligonucléotides en batterie , Valeur prédictive des tests , Courbe ROC , Réaction de polymérisation en chaine en temps réel , Reproductibilité des résultats , RT-PCR , Facteurs de risque , Résultat thérapeutique
11.
Transpl Immunol ; 33(3): 176-84, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26478531

RÉSUMÉ

Belatacept offers a new option for renal allograft recipients who are suffering from side effects of calcineurin inhibitors or mTOR inhibitors,which may result in renal and extrarenal benefits.We prospectively performed flow cytometric immunophenotyping with a T-cell panel. In total we were able to fully investigate the immunophenotypic change in 8 patients before and after conversion from calcineurin inhibitor (n = 5) or mTOR inhibitor (n=2) to Belatacept or additional administration (n=1). Cells were analysed pre conversion, 1 month, 3 months, 6 months and 12 months after first Belatacept administration. The percentage of central memory, naïve, effector memory and terminally differentiated effector memory CD4+ and CD4− T-cells was determined. CD28, CD25 and CD69 expression on CD4+ and CD4− T-cells was measured ex vivo and also after 3 days of mitogen stimulation. Intracellular cytokines IFNgamma and IL-2 were measured after polyclonal cellular stimulation. The expression of activation markers and intracellular cytokines as well as the percentage of T-cell subsets did not change significantly during the observation period compared to the time-point pre conversion. Therefore the conversion of calcineurin inhibitor or mTOR inhibitor to Belatacept seems to have no obvious impact on the immunophenotype of T-cells in patients after kidney transplantation.


Sujet(s)
Abatacept/usage thérapeutique , Rejet du greffon/traitement médicamenteux , Transplantation rénale , Sous-populations de lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/effets des médicaments et des substances chimiques , Antigènes CD/métabolisme , Inhibiteurs de la calcineurine/usage thérapeutique , Séparation cellulaire , Substitution de médicament , Cytométrie en flux , Rejet du greffon/étiologie , Humains , Mémoire immunologique , Immunophénotypage , Immunosuppresseurs/usage thérapeutique , Activation des lymphocytes , Études prospectives , Lymphocytes T/immunologie , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs
12.
Eur J Immunol ; 45(4): 1192-205, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25486906

RÉSUMÉ

Repeatedly activated T helper 1 (Th1) cells present during chronic inflammation can efficiently adapt to the inflammatory milieu, for example, by expressing the transcription factor Twist1, which limits the immunopathology caused by Th1 cells. Here, we show that in repeatedly activated murine Th1 cells, Twist1 and T-bet induce expression of microRNA-148a (miR-148a). miR-148a regulates expression of the proapoptotic gene Bim, resulting in a decreased Bim/Bcl2 ratio. Inhibition of miR-148a by antagomirs in repeatedly activated Th1 cells increases the expression of Bim, leading to enhanced apoptosis. Knockdown of Bim expression by siRNA in miR-148a antagomir-treated cells restores viability of the Th1 cells, demonstrating that miR-148a controls survival by regulating Bim expression. Thus, Twist1 and T-bet not only control the differentiation and function of Th1 cells, but also their persistence in chronic inflammation.


Sujet(s)
Protéines régulatrices de l'apoptose/génétique , Apoptose/génétique , Régulation de l'expression des gènes , Protéines membranaires/génétique , microARN/physiologie , Protéines nucléaires/métabolisme , Protéines proto-oncogènes/génétique , Protéines à domaine boîte-T/physiologie , Lymphocytes auxiliaires Th1/immunologie , Protéine-1 apparentée à Twist/métabolisme , Animaux , Polyarthrite rhumatoïde/immunologie , Protéine-11 analogue à Bcl-2 , Survie cellulaire/immunologie , Cellules cultivées , Humains , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris knockout , microARN/antagonistes et inhibiteurs , microARN/génétique , Protéines nucléaires/génétique , Interférence par ARN , Petit ARN interférent , Protéines à domaine boîte-T/génétique , Protéine-1 apparentée à Twist/génétique
13.
Int Immunopharmacol ; 15(4): 718-25, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23499640

RÉSUMÉ

A considerable fraction of renal transplanted patients is susceptible to humoral rejection. Today well-established therapy regimens are available to control antibody-mediated rejection in the short term. Nevertheless, donor-specific antibodies persist and graft function deteriorates over time. This might be due to insufficient maintenance immunosuppression - which always consists of two to three drugs with different mechanisms of action. Since T- and B-cell functions always depend on each other in the alloimmune response it is of interest to analyze the effects of combined standard and new immunosuppressive substances with T-cell inhibitory properties on B-cell function. The effectiveness of complementary administrations of sotrastaurin, mycophenolic acid and everolimus on the activation and function of human primary B-lymphocytes was tested. Everolimus and mycophenolic acid alone and in combination proved to be highly effective in suppressing B-cell activation, whereas the proteinkinase C inhibitor sotrastaurin had an unexpected and reverse impact on various B-cell functions when applied in combination with the mammalian target of rapamycin and the inosine monophosphate dehydrogenase inhibitor.


Sujet(s)
Lymphocytes B/effets des médicaments et des substances chimiques , Immunosuppression thérapeutique/normes , Immunosuppresseurs/pharmacologie , Acide mycophénolique/pharmacologie , Pyrroles/pharmacologie , Quinazolines/pharmacologie , Sirolimus/analogues et dérivés , Apoptose/effets des médicaments et des substances chimiques , Apoptose/immunologie , Lymphocytes B/immunologie , Antigène CD80/biosynthèse , Antigène CD80/immunologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytokines/immunologie , Relation dose-effet des médicaments , Synergie des médicaments , Association de médicaments , Évérolimus , Humains , Immunité humorale/effets des médicaments et des substances chimiques , Immunoglobuline G/biosynthèse , Immunoglobuline G/immunologie , Immunosuppression thérapeutique/méthodes , Immunosuppresseurs/administration et posologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Acide mycophénolique/administration et posologie , Culture de cellules primaires , Pyrroles/administration et posologie , Quinazolines/administration et posologie , Sirolimus/administration et posologie , Sirolimus/pharmacologie
14.
Expert Opin Drug Metab Toxicol ; 8(10): 1337-56, 2012 Oct.
Article de Anglais | MEDLINE | ID: mdl-22928953

RÉSUMÉ

INTRODUCTION: Sirolimus is a powerful antiproliferative immunosuppressive drug approved for the prevention of kidney allograft rejection. By its unique mechanism of action, sirolimus provides a multitude of clinical potential and has been used effectively in different drug combinations. Extensive experience has been gained regarding the best timing of its application, side effect profile and potential benefits and limitations compared with other immunosuppressive drugs. AREAS COVERED: The authors evaluate the recent experience with sirolimus in kidney transplantation. Pivotal randomized controlled trials were used to provide an overview with special attention to pharmacokinetic and dynamic aspects of sirolimus, its current clinical use as well as perspectives for its future role. EXPERT OPINION: Sirolimus enriches the possibilities of immunosuppressive therapies after renal transplantation. Beneficial effects toward kidney function by allowing CNI sparing, lower incidence of malignancies and less viral infections have been suggested. Sirolimus should be used cautiously in de novo patients for reasons of wound healing. An early conversion to a sirolimus-based CNI-free regimen has shown promising results, whereas late conversion is more challenging. Finally, sirolimus-associated side effects are causing tolerability concerns and frequent discontinuations. Future research should aim to better define the therapeutic window and those patients most likely to benefit.


Sujet(s)
Rejet du greffon/prévention et contrôle , Immunosuppresseurs/usage thérapeutique , Transplantation rénale , Rein/effets des médicaments et des substances chimiques , Sirolimus/pharmacocinétique , Sirolimus/usage thérapeutique , Calcineurine/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Immunosuppresseurs/effets indésirables , Immunosuppresseurs/pharmacocinétique , Rein/cytologie , Guides de bonnes pratiques cliniques comme sujet , Essais contrôlés randomisés comme sujet , Sirolimus/effets indésirables
15.
Drugs ; 72(10): 1335-54, 2012 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-22747448

RÉSUMÉ

Sensitization is generally referred to as the development of alloantibodies, specifically anti-human leukocyte antigen (HLA) immunoglobulin G (IgG) antibodies, most commonly caused by pregnancy, blood transfusion or a previous transplant. Despite being a well known phenomenon, there has not been a general consensus on its definition, monitoring or management. Today, 25% of the patients waitlisted for kidney transplant in the US have a panel reactive antibody (PRA) of >10% while, in the Eurotransplant zone, 14% have a PRA of >5%. Sensitized patients have more difficulty in finding a well HLA-matched donor, and have a higher risk of experiencing longer waiting times, more rejection episodes and eventually inferior long-term graft or patient survival. We review the currently available strategies in identifying and managing highly sensitized patients undergoing renal transplantation. We discuss the progress and limitations in laboratory techniques to elaborate on challenges in defining sensitized patients. The main management options (i.e. the Acceptable Mismatch Program, donor exchange programmes and the desensitization approach) and their mechanisms, related policies, advantages and outcomes, as well as medications and methods being investigated, are updated. In addition, particular emphasis is given to sensitization prevention, a practice that is neglected with our increasing ability to suppress the immune system.


Sujet(s)
Incompatibilité sanguine , Survie du greffon/immunologie , Antigènes HLA/immunologie , Alloanticorps/sang , Transplantation rénale/immunologie , Incompatibilité sanguine/diagnostic , Incompatibilité sanguine/prévention et contrôle , Désensibilisation immunologique , Test d'histocompatibilité , Humains , Plasmaphérèse , Donneurs de tissus
16.
Transpl Int ; 25(10): 1106-16, 2012 Oct.
Article de Anglais | MEDLINE | ID: mdl-22816666

RÉSUMÉ

Humoral rejection processes may lead to allograft injury and subsequent dysfunction. Today, only one B-cell-specific agent is in clinical use and the effects of standard and new immunosuppressant substances on B-cell activation and function are not fully clarified. The impact of sotrastaurin, mycophenolic acid and everolimus on human B-lymphocyte function was assessed by analysing proliferation, apoptosis, CD80/CD86 expression and immunoglobulin and IL-10 production in primary stimulated B cells. In addition, B-cell co-cultures with pre-activated T cells were performed to evaluate the effect of the different immunosuppressive agents on T-cell-dependent immunoglobulin production. Sotrastaurin did not inhibit B-cell proliferation, CD80/CD86 expression, and IgG production and had only minor effects on IgM levels at the highest concentration administered. In contrast, mycophenolic acid and everolimus had strong effects on all B-cell functions in a dose-dependent manner. All immunosuppressive agents caused decreased immunoglobulin levels in T-cell-dependent B-cell cultures. The data provided here suggest that mycophenolic acid and everolimus, but not sotrastaurin, are potent inhibitors of human B-lymphocyte function and activation.


Sujet(s)
Lymphocytes B/immunologie , Acide mycophénolique/usage thérapeutique , Pyrroles/usage thérapeutique , Quinazolines/usage thérapeutique , Sirolimus/analogues et dérivés , Apoptose , Lymphocytes B/effets des médicaments et des substances chimiques , Antigène CD80/biosynthèse , Antigène CD86/biosynthèse , Membrane cellulaire/métabolisme , Prolifération cellulaire , Relation dose-effet des médicaments , Antienzymes/pharmacologie , Évérolimus , Rejet du greffon , Humains , Immunité humorale , Immunoglobuline G/métabolisme , Immunoglobuline M/métabolisme , Immunoglobulines/métabolisme , Immunosuppresseurs/pharmacologie , Immunosuppresseurs/usage thérapeutique , Interleukine-10/métabolisme , Agranulocytes/cytologie , Sirolimus/usage thérapeutique , Transplantation homologue
18.
Ann N Y Acad Sci ; 1247: 46-55, 2012 Jan.
Article de Anglais | MEDLINE | ID: mdl-22236474

RÉSUMÉ

The Forkhead box O (FoxO) family of transcription factors is important for the maintenance of immunological homeostasis and tolerance by controlling the development and function of B and T lymphocytes. Because dysregulation in FoxO activity can result in chronic inflammation and autoimmunity, the transcriptional activity of FoxO proteins is tightly controlled and generally dependent on complex posttranslational modifications that lead either to their nuclear entry and subsequent activation or, alternatively, to their nuclear export. The phosphatidylinositol 3-kinase (PI3K)-protein kinase B (PKB/Akt) axis represents the major pathway phosphorylating and thereby inactivating FoxO proteins. However, recent results have revealed an additional posttranscriptional mechanism of FoxO inactivation by microRNAs. The discovery of this molecular pathway may provide a new therapeutic avenue for the modulation of FoxO activity in immune-mediated diseases using either microRNA targeting antagomirs or synthetic microRNA mimics, a topic that is addressed in this review.


Sujet(s)
Facteurs de transcription Forkhead/génétique , Lymphocytes/métabolisme , microARN/métabolisme , Transduction du signal , Immunité acquise/génétique , Animaux , Facteurs de transcription Forkhead/métabolisme , Humains , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Maturation post-transcriptionnelle des ARN
19.
Expert Opin Drug Metab Toxicol ; 7(1): 103-13, 2011 Jan.
Article de Anglais | MEDLINE | ID: mdl-21142580

RÉSUMÉ

IMPORTANCE OF THE FIELD: The prevalence of acute renal allograft rejection has decreased substantially in past decades due to new and more specific immunosuppressive compounds but improvements in long-term graft function have not been achieved. There is a large need for new immunosuppressive agents that lack toxicity of current agents such as calcineurin inhibitors but show high synergistic efficiency in preventing rejection processes. AREAS COVERED IN THIS REVIEW: This review summarizes data concerning the pharmacokinetics, pharmacodynamics and clinical efficacy of the new PKC inhibitor sotrastaurin with a focus on renal transplantation. The article contains information that has been presented at international transplant meetings and congresses and that has been published between 2006 and 2010. Additionally, current ongoing trials are described in detail. WHAT THE READER WILL GAIN: Immunosuppressive regimens after kidney transplantation consist of a combination of several agents in order to minimize drug toxicity. Therefore, the reader is presented with the most up-to-date/current developments in sotrastaurin applications in Phase I and II trials with emphasis on data maintained from studies that combined sotrastaurin with established agents such as mycophenolic acid and tacrolimus. TAKE HOME MESSAGE: Several trials are ongoing and planned to determine the optimal immunosuppressive regimen to benefit from sotrastaurin's distinct mechanism of action.


Sujet(s)
Évaluation de médicament , Rejet du greffon/prévention et contrôle , Immunosuppresseurs/pharmacologie , Transplantation rénale , Inhibiteurs de protéines kinases/pharmacologie , Pyrroles/pharmacocinétique , Quinazolines/pharmacocinétique , Inhibiteurs de la calcineurine , Essais cliniques de phase I comme sujet , Essais cliniques de phase II comme sujet , Ciclosporine/pharmacologie , Association de médicaments , Effets secondaires indésirables des médicaments/métabolisme , Humains , Acide mycophénolique/pharmacocinétique , Protéine kinase C/antagonistes et inhibiteurs , Sirolimus/pharmacologie , Tacrolimus/pharmacocinétique , Résultat thérapeutique
20.
Nat Immunol ; 11(11): 1057-62, 2010 Nov.
Article de Anglais | MEDLINE | ID: mdl-20935646

RÉSUMÉ

After being activated by antigen, helper T lymphocytes switch from a resting state to clonal expansion. This switch requires inactivation of the transcription factor Foxo1, a suppressor of proliferation expressed in resting helper T lymphocytes. In the early antigen-dependent phase of expansion, Foxo1 is inactivated by antigen receptor-mediated post-translational modifications. Here we show that in the late phase of expansion, Foxo1 was no longer post-translationally regulated but was inhibited post-transcriptionally by the interleukin 2 (IL-2)-induced microRNA miR-182. Specific inhibition of miR-182 in helper T lymphocytes limited their population expansion in vitro and in vivo. Our results demonstrate a central role for miR-182 in the physiological regulation of IL-2-driven helper T cell-mediated immune responses and open new therapeutic possibilities.


Sujet(s)
Interleukine-2/immunologie , microARN/immunologie , Lymphocytes T auxiliaires/cytologie , Lymphocytes T auxiliaires/immunologie , Animaux , Arthrite/immunologie , Prolifération cellulaire , Cellules cultivées , Modèles animaux de maladie humaine , Humains , Souris , Souris de lignée BALB C , Souris de lignée C57BL
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...