Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.904
Filtrer
1.
Front Immunol ; 15: 1409945, 2024.
Article de Anglais | MEDLINE | ID: mdl-39072320

RÉSUMÉ

In this study, we try to find the pathogenic role of immune-related genes in the bone marrow microenvironment of AML. Through WGCNA, seven modules were obtained, among which the turquoise module containing 1793 genes was highly correlated with the immune infiltration score. By unsupervised clustering, the turquoise module was divided into two clusters: the intersection of clinically significant genes in the TCGA and DEGs to obtain 178 genes for mutation analysis, followed by obtaining 17 genes with high mutation frequency. Subsequently, these 17 genes were subjected to LASSO regression analysis to construct a riskscore model of 8 hub genes. The TIMER database, ImmuCellAI portal website, and ssGSEA elucidate that the hub genes and risk scores are closely related to immune cell infiltration into the bone marrow microenvironment. In addition, we also validated the relative expression levels of hub genes using the TCGA database and GSE114868, and additional expression levels of hub genes in AML cell lines in vitro. Therefore, we constructed an immune infiltration-related gene model that identify 8 hub genes with good risk stratification and predictive prognosis for AML.


Sujet(s)
Moelle osseuse , Leucémie aigüe myéloïde , Microenvironnement tumoral , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/immunologie , Humains , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Moelle osseuse/immunologie , Moelle osseuse/anatomopathologie , Analyse de profil d'expression de gènes , Évolution de la maladie , Bases de données génétiques , Réseaux de régulation génique , Régulation de l'expression des gènes dans la leucémie , Marqueurs biologiques tumoraux/génétique , Pronostic , Transcriptome , Biologie informatique/méthodes , Mutation
2.
Nat Commun ; 15(1): 4691, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38824171

RÉSUMÉ

Self-reactive and polyreactive B cells generated during B cell development are silenced by either apoptosis, clonal deletion, receptor editing or anergy to avoid autoimmunity. The specific contribution of apoptosis to normal B cell development and self-tolerance is incompletely understood. Here, we quantify self-reactivity, polyreactivity and apoptosis during physiologic B lymphocyte development. Self-reactivity and polyreactivity are most abundant in early immature B cells and diminish significantly during maturation within the bone marrow. Minimal apoptosis still occurs at this site, however B cell receptors cloned from apoptotic B cells show comparable self-reactivity to that of viable cells. Apoptosis increases dramatically only following immature B cells leaving the bone marrow sinusoids, but above 90% of cloned apoptotic transitional B cells are not self-reactive/polyreactive. Our data suggests that an apoptosis-independent mechanism, such as receptor editing, removes most self-reactive B cells in the bone marrow. Mechanistically, lack of survival signaling rather than clonal deletion appears to be the underpinning cause of apoptosis in most transitional B cells in the periphery.


Sujet(s)
Apoptose , Lymphocytes B , Délétion clonale , Souris de lignée C57BL , Animaux , Apoptose/immunologie , Délétion clonale/immunologie , Lymphocytes B/immunologie , Souris , Récepteurs pour l'antigène des lymphocytes B/métabolisme , Récepteurs pour l'antigène des lymphocytes B/immunologie , Récepteurs pour l'antigène des lymphocytes B/génétique , Différenciation cellulaire/immunologie , Moelle osseuse/immunologie , Femelle , Précurseurs lymphoïdes B/immunologie
3.
Commun Biol ; 7(1): 769, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918571

RÉSUMÉ

Innate lymphoid cells (ILCs) are largely tissue-resident, mostly described within the mucosal tissues. However, their presence and functions in the human draining lymph nodes (LNs) are unknown. Our study unravels the tissue-specific transcriptional profiles of 47,287 CD127+ ILCs within the human abdominal and thoracic LNs. LNs contain a higher frequency of CD127+ ILCs than in BM or spleen. We define independent stages of ILC development, including EILP and pILC in the BM. These progenitors exist in LNs in addition to naïve ILCs (nILCs) that can differentiate into mature ILCs. We define three ILC1 and four ILC3 sub-clusters in the LNs. ILC1 and ILC3 subsets have clusters with high heat shock protein-encoding genes. We identify previously unrecognized regulons, including the BACH2 family for ILC1 and the ATF family for ILC3. Our study is the comprehensive characterization of ILCs in LNs, providing an in-depth understanding of ILC-mediated immunity in humans.


Sujet(s)
Immunité innée , Noeuds lymphatiques , Lymphocytes , Rate , Transcriptome , Humains , Immunité innée/génétique , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/cytologie , Lymphocytes/immunologie , Lymphocytes/métabolisme , Rate/immunologie , Rate/cytologie , Moelle osseuse/immunologie , Analyse de profil d'expression de gènes , Mâle
4.
Cell Rep Med ; 5(6): 101584, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38776911

RÉSUMÉ

Iberdomide is a potent cereblon E3 ligase modulator (CELMoD agent) with promising efficacy and safety as a monotherapy or in combination with other therapies in patients with relapsed/refractory multiple myeloma (RRMM). Using a custom mass cytometry panel designed for large-scale immunophenotyping of the bone marrow tumor microenvironment (TME), we demonstrate significant increases of effector T and natural killer (NK) cells in a cohort of 93 patients with multiple myeloma (MM) treated with iberdomide, correlating findings to disease characteristics, prior therapy, and a peripheral blood immune phenotype. Notably, changes are dose dependent, associated with objective response, and independent of prior refractoriness to MM therapies. This suggests that iberdomide broadly induces innate and adaptive immune activation in the TME, contributing to its antitumor efficacy. Our approach establishes a strategy to study treatment-induced changes in the TME of patients with MM and, more broadly, patients with cancer and establishes rational combination strategies for iberdomide with immune-enhancing therapies to treat MM.


Sujet(s)
Moelle osseuse , Immunité innée , Myélome multiple , Humains , Myélome multiple/traitement médicamenteux , Myélome multiple/immunologie , Myélome multiple/anatomopathologie , Immunité innée/effets des médicaments et des substances chimiques , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/anatomopathologie , Moelle osseuse/immunologie , Immunité acquise/effets des médicaments et des substances chimiques , Femelle , Mâle , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Sujet âgé , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/immunologie , Récidive tumorale locale/traitement médicamenteux
5.
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
6.
Front Immunol ; 15: 1353513, 2024.
Article de Anglais | MEDLINE | ID: mdl-38680490

RÉSUMÉ

The recent identification of skull bone marrow as a reactive hematopoietic niche that can contribute to and direct leukocyte trafficking into the meninges and brain has transformed our view of this bone structure from a solid, protective casing to a living, dynamic tissue poised to modulate brain homeostasis and neuroinflammation. This emerging concept may be highly relevant to injuries that directly impact the skull such as in traumatic brain injury (TBI). From mild concussion to severe contusion with skull fracturing, the bone marrow response of this local myeloid cell reservoir has the potential to impact not just the acute inflammatory response in the brain, but also the remodeling of the calvarium itself, influencing its response to future head impacts. If we borrow understanding from recent discoveries in other CNS immunological niches and extend them to this nascent, but growing, subfield of neuroimmunology, it is not unreasonable to consider the hematopoietic compartment in the skull may similarly play an important role in health, aging, and neurodegenerative disease following TBI. This literature review briefly summarizes the traditional role of the skull in TBI and offers some additional insights into skull-brain interactions and their potential role in affecting secondary neuroinflammation and injury outcomes.


Sujet(s)
Lésions traumatiques de l'encéphale , Encéphale , Crâne , Humains , Lésions traumatiques de l'encéphale/anatomopathologie , Animaux , Encéphale/immunologie , Encéphale/anatomopathologie , Encéphale/métabolisme , Crâne/traumatismes , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/anatomopathologie , Maladies neuro-inflammatoires/étiologie , Moelle osseuse/métabolisme , Moelle osseuse/anatomopathologie , Moelle osseuse/immunologie
7.
Nat Immunol ; 25(5): 820-833, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38600356

RÉSUMÉ

Human bone marrow permanently harbors high numbers of neutrophils, and a tumor-supportive bias of these cells could significantly impact bone marrow-confined malignancies. In individuals with multiple myeloma, the bone marrow is characterized by inflammatory stromal cells with the potential to influence neutrophils. We investigated myeloma-associated alterations in human marrow neutrophils and the impact of stromal inflammation on neutrophil function. Mature neutrophils in myeloma marrow are activated and tumor supportive and transcribe increased levels of IL1B and myeloma cell survival factor TNFSF13B (BAFF). Interactions with inflammatory stromal cells induce neutrophil activation, including BAFF secretion, in a STAT3-dependent manner, and once activated, neutrophils gain the ability to reciprocally induce stromal activation. After first-line myeloid-depleting antimyeloma treatment, human bone marrow retains residual stromal inflammation, and newly formed neutrophils are reactivated. Combined, we identify a neutrophil-stromal cell feed-forward loop driving tumor-supportive inflammation that persists after treatment and warrants novel strategies to target both stromal and immune microenvironments in multiple myeloma.


Sujet(s)
Facteur d'activation des lymphocytes B , Interleukine-1 bêta , Myélome multiple , Granulocytes neutrophiles , Cellules stromales , Microenvironnement tumoral , Myélome multiple/immunologie , Myélome multiple/anatomopathologie , Humains , Microenvironnement tumoral/immunologie , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Cellules stromales/métabolisme , Cellules stromales/immunologie , Facteur d'activation des lymphocytes B/métabolisme , Interleukine-1 bêta/métabolisme , Activation des neutrophiles , Facteur de transcription STAT-3/métabolisme , Moelle osseuse/immunologie , Moelle osseuse/anatomopathologie
8.
Mol Genet Genomics ; 299(1): 47, 2024 Apr 22.
Article de Anglais | MEDLINE | ID: mdl-38649532

RÉSUMÉ

Multiple myeloma (MM) is a plasma cell dyscrasia that is characterized by the uncontrolled proliferation of malignant PCs in the bone marrow. Due to immunotherapy, attention has returned to the immune system in MM, and it appears necessary to identify biomarkers in this area. In this study, we created a prognostic model for MM using immune-related gene pairs (IRGPs), with the advantage that it is not affected by technical bias. After retrieving microarray data of MM patients, bioinformatics analyses like COX regression and least absolute shrinkage and selection operator (LASSO) were used to construct the signature. Then its prognostic value is assessed via time-dependent receiver operating characteristic (ROC) and the Kaplan-Meier (KM) analysis. We also used XCELL to examine the status of immune cell infiltration among MM patients. 6-IRGP signatures were developed and proved to predict MM prognosis with a P-value of 0.001 in the KM analysis. Moreover, the risk score was significantly associated with clinicopathological characteristics and was an independent prognostic factor. Of note, the combination of age and ß2-microglobulin with risk score could improve the accuracy of determining patients' prognosis with the values of the area under the curve (AUC) of 0.73 in 5 years ROC curves. Our model was also associated with the distribution of immune cells. This novel signature, either alone or in combination with age and ß2-microglobulin, showed a good prognostic predictive value and might be used to guide the management of MM patients in clinical practice.


Sujet(s)
Moelle osseuse , Analyse de profil d'expression de gènes , Myélome multiple , Myélome multiple/génétique , Myélome multiple/immunologie , Myélome multiple/mortalité , Humains , Femelle , Pronostic , Mâle , Analyse de profil d'expression de gènes/méthodes , Moelle osseuse/anatomopathologie , Moelle osseuse/immunologie , Adulte d'âge moyen , Sujet âgé , Régulation de l'expression des gènes tumoraux , bêta-2-Microglobuline/génétique , Marqueurs biologiques tumoraux/génétique , Estimation de Kaplan-Meier , Courbe ROC , Transcriptome/génétique
9.
Int Immunol ; 36(7): 339-352, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38430523

RÉSUMÉ

Bone marrow is a dynamic organ composed of stem cells that constantly receive signals from stromal cells and other hematopoietic cells in the niches of the bone marrow to maintain hematopoiesis and generate immune cells. Perturbation of the bone marrow microenvironment by infection and inflammation affects hematopoiesis and may affect immune cell development. Little is known about the effect of malaria on the bone marrow stromal cells that govern the hematopoietic stem cell (HSC) niche. In this study, we demonstrate that the mesenchymal stromal CXCL12-abundant reticular (CAR) cell population is reduced during acute malaria infection. The reduction of CXCL12 and interleukin-7 signals in the bone marrow impairs the lymphopoietic niche, leading to the depletion of common lymphoid progenitors, B cell progenitors, and mature B cells, including plasma cells in the bone marrow. We found that interferon-γ (IFNγ) is responsible for the upregulation of Sca1 on CAR cells, yet the decline in CAR cell and B cell populations in the bone marrow is IFNγ-independent. In contrast to the decline in B cell populations, HSCs and multipotent progenitors increased with the expansion of myelopoiesis and erythropoiesis, indicating a bias in the differentiation of multipotent progenitors during malaria infection. These findings suggest that malaria may affect host immunity by modulating the bone marrow niche.


Sujet(s)
Lymphocytes B , Moelle osseuse , Chimiokine CXCL12 , Paludisme , Souris de lignée C57BL , Animaux , Chimiokine CXCL12/métabolisme , Chimiokine CXCL12/immunologie , Souris , Paludisme/immunologie , Paludisme/parasitologie , Lymphocytes B/immunologie , Moelle osseuse/immunologie , Moelle osseuse/parasitologie , Niche de cellules souches/immunologie , Interféron gamma/métabolisme , Interféron gamma/immunologie , Cellules souches hématopoïétiques/immunologie , Cellules souches hématopoïétiques/métabolisme
10.
Nature ; 628(8008): 612-619, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38509366

RÉSUMÉ

There is increasing interest in how immune cells in the meninges-the membranes that surround the brain and spinal cord-contribute to homeostasis and disease in the central nervous system1,2. The outer layer of the meninges, the dura mater, has recently been described to contain both innate and adaptive immune cells, and functions as a site for B cell development3-6. Here we identify organized lymphoid structures that protect fenestrated vasculature in the dura mater. The most elaborate of these dural-associated lymphoid tissues (DALT) surrounded the rostral-rhinal confluence of the sinuses and included lymphatic vessels. We termed this structure, which interfaces with the skull bone marrow and a comparable venous plexus at the skull base, the rostral-rhinal venolymphatic hub. Immune aggregates were present in DALT during homeostasis and expanded with age or after challenge with systemic or nasal antigens. DALT contain germinal centre B cells and support the generation of somatically mutated, antibody-producing cells in response to a nasal pathogen challenge. Inhibition of lymphocyte entry into the rostral-rhinal hub at the time of nasal viral challenge abrogated the generation of germinal centre B cells and class-switched plasma cells, as did perturbation of B-T cell interactions. These data demonstrate a lymphoid structure around vasculature in the dura mater that can sample antigens and rapidly support humoral immune responses after local pathogen challenge.


Sujet(s)
Dure-mère , Immunité humorale , Tissu lymphoïde , Veines , Administration par voie nasale , Antigènes/administration et posologie , Antigènes/immunologie , Moelle osseuse/immunologie , Système nerveux central/vascularisation , Système nerveux central/immunologie , Dure-mère/vascularisation , Dure-mère/immunologie , Centre germinatif/cytologie , Centre germinatif/immunologie , Vaisseaux lymphatiques/immunologie , Tissu lymphoïde/vascularisation , Tissu lymphoïde/immunologie , Plasmocytes/immunologie , Crâne/vascularisation , Lymphocytes T/immunologie , Veines/physiologie , Humains , Mâle , Femelle , Adulte , Adulte d'âge moyen , Animaux , Souris , Sujet âgé de 80 ans ou plus
11.
Nat Rev Immunol ; 24(8): 543-558, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38491073

RÉSUMÉ

Tissue inflammation is a hallmark of tumour microenvironments. In the bone marrow, tumour-associated inflammation impacts normal niches for haematopoietic progenitor cells and mature immune cells and supports the outgrowth and survival of malignant cells residing in these niche compartments. This Review provides an overview of our current understanding of inflammatory changes in the bone marrow microenvironment of myeloid and lymphoid malignancies, using acute myeloid leukaemia and multiple myeloma as examples and highlights unique and shared features of inflammation in niches for progenitor cells and plasma cells. Importantly, inflammation exerts profoundly different effects on normal bone marrow niches in these malignancies, and we provide context for possible drivers of these divergent effects. We explore the role of tumour cells in inflammatory changes, as well as the role of cellular constituents of normal bone marrow niches, including myeloid cells and stromal cells. Integrating knowledge of disease-specific dynamics of malignancy-associated bone marrow inflammation will provide a necessary framework for future targeting of these processes to improve patient outcome.


Sujet(s)
Moelle osseuse , Tumeurs hématologiques , Inflammation , Microenvironnement tumoral , Humains , Inflammation/immunologie , Inflammation/anatomopathologie , Moelle osseuse/anatomopathologie , Moelle osseuse/immunologie , Tumeurs hématologiques/immunologie , Tumeurs hématologiques/anatomopathologie , Microenvironnement tumoral/immunologie , Animaux , Cellules souches hématopoïétiques/immunologie , Cellules souches hématopoïétiques/anatomopathologie , Myélome multiple/immunologie , Myélome multiple/anatomopathologie , Niche de cellules souches/immunologie
12.
Nat Immunol ; 24(8): 1265-1280, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37414907

RÉSUMÉ

High-dimensional approaches have revealed heterogeneity amongst dendritic cells (DCs), including a population of transitional DCs (tDCs) in mice and humans. However, the origin and relationship of tDCs to other DC subsets has been unclear. Here we show that tDCs are distinct from other well-characterized DCs and conventional DC precursors (pre-cDCs). We demonstrate that tDCs originate from bone marrow progenitors shared with plasmacytoid DCs (pDCs). In the periphery, tDCs contribute to the pool of ESAM+ type 2 DCs (DC2s), and these DC2s have pDC-related developmental features. Different from pre-cDCs, tDCs have less turnover, capture antigen, respond to stimuli and activate antigen-specific naïve T cells, all characteristics of differentiated DCs. Different from pDCs, viral sensing by tDCs results in IL-1ß secretion and fatal immune pathology in a murine coronavirus model. Our findings suggest that tDCs are a distinct pDC-related subset with a DC2 differentiation potential and unique proinflammatory function during viral infections.


Sujet(s)
Moelle osseuse , Cellules dendritiques , Animaux , Souris , Antiviraux , Moelle osseuse/immunologie , Différenciation cellulaire , Cellules dendritiques/classification , Cellules dendritiques/immunologie
13.
Scand J Immunol ; 98(2): e13273, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-39007933

RÉSUMÉ

In recent years, chimeric antigen receptor-T (CAR-T) cell therapy has emerged as a novel immunotherapy method. It has shown significant therapeutic efficacy in the treatment of haematological B cell malignancies. In particular, the CAR-T therapy targeting CD19 has yielded unprecedented efficacy for acute B-lymphocytic leukaemia (B-ALL) and non-Hodgkin's lymphoma (NHL). In haematologic malignancies, tumour stem cells are more prone to stay in the regulatory bone marrow (BM) microenvironment (called niches), which provides a protective environment against immune attack. However, how the BM microenvironment affects the anti-tumour efficacy of CAR-T cells and its underlying mechanism is worthy of attention. In this review, we discuss the role of the BM microenvironment on the efficacy of CAR-T in haematological malignancies and propose corresponding strategies to enhance the anti-tumour activity of CAR-T therapy.


Sujet(s)
Moelle osseuse , Tumeurs hématologiques , Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Immunothérapie adoptive/méthodes , Tumeurs hématologiques/thérapie , Tumeurs hématologiques/immunologie , Moelle osseuse/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Animaux , Antigènes CD19/immunologie , Lymphocytes T/immunologie
14.
Blood Cancer Discov ; 3(6): 478-480, 2022 11 02.
Article de Anglais | MEDLINE | ID: mdl-36026510

RÉSUMÉ

In this issue of Blood Cancer Discovery, Dhodapkar and colleagues find that myeloid, dendritic, and endogenous T-cell populations in the bone marrow microenvironment are associated with progression-free survival (PFS) in multiple myeloma patients responding to B-cell maturation antigen-targeted CAR T cells. Immunosuppressive myeloid cells are associated with short PFS, but a diverse T-cell receptor repertoire and more dendritic cells are associated with a longer PFS, suggesting a potential role for epitope spreading. See related article by Dhodapkar et al., p. 490 (6).


Sujet(s)
Tumeurs de la moelle osseuse , Myélome multiple , Récepteurs chimériques pour l'antigène , Humains , Antigène de maturation des cellules B/immunologie , Myélome multiple/sang , Moelle osseuse/immunologie , Lymphocytes T/immunologie , Microenvironnement tumoral
15.
Nat Commun ; 13(1): 121, 2022 01 10.
Article de Anglais | MEDLINE | ID: mdl-35013215

RÉSUMÉ

HIV is difficult to eradicate due to the persistence of a long-lived reservoir of latently infected cells. Previous studies have shown that natural killer cells are important to inhibiting HIV infection, but it is unclear whether the administration of natural killer cells can reduce rebound viremia when anti-retroviral therapy is discontinued. Here we show the administration of allogeneic human peripheral blood natural killer cells delays viral rebound following interruption of anti-retroviral therapy in humanized mice infected with HIV-1. Utilizing genetically barcoded virus technology, we show these natural killer cells efficiently reduced viral clones rebounding from latency. Moreover, a kick and kill strategy comprised of the protein kinase C modulator and latency reversing agent SUW133 and allogeneic human peripheral blood natural killer cells during anti-retroviral therapy eliminated the viral reservoir in a subset of mice. Therefore, combinations utilizing latency reversal agents with targeted cellular killing agents may be an effective approach to eradicating the viral reservoir.


Sujet(s)
Agents antiVIH/pharmacologie , Lymphocytes T CD4+/immunologie , Infections à VIH/thérapie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/immunologie , Inhibiteurs de protéines kinases/pharmacologie , Virémie/thérapie , Animaux , Moelle osseuse/effets des médicaments et des substances chimiques , Moelle osseuse/immunologie , Moelle osseuse/virologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/virologie , Techniques de coculture , Femelle , Infections à VIH/génétique , Infections à VIH/immunologie , Infections à VIH/virologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Interactions hôte-pathogène/effets des médicaments et des substances chimiques , Interactions hôte-pathogène/génétique , Interactions hôte-pathogène/immunologie , Humains , Cellules tueuses naturelles/transplantation , Mâle , Souris , Souris transgéniques , Protéine kinase C/génétique , Protéine kinase C/immunologie , Rate/effets des médicaments et des substances chimiques , Rate/immunologie , Rate/virologie , Charge virale/effets des médicaments et des substances chimiques , Virémie/génétique , Virémie/immunologie , Virémie/virologie , Latence virale/effets des médicaments et des substances chimiques , Réplication virale/effets des médicaments et des substances chimiques
16.
Sci Rep ; 12(1): 215, 2022 01 07.
Article de Anglais | MEDLINE | ID: mdl-34997115

RÉSUMÉ

Disability and distress caused by chronic low back pain (LBP) lacking clear pathoanatomical explanations cause huge problems both for patients and society. A subgroup of patients has Modic changes (MC), identifiable by MRI as vertebral bone marrow lesions. The cause of such changes and their relationship to pain are not yet understood. We explored the pathobiology of these lesions using profiling of gene expression in blood, coupled with an edema-sensitive MRI technique known as short tau inversion recovery (STIR) imaging. STIR images and total RNA from blood were collected from 96 patients with chronic LBP and MC type I, the most inflammatory MC state. We found the expression of 37 genes significantly associated with STIR signal volume, ten genes with edema abundancy (a constructed combination of STIR signal volume, height, and intensity), and one gene with expression levels significantly associated with maximum STIR signal intensity. Gene sets related to interferon signaling, mitochondrial metabolism and defense response to virus were identified as significantly enriched among the upregulated genes in all three analyses. Our results point to inflammation and immunological defense as important players in MC biology in patients with chronic LBP.


Sujet(s)
Moelle osseuse/imagerie diagnostique , Douleur chronique/imagerie diagnostique , Analyse de profil d'expression de gènes , Lombalgie/imagerie diagnostique , Imagerie par résonance magnétique , Rachis/imagerie diagnostique , Transcriptome , Adulte , Moelle osseuse/immunologie , Douleur chronique/génétique , Douleur chronique/immunologie , Femelle , Régulation de l'expression des gènes , Humains , Lombalgie/génétique , Lombalgie/immunologie , Mâle , Adulte d'âge moyen , Valeur prédictive des tests , Essais contrôlés randomisés comme sujet , Rachis/immunologie
17.
J Extracell Vesicles ; 11(1): e12176, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34973063

RÉSUMÉ

Natural killer (NK) cells are innate cytotoxic lymphocytes that play a key role in cancer immunosurveillance thanks to their ability to recognize and kill cancer cells. NKG2D is an activating receptor that binds to MIC and ULBP molecules typically induced on damaged, transformed or infected cells. The release of NKG2D ligands (NKG2DLs) in the extracellular milieu through protease-mediated cleavage or by extracellular vesicle (EV) secretion allows cancer cells to evade NKG2D-mediated immunosurveillance. In this work, we investigated the immunomodulatory properties of the NKG2D ligand MICA*008 associated to distinct populations of EVs (i.e., small extracellular vesicles [sEVs] and medium size extracellular vesicles [mEVs]). By using as model a human MICA*008-transfected multiple myeloma (MM) cell line, we found that this ligand is present on both vesicle populations. Interestingly, our findings reveal that NKG2D is specifically involved in the uptake of vesicles expressing its cognate ligand. We provide evidence that MICA*008-expressing sEVs and mEVs are able on one hand to activate NK cells but, following prolonged stimulation induce a sustained NKG2D downmodulation leading to impaired NKG2D-mediated functions. Moreover, our findings show that MICA*008 can be transferred by vesicles to NK cells causing fratricide. Focusing on MM as a clinically and biologically relevant model of tumour-NK cell interactions, we found enrichment of EVs expressing MICA in the bone marrow of a cohort of patients. All together our results suggest that the accumulation of NKG2D ligands associated to vesicles in the tumour microenvironment could favour the suppression of NK cell activity either by NKG2D down-modulation or by fratricide of NK cell dressed with EV-derived NKG2D ligands.


Sujet(s)
Vésicules extracellulaires/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Surveillance immunologique , Cellules tueuses naturelles/immunologie , Myélome multiple/immunologie , Sujet âgé , Sujet âgé de 80 ans ou plus , Moelle osseuse/immunologie , Mort cellulaire/immunologie , Lignée cellulaire , Extracellular Signal-Regulated MAP Kinases/métabolisme , Femelle , Humains , Immunomodulation , Interféron gamma/métabolisme , Ligands , Mâle , Adulte d'âge moyen , Sous-famille K des récepteurs de cellules NK de type lectine/immunologie , Échappement de la tumeur à la surveillance immunitaire
18.
Cancer Sci ; 113(1): 41-52, 2022 Jan.
Article de Anglais | MEDLINE | ID: mdl-34716967

RÉSUMÉ

Due to the considerable success of cancer immunotherapy for leukemia, the tumor immune environment has become a focus of intense research; however, there are few reports on the dynamics of the tumor immune environment in leukemia. Here, we analyzed the tumor immune environment in pediatric B cell precursor acute lymphoblastic leukemia by analyzing serial bone marrow samples from nine patients with primary and recurrent disease by mass cytometry using 39 immunophenotype markers, and transcriptome analysis. High-dimensional single-cell mass cytometry analysis elucidated a dynamic shift of T cells from naïve to effector subsets, and clarified that, during relapse, the tumor immune environment comprised a T helper 1-polarized immune profile, together with an increased number of effector regulatory T cells. These results were confirmed in a validation cohort using conventional flow cytometry. Furthermore, RNA transcriptome analysis identified the upregulation of immune-related pathways in B cell precursor acute lymphoblastic leukemia cells during relapse, suggesting interaction with the surrounding environment. In conclusion, a tumor immune environment characterized by a T helper 1-polarized immune profile, with an increased number of effector regulatory T cells, could contribute to the pathophysiology of recurrent B cell precursor acute lymphoblastic leukemia. This information could contribute to the development of effective immunotherapeutic approaches against B cell precursor acute lymphoblastic leukemia relapse.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Moelle osseuse/immunologie , Analyse de profil d'expression de gènes/méthodes , Récidive tumorale locale/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Adolescent , Moelle osseuse/composition chimique , Enfant , Enfant d'âge préscolaire , Femelle , Cytométrie en flux , Régulation de l'expression des gènes tumoraux , Humains , Nourrisson , Mâle , Récidive tumorale locale/immunologie , Leucémie-lymphome lymphoblastique à précurseurs B/immunologie , Analyse de séquence d'ARN , Analyse sur cellule unique , Microenvironnement tumoral , Régulation positive , Jeune adulte
19.
Leukemia ; 36(3): 790-800, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34584204

RÉSUMÉ

Multiple myeloma remains a largely incurable disease of clonally expanding malignant plasma cells. The bone marrow microenvironment harbors treatment-resistant myeloma cells, which eventually lead to disease relapse in patients. In the bone marrow, CD4+FoxP3+ regulatory T cells (Tregs) are highly abundant amongst CD4+ T cells providing an immune protective niche for different long-living cell populations, e.g., hematopoietic stem cells. Here, we addressed the functional role of Tregs in multiple myeloma dissemination to bone marrow compartments and disease progression. To investigate the immune regulation of multiple myeloma, we utilized syngeneic immunocompetent murine multiple myeloma models in two different genetic backgrounds. Analyzing the spatial immune architecture of multiple myeloma revealed that the bone marrow Tregs accumulated in the vicinity of malignant plasma cells and displayed an activated phenotype. In vivo Treg depletion prevented multiple myeloma dissemination in both models. Importantly, short-term in vivo depletion of Tregs in mice with established multiple myeloma evoked a potent CD8 T cell- and NK cell-mediated immune response resulting in complete and stable remission. Conclusively, this preclinical in-vivo study suggests that Tregs are an attractive target for the treatment of multiple myeloma.


Sujet(s)
Myélome multiple/immunologie , Lymphocytes T régulateurs/immunologie , Animaux , Moelle osseuse/immunologie , Évolution de la maladie , Humains , Activation des lymphocytes , Souris , Microenvironnement tumoral
20.
Immunology ; 165(1): 88-98, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34435359

RÉSUMÉ

B-cell development undergoes a series of steps from the bone marrow to the secondary lymphoid organs. A defect in B-cell development can lead to immunodeficiency or malignant disorders, such as leukaemia or lymphoma. Long non-coding RNAs have been reported to act as important regulators of many pathological processes. However, very little is known regarding the role of lncRNAs during B-cell development and the regulation of their expression. In this study, we explored the expression and role of lncRNA Gme00492 in B-cell development. We observed that lnc00492 was highly expressed in B-cell development and primarily expressed in the nucleus. Lnc00492-deficient mice had fewer marginal zone B cells in the spleen, likely due to a developmental block. Importantly, lnc00492 interacts with CTBP1 and targets it for ubiquitination and degradation during B-cell development, whereas the transcriptional corepressor factor CTBP1 plays a critical role in Notch2 signalling. Thus, we identified a novel regulatory axis between lnc00492 and CTBP1 in B cells, suggesting that lnc00492 is essential for marginal zone B-cell development.


Sujet(s)
Lymphocytes B/cytologie , Lymphocytes B/métabolisme , Différenciation cellulaire/génétique , Lymphopoïèse/génétique , ARN long non codant/génétique , Alcohol oxidoreductases/métabolisme , Animaux , Lymphocytes B/immunologie , Marqueurs biologiques , Moelle osseuse/immunologie , Moelle osseuse/métabolisme , Moelle osseuse/anatomopathologie , Différenciation cellulaire/immunologie , Protéines de liaison à l'ADN/métabolisme , Régulation de l'expression des gènes , Immunophénotypage , Souris , Souris knockout , Modèles biologiques , Liaison aux protéines , Récepteur Notch2/métabolisme , Transduction du signal , Rate/cytologie , Rate/immunologie , Rate/métabolisme , Ubiquitination
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE