Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Nat Chem Biol ; 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075252

RÉSUMÉ

Molecular glues are proximity-inducing small molecules that have emerged as an attractive therapeutic approach. However, developing molecular glues remains challenging, requiring innovative mechanistic strategies to stabilize neoprotein interfaces and expedite discovery. Here we unveil a trans-labeling covalent molecular glue mechanism, termed 'template-assisted covalent modification'. We identified a new series of BRD4 molecular glue degraders that recruit CUL4DCAF16 ligase to the second bromodomain of BRD4 (BRD4BD2). Through comprehensive biochemical, structural and mutagenesis analyses, we elucidated how pre-existing structural complementarity between DCAF16 and BRD4BD2 serves as a template to optimally orient the degrader for covalent modification of DCAF16Cys58. This process stabilizes the formation of BRD4-degrader-DCAF16 ternary complex and facilitates BRD4 degradation. Supporting generalizability, we found that a subset of degraders also induces GAK-BRD4BD2 interaction through trans-labeling of GAK. Together, our work establishes 'template-assisted covalent modification' as a mechanism for covalent molecular glues, which opens a new path to proximity-driven pharmacology.

2.
Nat Rev Mol Cell Biol ; 25(9): 740-757, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38684868

RÉSUMÉ

Targeted protein degradation refers to the use of small molecules to induce the selective degradation of proteins. In its most common form, this degradation is achieved through ligand-mediated neo-interactions between ubiquitin E3 ligases - the principal waste disposal machines of a cell - and the protein targets of interest, resulting in ubiquitylation and subsequent proteasomal degradation. Notable advances have been made in biological and mechanistic understanding of serendipitously discovered degraders. This improved understanding and novel chemistry has not only provided clinical proof of concept for targeted protein degradation but has also led to rapid growth of the field, with dozens of investigational drugs in active clinical trials. Two distinct classes of protein degradation therapeutics are being widely explored: bifunctional PROTACs and molecular glue degraders, both of which have their unique advantages and challenges. Here, we review the current landscape of targeted protein degradation approaches and how they have parallels in biological processes. We also outline the ongoing clinical exploration of novel degraders and provide some perspectives on the directions the field might take.


Sujet(s)
Proteasome endopeptidase complex , Protéolyse , Ubiquitin-protein ligases , Ubiquitination , Humains , Ubiquitin-protein ligases/métabolisme , Proteasome endopeptidase complex/métabolisme , Animaux , Protéines/métabolisme
3.
J Mol Diagn ; 25(9): 665-681, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37419244

RÉSUMÉ

Recognition of aberrant gene isoforms due to DNA events can impact risk stratification and molecular classification of hematolymphoid tumors. In myelodysplastic syndromes, KMT2A partial tandem duplication (PTD) was one of the top adverse predictors in the International Prognostic Scoring System-Molecular study. In B-cell acute lymphoblastic leukemia (B-ALL), ERG isoforms have been proposed as markers of favorable-risk DUX4 rearrangements, whereas deletion-mediated IKZF1 isoforms are associated with adverse prognosis and have been extended to the high-risk IKZF1plus signature defined by codeletions, including PAX5. In this limited study, outlier expression of isoforms as markers of IKZF1 intragenic or 3' deletions, DUX4 rearrangements, or PAX5 intragenic deletions were 92.3% (48/52), 90% (9/10), or 100% (9/9) sensitive, respectively, and 98.7% (368/373), 100% (35/35), or 97.1% (102/105) specific, respectively, by targeted RNA sequencing, and 84.0% (21/25), 85.7% (6/7), or 81.8% (9/11) sensitive, respectively, and 98.2% (109/111), 98.4% (127/129), or 98.7% (78/79) specific, respectively, by total RNA sequencing. Comprehensive split-read analysis identified expressed DNA breakpoints, cryptic splice sites associated with IKZF1 3' deletions, PTD of IKZF1 exon 5 spanning N159Y in B-ALL with mutated IKZF1 N159Y, and truncated KMT2A-PTD isoforms. Outlier isoforms were also effective targeted RNA markers for PAX5 intragenic amplifications (B-ALL), KMT2A-PTD (myeloid malignant cancers), and rare NOTCH1 intragenic deletions (T-cell acute lymphoblastic leukemia). These findings support the use of outlier isoform analysis as a robust strategy for detecting clinically significant DNA events.


Sujet(s)
Tumeurs , Leucémie-lymphome lymphoblastique à précurseurs B , Humains , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Isoformes de protéines/génétique , Analyse de séquence d'ARN , Génomique
4.
Mol Cell ; 83(15): 2753-2767.e10, 2023 08 03.
Article de Anglais | MEDLINE | ID: mdl-37478846

RÉSUMÉ

Nuclear hormone receptors (NRs) are ligand-binding transcription factors that are widely targeted therapeutically. Agonist binding triggers NR activation and subsequent degradation by unknown ligand-dependent ubiquitin ligase machinery. NR degradation is critical for therapeutic efficacy in malignancies that are driven by retinoic acid and estrogen receptors. Here, we demonstrate the ubiquitin ligase UBR5 drives degradation of multiple agonist-bound NRs, including the retinoic acid receptor alpha (RARA), retinoid x receptor alpha (RXRA), glucocorticoid, estrogen, liver-X, progesterone, and vitamin D receptors. We present the high-resolution cryo-EMstructure of full-length human UBR5 and a negative stain model representing its interaction with RARA/RXRA. Agonist ligands induce sequential, mutually exclusive recruitment of nuclear coactivators (NCOAs) and UBR5 to chromatin to regulate transcriptional networks. Other pharmacological ligands such as selective estrogen receptor degraders (SERDs) degrade their receptors through differential recruitment of UBR5 or RNF111. We establish the UBR5 transcriptional regulatory hub as a common mediator and regulator of NR-induced transcription.


Sujet(s)
Chromatine , Facteurs de transcription , Humains , Ligands , Chromatine/génétique , Facteurs de transcription/métabolisme , Récepteurs cytoplasmiques et nucléaires/génétique , Ubiquitines , Ubiquitin-protein ligases/génétique
5.
iScience ; 26(5): 106601, 2023 May 19.
Article de Anglais | MEDLINE | ID: mdl-37095859

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) hijacks multiple human proteins during infection and viral replication. To examine whether any viral proteins employ human E3 ubiquitin ligases, we evaluated the stability of SARS-CoV-2 proteins with inhibition of the ubiquitin proteasome pathway. Using genetic screens to dissect the molecular machinery involved in the degradation of candidate viral proteins, we identified human E3 ligase RNF185 as a regulator of protein stability for the SARS-CoV-2 envelope protein. We found that RNF185 and the SARS-CoV-2 envelope co-localize to the endoplasmic reticulum (ER). Finally, we demonstrate that the depletion of RNF185 significantly increases SARS-CoV-2 viral titer in a cellular model. Modulation of this interaction could provide opportunities for novel antiviral therapies.

6.
bioRxiv ; 2023 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-36824856

RÉSUMÉ

Small molecules that induce protein-protein interactions to exert proximity-driven pharmacology such as targeted protein degradation are a powerful class of therapeutics1-3. Molecular glues are of particular interest given their favorable size and chemical properties and represent the only clinically approved degrader drugs4-6. The discovery and development of molecular glues for novel targets, however, remains challenging. Covalent strategies could in principle facilitate molecular glue discovery by stabilizing the neo-protein interfaces. Here, we present structural and mechanistic studies that define a trans-labeling covalent molecular glue mechanism, which we term "template-assisted covalent modification". We found that a novel series of BRD4 molecular glue degraders act by recruiting the CUL4DCAF16 ligase to the second bromodomain of BRD4 (BRD4BD2). BRD4BD2, in complex with DCAF16, serves as a structural template to facilitate covalent modification of DCAF16, which stabilizes the BRD4-degrader-DCAF16 ternary complex formation and facilitates BRD4 degradation. A 2.2 Å cryo-electron microscopy structure of the ternary complex demonstrates that DCAF16 and BRD4BD2 have pre-existing structural complementarity which optimally orients the reactive moiety of the degrader for DCAF16Cys58 covalent modification. Systematic mutagenesis of both DCAF16 and BRD4BD2 revealed that the loop conformation around BRD4His437, rather than specific side chains, is critical for stable interaction with DCAF16 and BD2 selectivity. Together our work establishes "template-assisted covalent modification" as a mechanism for covalent molecular glues, which opens a new path to proximity driven pharmacology.

7.
J Exp Med ; 218(12)2021 12 06.
Article de Anglais | MEDLINE | ID: mdl-34698806

RÉSUMÉ

Osteoporosis is caused by an imbalance of osteoclasts and osteoblasts, occurring in close proximity to hematopoietic cells in the bone marrow. Recurrent somatic mutations that lead to an expanded population of mutant blood cells is termed clonal hematopoiesis of indeterminate potential (CHIP). Analyzing exome sequencing data from the UK Biobank, we found CHIP to be associated with increased incident osteoporosis diagnoses and decreased bone mineral density. In murine models, hematopoietic-specific mutations in Dnmt3a, the most commonly mutated gene in CHIP, decreased bone mass via increased osteoclastogenesis. Dnmt3a-/- demethylation opened chromatin and altered activity of inflammatory transcription factors. Bone loss was driven by proinflammatory cytokines, including Irf3-NF-κB-mediated IL-20 expression from Dnmt3a mutant macrophages. Increased osteoclastogenesis due to the Dnmt3a mutations was ameliorated by alendronate or IL-20 neutralization. These results demonstrate a novel source of osteoporosis-inducing inflammation.


Sujet(s)
Hématopoïèse clonale/génétique , DNA methyltransferase 3A/génétique , Ostéoporose/génétique , Adulte , Sujet âgé , Alendronate/pharmacologie , Animaux , Anticorps neutralisants/pharmacologie , Différenciation cellulaire/génétique , Hématopoïèse clonale/physiologie , DNA methyltransferase 3A/métabolisme , Femelle , Humains , Interleukines/immunologie , Interleukines/métabolisme , Mâle , Souris knockout , Adulte d'âge moyen , Ostéoclastes/anatomopathologie , Ostéoporose/sang , Ostéoporose/traitement médicamenteux , Ostéoporose/physiopathologie
8.
Oncologist ; 26(12): e2297-e2301, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34347347

RÉSUMÉ

Comprehensive genetic profiling using next-generation sequencing technologies has become an integral part of precision oncology. Variant annotation requires translating the DNA findings into protein level predictions. In this article we highlight inconsistencies in variant annotation for the MET D1228N exon 19 resistance mutations. MET D1228N and D1246N represent the same resistance mutation in MET exon 14 skipping alterations annotated on different transcripts. Additional examples of relevant variants annotated on different transcripts emphasize the importance of avoiding erroneous interpretation when realizing precision oncology.


Sujet(s)
Exons , Tumeurs , Protéines proto-oncogènes c-met/génétique , Humains , Mutation , Tumeurs/génétique , Médecine de précision
9.
J Appl Lab Med ; 6(5): 1338-1354, 2021 09 01.
Article de Anglais | MEDLINE | ID: mdl-33822967

RÉSUMÉ

The COVID-19 pandemic has made a devastating impact on global health and continues to challenge healthcare infrastructure and delivery. The clinical laboratories were no exception as they are responsible for diagnostic testing that dictates many clinical, infection control, and public health decisions. Information technology and laboratory management tools are critical assets for maintaining and adapting operations in response to crises. When utilized effectively, they promote the integration between the clinical laboratory specialties (e.g., chemistry, hematology, microbiology, and molecular pathology). During the COVID-19 pandemic, our systems and processes were strained due to high testing volumes, demand for rapid turnaround times, supply chain constraints, and constantly evolving testing algorithms and result interpretations as our knowledge of the virus and of diagnostics increased over time. In this report, we describe those challenges and subsequent adaptations made by each clinical laboratory section. We hope these details help to provide potential solutions and approaches for other hospitals facing COVID-19 surges or other future pandemics.


Sujet(s)
COVID-19 , Services de laboratoire d'analyses médicales , Humains , Laboratoires , Pandémies/prévention et contrôle , SARS-CoV-2
10.
Nature ; 588(7836): 164-168, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33208943

RÉSUMÉ

Effective and sustained inhibition of non-enzymatic oncogenic driver proteins is a major pharmacological challenge. The clinical success of thalidomide analogues demonstrates the therapeutic efficacy of drug-induced degradation of transcription factors and other cancer targets1-3, but a substantial subset of proteins are resistant to targeted degradation using existing approaches4,5. Here we report an alternative mechanism of targeted protein degradation, in which a small molecule induces the highly specific, reversible polymerization of a target protein, followed by its sequestration into cellular foci and subsequent degradation. BI-3802 is a small molecule that binds to the Broad-complex, Tramtrack and Bric-à-brac (BTB) domain of the oncogenic transcription factor B cell lymphoma 6 (BCL6) and leads to the proteasomal degradation of BCL66. We use cryo-electron microscopy to reveal how the solvent-exposed moiety of a BCL6-binding molecule contributes to a composite ligand-protein surface that engages BCL6 homodimers to form a supramolecular structure. Drug-induced formation of BCL6 filaments facilitates ubiquitination by the SIAH1 E3 ubiquitin ligase. Our findings demonstrate that a small molecule such as BI-3802 can induce polymerization coupled to highly specific protein degradation, which in the case of BCL6 leads to increased pharmacological activity compared to the effects induced by other BCL6 inhibitors. These findings open new avenues for the development of therapeutic agents and synthetic biology.


Sujet(s)
Polymérisation/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-bcl-6/composition chimique , Protéines proto-oncogènes c-bcl-6/métabolisme , Cryomicroscopie électronique , Humains , Techniques in vitro , Ligands , Modèles moléculaires , Protéines nucléaires/métabolisme , Proteasome endopeptidase complex/effets des médicaments et des substances chimiques , Proteasome endopeptidase complex/métabolisme , Protéines proto-oncogènes c-bcl-6/ultrastructure , Solvants , Biologie synthétique , Ubiquitin-protein ligases/métabolisme , Ubiquitination/effets des médicaments et des substances chimiques
11.
Blood Adv ; 3(18): 2713-2721, 2019 09 24.
Article de Anglais | MEDLINE | ID: mdl-31519647

RÉSUMÉ

Peritoneal adhesions are pathological fibroses that ensnare organs after abdominal surgery. This dense connective tissue can cause small bowel obstruction, female infertility, and chronic abdominal pain. The pathogenesis of adhesions is a fibrotic response to tissue damage coordinated between mesothelial cells, fibroblasts, and immune cells. We have previously demonstrated that peritoneal adhesions are a consequence of mechanical injury to the mesothelial layer sustained during surgery. Neutrophils are among the first leukocytes involved in the early response to tissue damage. Here, we show that when subjected to mechanical stress, activated mesothelial cells directly recruit neutrophils and monocytes through upregulation of chemokines such as CXCL1 and monocyte chemoattractant protein 1 (MCP-1). We find that neutrophils within the adhesion sites undergo cell death and form neutrophil extracellular traps (NETosis) that contribute to pathogenesis. Conversely, tissue-resident macrophages were profoundly depleted throughout the disease time course. We show that this is distinct from traditional inflammatory kinetics such as after sham surgery or chemically induced peritonitis, and suggest that adhesions result from a primary difference in inflammatory kinetics. We find that transient depletion of circulating neutrophils significantly decreases adhesion burden, and further recruitment of monocytes with thioglycolate or MCP-1 also improves outcomes. Our findings suggest that the combination of neutrophil depletion and monocyte recruitment is sufficient to prevent adhesion formation, thus providing insight for potential clinical interventions.


Sujet(s)
Monocytes/métabolisme , Granulocytes neutrophiles/métabolisme , Adhérences tissulaires/métabolisme , Animaux , Femelle , Humains , Souris
12.
Nature ; 564(7736): 425-429, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30518860

RÉSUMÉ

Haematopoiesis is an essential process that evolved in multicellular animals. At the heart of this process are haematopoietic stem cells (HSCs), which are multipotent and self-renewing, and generate the entire repertoire of blood and immune cells throughout an animal's life1. Although there have been comprehensive studies on self-renewal, differentiation, physiological regulation and niche occupation in vertebrate HSCs, relatively little is known about the evolutionary origin and niches of these cells. Here we describe the haematopoietic system of Botryllus schlosseri, a colonial tunicate that has a vasculature and circulating blood cells, and interesting stem-cell biology and immunity characteristics2-8. Self-recognition between genetically compatible B. schlosseri colonies leads to the formation of natural parabionts with shared circulation, whereas incompatible colonies reject each other3,4,7. Using flow cytometry, whole-transcriptome sequencing of defined cell populations and diverse functional assays, we identify HSCs, progenitors, immune effector cells and an HSC niche, and demonstrate that self-recognition inhibits allospecific cytotoxic reactions. Our results show that HSC and myeloid lineage immune cells emerged in a common ancestor of tunicates and vertebrates, and also suggest that haematopoietic bone marrow and the B. schlosseri endostyle niche evolved from a common origin.


Sujet(s)
Hématopoïèse , Système hématopoïétique/cytologie , Mammifères/sang , Phylogenèse , Urochordata/cytologie , Animaux , Différenciation cellulaire , Lignage cellulaire , Cytotoxicité immunologique , Femelle , Cytométrie en flux , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/immunologie , Immunité cellulaire , Isoantigènes/immunologie , Mâle , Mammifères/anatomie et histologie , Cellules myéloïdes/cytologie , Cellules myéloïdes/immunologie , Phagocytose/immunologie , Niche de cellules souches , Transcriptome/génétique , Urochordata/anatomie et histologie , Urochordata/génétique , Urochordata/immunologie
13.
Sci Transl Med ; 10(469)2018 11 28.
Article de Anglais | MEDLINE | ID: mdl-30487249

RÉSUMÉ

Peritoneal adhesions are fibrous tissues that tether organs to one another or to the peritoneal wall and are a major cause of postsurgical and infectious morbidity. The primary molecular chain of events leading to the initiation of adhesions has been elusive, chiefly due to the lack of an identifiable cell of origin. Using clonal analysis and lineage tracing, we have identified injured surface mesothelium expressing podoplanin (PDPN) and mesothelin (MSLN) as a primary instigator of peritoneal adhesions after surgery in mice. We demonstrate that an anti-MSLN antibody diminished adhesion formation in a mouse model where adhesions were induced by surgical ligation to form ischemic buttons and subsequent surgical abrasion of the peritoneum. RNA sequencing and bioinformatics analyses of mouse mesothelial cells from injured mesothelium revealed aspects of the pathological mechanism of adhesion development and yielded several potential regulators of this process. Specifically, we show that PDPN+MSLN+ mesothelium responded to hypoxia by early up-regulation of hypoxia-inducible factor 1 alpha (HIF1α) that preceded adhesion development. Inhibition of HIF1α with small molecules ameliorated the injury program in damaged mesothelium and was sufficient to diminish adhesion severity in a mouse model. Analyses of human adhesion tissue suggested that similar surface markers and signaling pathways may contribute to surgical adhesions in human patients.


Sujet(s)
Anticorps/pharmacologie , Marqueurs biologiques/métabolisme , Épithélium/anatomopathologie , Adhérences tissulaires/anatomopathologie , Animaux , Lignage cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Épithélium/effets des médicaments et des substances chimiques , Épithélium/métabolisme , Épithélium/ultrastructure , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/antagonistes et inhibiteurs , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Mésothéline , Souris de lignée BALB C , Souris de lignée C57BL , Péritoine/effets des médicaments et des substances chimiques , Péritoine/traumatismes , Péritoine/anatomopathologie , Adhérences tissulaires/génétique , Transcription génétique
14.
Proc Natl Acad Sci U S A ; 115(26): E5954-E5962, 2018 06 26.
Article de Anglais | MEDLINE | ID: mdl-29891676

RÉSUMÉ

In the adult mouse spinal cord, the ependymal cell population that surrounds the central canal is thought to be a promising source of quiescent stem cells to treat spinal cord injury. Relatively little is known about the cellular origin of ependymal cells during spinal cord development, or the molecular mechanisms that regulate ependymal cells during adult homeostasis. Using genetic lineage tracing based on the Wnt target gene Axin2, we have characterized Wnt-responsive cells during spinal cord development. Our results revealed that Wnt-responsive progenitor cells are restricted to the dorsal midline throughout spinal cord development, which gives rise to dorsal ependymal cells in a spatially restricted pattern. This is contrary to previous reports that suggested an exclusively ventral origin of ependymal cells, suggesting that ependymal cells may retain positional identities in relation to their neural progenitors. Our results further demonstrated that in the postnatal and adult spinal cord, all ependymal cells express the Wnt/ß-catenin signaling target gene Axin2, as well as Wnt ligands. Genetic elimination of ß-catenin or inhibition of Wnt secretion in Axin2-expressing ependymal cells in vivo both resulted in impaired proliferation, indicating that Wnt/ß-catenin signaling promotes ependymal cell proliferation. These results demonstrate the continued importance of Wnt/ß-catenin signaling for both ependymal cell formation and regulation. By uncovering the molecular signals underlying the formation and regulation of spinal cord ependymal cells, our findings thus enable further targeting and manipulation of this promising source of quiescent stem cells for therapeutic interventions.


Sujet(s)
Axine/métabolisme , Prolifération cellulaire , Névroglie/métabolisme , Moelle spinale/croissance et développement , Voie de signalisation Wnt/physiologie , Animaux , Axine/génétique , Souris , Souris transgéniques , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Névroglie/cytologie , Moelle spinale/cytologie , Protéines de type Wingless/génétique , Protéines de type Wingless/métabolisme , bêta-Caténine/génétique , bêta-Caténine/métabolisme
15.
J Vis Exp ; (135)2018 05 31.
Article de Anglais | MEDLINE | ID: mdl-29912198

RÉSUMÉ

Morphological organ regeneration following acute tissue loss is common among lower vertebrates, but is rarely observed in mammalian postnatal life. Adult liver regeneration after 70% partial hepatectomy results in hepatocyte hypertrophy with some replication in remaining lobes with restoration of metabolic activity, but with permanent loss of the injured lobe's morphology and architecture. Here, we detail a new surgical method in the neonate that leaves a physiologic environment conducive to regeneration. This model involves amputation of the left lobe apex and a subsequent conservative management regimen, and lacks the necessity for ligation of major liver vessels or chemical injury, leaving a physiologic environment where regeneration may occur. We extend this protocol to amputations on juvenile (P7-14) mice, during which the injured liver transitions from organ regeneration to compensatory growth by hypertrophy. The presented, brief 30 min protocol provides a framework to study the mechanisms of regeneration, its age-associated decline in mammals, and the characterization of putative hepatic stem or progenitors.


Sujet(s)
Hépatectomie/méthodes , Régénération hépatique/physiologie , Foie/chirurgie , Animaux , Foie/anatomopathologie , Mâle , Souris
16.
Nat Immunol ; 19(1): 76-84, 2018 Jan.
Article de Anglais | MEDLINE | ID: mdl-29180808

RÉSUMÉ

Exciting progress in the field of cancer immunotherapy has renewed the urgency of the need for basic studies of immunoregulation in both adaptive cell lineages and innate cell lineages. Here we found a central role for major histocompatibility complex (MHC) class I in controlling the phagocytic function of macrophages. Our results demonstrated that expression of the common MHC class I component ß2-microglobulin (ß2M) by cancer cells directly protected them from phagocytosis. We further showed that this protection was mediated by the inhibitory receptor LILRB1, whose expression was upregulated on the surface of macrophages, including tumor-associated macrophages. Disruption of either MHC class I or LILRB1 potentiated phagocytosis of tumor cells both in vitro and in vivo, which defines the MHC class I-LILRB1 signaling axis as an important regulator of the effector function of innate immune cells, a potential biomarker for therapeutic response to agents directed against the signal-regulatory protein CD47 and a potential target of anti-cancer immunotherapy.


Sujet(s)
Antigènes d'histocompatibilité de classe I/immunologie , Récepteur B1 de type immunoglobuline des leucocytes/immunologie , Macrophages/immunologie , Tumeurs/immunologie , Phagocytose/immunologie , Animaux , Lignée cellulaire tumorale , Antigènes d'histocompatibilité de classe I/métabolisme , Humains , Immunothérapie/méthodes , Récepteur B1 de type immunoglobuline des leucocytes/métabolisme , Macrophages/métabolisme , Souris de lignée BALB C , Souris de lignée C57BL , Souris de lignée NOD , Souris knockout , Souris SCID , Tumeurs/métabolisme , Tumeurs/thérapie , Tumeurs expérimentales/immunologie , Tumeurs expérimentales/métabolisme , Tumeurs expérimentales/thérapie
17.
BMC Cell Biol ; 18(1): 30, 2017 08 29.
Article de Anglais | MEDLINE | ID: mdl-28851289

RÉSUMÉ

BACKGROUND: Generalized methods for understanding the cell biology of non-model species are quite rare, yet very much needed. In order to address this issue, we have modified a technique traditionally used in the biomedical field for ecological and evolutionary research. Fluorescent activated cell sorting (FACS) is often used for sorting and identifying cell populations. In this study, we developed a method to identify and isolate different cell populations in corals and other cnidarians. METHODS: Using fluorescence-activated cell sorting (FACS), coral cell suspension were sorted into different cellular populations using fluorescent cell markers that are non-species specific. Over 30 different cell markers were tested. Additionally, cell suspension from Aiptasia pallida was also tested, and a phagocytosis test was done as a downstream functional assay. RESULTS: We found that 24 of the screened markers positively labeled coral cells and 16 differentiated cell sub-populations. We identified 12 different cellular sub-populations using three markers, and found that each sub-population is primarily homogeneous. Lastly, we verified this technique in a sea anemone, Aiptasia pallida, and found that with minor modifications, a similar gating strategy can be successfully applied. Additionally, within A. pallida, we show elevated phagocytosis of sorted cells based on an immune associated marker. CONCLUSIONS: In this study, we successfully adapted FACS for isolating coral cell populations and conclude that this technique is translatable for future use in other species. This technique has the potential to be used for different types of studies on the cellular stress response and other immunological studies.


Sujet(s)
Anthozoa/cytologie , Marqueurs biologiques/analyse , Séparation cellulaire/méthodes , Cytométrie en flux , Animaux , Reproductibilité des résultats , Anémones de mer/cytologie , Coloration et marquage
18.
Nature ; 545(7655): 495-499, 2017 05 25.
Article de Anglais | MEDLINE | ID: mdl-28514441

RÉSUMÉ

Programmed cell death protein 1 (PD-1) is an immune checkpoint receptor that is upregulated on activated T cells for the induction of immune tolerance. Tumour cells frequently overexpress the ligand for PD-1, programmed cell death ligand 1 (PD-L1), facilitating their escape from the immune system. Monoclonal antibodies that block the interaction between PD-1 and PD-L1, by binding to either the ligand or receptor, have shown notable clinical efficacy in patients with a variety of cancers, including melanoma, colorectal cancer, non-small-cell lung cancer and Hodgkin's lymphoma. Although it is well established that PD-1-PD-L1 blockade activates T cells, little is known about the role that this pathway may have in tumour-associated macrophages (TAMs). Here we show that both mouse and human TAMs express PD-1. TAM PD-1 expression increases over time in mouse models of cancer and with increasing disease stage in primary human cancers. TAM PD-1 expression correlates negatively with phagocytic potency against tumour cells, and blockade of PD-1-PD-L1 in vivo increases macrophage phagocytosis, reduces tumour growth and lengthens the survival of mice in mouse models of cancer in a macrophage-dependent fashion. This suggests that PD-1-PD-L1 therapies may also function through a direct effect on macrophages, with substantial implications for the treatment of cancer with these agents.


Sujet(s)
Tumeurs du côlon/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Phagocytose , Récepteur-1 de mort cellulaire programmée/métabolisme , Animaux , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs du côlon/traitement médicamenteux , Tumeurs du côlon/anatomopathologie , Modèles animaux de maladie humaine , Femelle , Humains , Macrophages/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de lignée BALB C , Stadification tumorale , Phagocytose/effets des médicaments et des substances chimiques , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Tests d'activité antitumorale sur modèle de xénogreffe
19.
Proc Natl Acad Sci U S A ; 114(18): 4757-4762, 2017 05 02.
Article de Anglais | MEDLINE | ID: mdl-28424250

RÉSUMÉ

Fibrotic diseases are not well-understood. They represent a number of different diseases that are characterized by the development of severe organ fibrosis without any obvious cause, such as the devastating diseases idiopathic pulmonary fibrosis (IPF) and scleroderma. These diseases have a poor prognosis comparable with endstage cancer and are uncurable. Given the phenotypic differences, it was assumed that the different fibrotic diseases also have different pathomechanisms. Here, we demonstrate that many endstage fibrotic diseases, including IPF; scleroderma; myelofibrosis; kidney-, pancreas-, and heart-fibrosis; and nonalcoholic steatohepatosis converge in the activation of the AP1 transcription factor c-JUN in the pathologic fibroblasts. Expression of the related AP1 transcription factor FRA2 was restricted to pulmonary artery hypertension. Induction of c-Jun in mice was sufficient to induce severe fibrosis in multiple organs and steatohepatosis, which was dependent on sustained c-Jun expression. Single cell mass cytometry revealed that c-Jun activates multiple signaling pathways in mice, including pAkt and CD47, which were also induced in human disease. αCD47 antibody treatment and VEGF or PI3K inhibition reversed various organ c-Jun-mediated fibroses in vivo. These data suggest that c-JUN is a central molecular mediator of most fibrotic conditions.


Sujet(s)
Fibrose pulmonaire idiopathique , Myélofibrose primitive , Protéines proto-oncogènes c-jun , Sclérodermie systémique , Facteur de transcription AP-1 , Antigène-2 apparenté à fos/génétique , Antigène-2 apparenté à fos/métabolisme , Humains , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/métabolisme , Myélofibrose primitive/génétique , Myélofibrose primitive/métabolisme , Protéines proto-oncogènes c-jun/génétique , Protéines proto-oncogènes c-jun/métabolisme , Sclérodermie systémique/génétique , Sclérodermie systémique/métabolisme , Facteur de transcription AP-1/génétique , Facteur de transcription AP-1/métabolisme
20.
Proc Natl Acad Sci U S A ; 114(14): 3654-3659, 2017 04 04.
Article de Anglais | MEDLINE | ID: mdl-28330992

RÉSUMÉ

The regeneration of organ morphology and function following tissue loss is critical to restore normal physiology, yet few cases are documented in mammalian postnatal life. Partial hepatectomy of the adult mammalian liver activates compensatory hepatocyte hypertrophy and cell division across remaining lobes, resulting in restitution of organ mass but with permanent alteration of architecture. Here, we identify a time window in early postnatal life wherein partial amputation culminates in a localized regeneration instead of global hypertrophy and proliferation. Quantifications of liver mass, enzymatic activity, and immunohistochemistry demonstrate that damaged lobes underwent multilineage regeneration, reforming a lobe often indistinguishable from undamaged ones. Clonal analysis during regeneration reveals local clonal expansions of hepatocyte stem/progenitors at injured sites that are lineage but not fate restricted. Tetrachimeric mice show clonal selection occurs during development with further selections following injury. Surviving progenitors associate mainly with central veins, in a pattern of selection different from that of normal development. These results illuminate a previously unknown program of liver regeneration after acute injury and allow for exploration of latent regenerative programs with potential applications to adult liver regeneration.


Sujet(s)
Régénération hépatique , Foie/cytologie , Foie/chirurgie , Cellules souches/cytologie , Animaux , Animaux nouveau-nés , Division cellulaire , Lignage cellulaire , Clones cellulaires , Foie/physiologie , Souris , Modèles biologiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE