Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Blood Adv ; 8(8): 1981-1990, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38507738

ABSTRACT

ABSTRACT: Bruton's tyrosine kinase (BTK) is an enzyme needed for B-cell survival, and its inhibitors have become potent targeted medicines for the treatment of B-cell malignancies. The initial activation event of cytoplasmic protein-tyrosine kinases is the phosphorylation of a conserved regulatory tyrosine in the catalytic domain, which in BTK is represented by tyrosine 551. In addition, the tyrosine 223 (Y223) residue in the SRC homology 3 (SH3) domain has, for more than 2 decades, generally been considered necessary for full enzymatic activity. The initial recognition of its potential importance stems from transformation assays using nonlymphoid cells. To determine the biological significance of this residue, we generated CRISPR-Cas-mediated knockin mice carrying a tyrosine to phenylalanine substitution (Y223F), maintaining aromaticity and bulkiness while prohibiting phosphorylation. Using a battery of assays to study leukocyte subsets and the morphology of lymphoid organs, as well as the humoral immune responses, we were unable to detect any difference between wild-type mice and the Y223F mutant. Mice resistant to irreversible BTK inhibitors, through a cysteine 481 to serine substitution (C481S), served as an additional immunization control and mounted similar humoral immune responses as Y223F and wild-type animals. Collectively, our findings suggest that phosphorylation of Y223 serves as a useful proxy for phosphorylation of phospholipase Cγ2 (PLCG2), the endogenous substrate of BTK. However, in contrast to a frequently held conception, this posttranslational modification is dispensable for the function of BTK.


Subject(s)
Protein-Tyrosine Kinases , src Homology Domains , Mice , Animals , Agammaglobulinaemia Tyrosine Kinase , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Tyrosine
3.
Nat Commun ; 14(1): 3092, 2023 05 29.
Article in English | MEDLINE | ID: mdl-37248239

ABSTRACT

In this study we use comparative genomics to uncover a gene with uncharacterized function (1700011H14Rik/C14orf105/CCDC198), which we hereby name FAME (Factor Associated with Metabolism and Energy). We observe that FAME shows an unusually high evolutionary divergence in birds and mammals. Through the comparison of single nucleotide polymorphisms, we identify gene flow of FAME from Neandertals into modern humans. We conduct knockout experiments on animals and observe altered body weight and decreased energy expenditure in Fame knockout animals, corresponding to genome-wide association studies linking FAME with higher body mass index in humans. Gene expression and subcellular localization analyses reveal that FAME is a membrane-bound protein enriched in the kidneys. Although the gene knockout results in structurally normal kidneys, we detect higher albumin in urine and lowered ferritin in the blood. Through experimental validation, we confirm interactions between FAME and ferritin and show co-localization in vesicular and plasma membranes.


Subject(s)
Energy Metabolism , Genome-Wide Association Study , Animals , Humans , Body Weight , Energy Metabolism/genetics , Ferritins/genetics , Kidney , Neanderthals
4.
Blood ; 142(1): 73-89, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37018663

ABSTRACT

Although tyrosine kinase inhibitors (TKIs) are effective in treating chronic myeloid leukemia (CML), they often fail to eradicate the leukemia-initiating stem cells (LSCs), causing disease persistence and relapse. Evidence indicates that LSC persistence may be because of bone marrow (BM) niche protection; however, little is known about the underlying mechanisms. Herein, we molecularly and functionally characterize BM niches in patients with CML at diagnosis and reveal the altered niche composition and function in these patients. Long-term culture initiating cell assay showed that the mesenchymal stem cells from patients with CML displayed an enhanced supporting capacity for normal and CML BM CD34+CD38- cells. Molecularly, RNA sequencing detected dysregulated cytokine and growth factor expression in the BM cellular niches of patients with CML. Among them, CXCL14 was lost in the BM cellular niches in contrast to its expression in healthy BM. Restoring CXCL14 significantly inhibited CML LSC maintenance and enhanced their response to imatinib in vitro, and CML engraftment in vivo in NSG-SGM3 mice. Importantly, CXCL14 treatment dramatically inhibited CML engraftment in patient-derived xenografted NSG-SGM3 mice, even to a greater degree than imatinib, and this inhibition persisted in patients with suboptimal TKI response. Mechanistically, CXCL14 upregulated inflammatory cytokine signaling but downregulated mTOR signaling and oxidative phosphorylation in CML LSCs. Together, we have discovered a suppressive role of CXCL14 in CML LSC growth. CXCL14 might offer a treatment option targeting CML LSCs.


Subject(s)
Bone Marrow , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Animals , Mice , Bone Marrow/metabolism , Chemokines, CXC/metabolism , Chemokines, CXC/pharmacology , Chemokines, CXC/therapeutic use , Cytokines/metabolism , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Neoplastic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction
5.
EMBO J ; 41(17): e108780, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35815410

ABSTRACT

Schwann cell precursors (SCPs) are nerve-associated progenitors that can generate myelinating and non-myelinating Schwann cells but also are multipotent like the neural crest cells from which they originate. SCPs are omnipresent along outgrowing peripheral nerves throughout the body of vertebrate embryos. By using single-cell transcriptomics to generate a gene expression atlas of the entire neural crest lineage, we show that early SCPs and late migratory crest cells have similar transcriptional profiles characterised by a multipotent "hub" state containing cells biased towards traditional neural crest fates. SCPs keep diverging from the neural crest after being primed towards terminal Schwann cells and other fates, with different subtypes residing in distinct anatomical locations. Functional experiments using CRISPR-Cas9 loss-of-function further show that knockout of the common "hub" gene Sox8 causes defects in neural crest-derived cells along peripheral nerves by facilitating differentiation of SCPs towards sympathoadrenal fates. Finally, specific tumour populations found in melanoma, neurofibroma and neuroblastoma map to different stages of SCP/Schwann cell development. Overall, SCPs resemble migrating neural crest cells that maintain multipotency and become transcriptionally primed towards distinct lineages.


Subject(s)
Neural Crest , Schwann Cells , Cell Differentiation/physiology , Neurogenesis/physiology , Peripheral Nerves , Schwann Cells/metabolism
6.
Front Immunol ; 13: 854312, 2022.
Article in English | MEDLINE | ID: mdl-35757763

ABSTRACT

Natural killer (NK) cells play roles in viral clearance and early surveillance against malignant transformation, yet our knowledge of the underlying mechanisms controlling their development and functions remain incomplete. To reveal cell fate-determining pathways in NK cell progenitors (NKP), we utilized an unbiased approach and generated comprehensive gene expression profiles of NK cell progenitors. We found that the NK cell program was gradually established in the CLP to preNKP and preNKP to rNKP transitions. In line with FOXO1 and FOXO3 being co-expressed through the NK developmental trajectory, the loss of both perturbed the establishment of the NK cell program and caused stalling in both NK cell development and maturation. In addition, we found that the combined loss of FOXO1 and FOXO3 caused specific changes to the composition of the non-cytotoxic innate lymphoid cell (ILC) subsets in bone marrow, spleen, and thymus. By combining transcriptome and chromatin profiling, we revealed that FOXO TFs ensure proper NK cell development at various lineage-commitment stages through orchestrating distinct molecular mechanisms. Combined FOXO1 and FOXO3 deficiency in common and innate lymphoid cell progenitors resulted in reduced expression of genes associated with NK cell development including ETS-1 and their downstream target genes. Lastly, we found that FOXO1 and FOXO3 controlled the survival of committed NK cells via gene regulation of IL-15Rß (CD122) on rNKPs and bone marrow NK cells. Overall, we revealed that FOXO1 and FOXO3 function in a coordinated manner to regulate essential developmental genes at multiple stages during murine NK cell and ILC lineage commitment.


Subject(s)
Forkhead Box Protein O1 , Forkhead Box Protein O3 , Killer Cells, Natural , Lymphoid Progenitor Cells , Animals , Cell Differentiation/immunology , Forkhead Box Protein O1/immunology , Forkhead Box Protein O3/immunology , Immunity, Innate , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Mice , Mice, Inbred C57BL
7.
Front Immunol ; 13: 880668, 2022.
Article in English | MEDLINE | ID: mdl-35603175

ABSTRACT

The development of B cells relies on an intricate network of transcription factors critical for developmental progression and lineage commitment. In the B cell developmental trajectory, a temporal switch from predominant Foxo3 to Foxo1 expression occurs at the CLP stage. Utilizing VAV-iCre mediated conditional deletion, we found that the loss of FOXO3 impaired B cell development from LMPP down to B cell precursors, while the loss of FOXO1 impaired B cell commitment and resulted in a complete developmental block at the CD25 negative proB cell stage. Strikingly, the combined loss of FOXO1 and FOXO3 resulted in the failure to restrict the myeloid potential of CLPs and the complete loss of the B cell lineage. This is underpinned by the failure to enforce the early B-lineage gene regulatory circuitry upon a predominantly pre-established open chromatin landscape. Altogether, this demonstrates that FOXO3 and FOXO1 cooperatively govern early lineage restriction and initiation of B-lineage commitment in CLPs.


Subject(s)
Hematopoiesis , Lymphoid Progenitor Cells , B-Lymphocytes/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Hematopoiesis/genetics , Lymphoid Progenitor Cells/metabolism , Precursor Cells, B-Lymphoid/metabolism
8.
Curr Biol ; 32(12): 2596-2609.e7, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35561678

ABSTRACT

Reef-building corals are endangered animals with a complex colonial organization. Physiological mechanisms connecting multiple polyps and integrating them into a coral colony are still enigmatic. Using live imaging, particle tracking, and mathematical modeling, we reveal how corals connect individual polyps and form integrated polyp groups via species-specific, complex, and stable networks of currents at their surface. These currents involve surface mucus of different concentrations, which regulate joint feeding of the colony. Inside the coral, within the gastrovascular system, we expose the complexity of bidirectional branching streams that connect individual polyps. This system of canals extends the surface area by 4-fold and might improve communication, nutrient supply, and symbiont transfer. Thus, individual polyps integrate via complex liquid dynamics on the surface and inside the colony.


Subject(s)
Anthozoa , Animals , Anthozoa/physiology , Coral Reefs , Environment , Species Specificity
9.
J Vis Exp ; (176)2021 10 28.
Article in English | MEDLINE | ID: mdl-34779437

ABSTRACT

Mouse and human teeth represent challenging organs for quick and efficient cell isolation for single-cell transcriptomic or other applications. The dental pulp tissue, rich in the extracellular matrix, requires a long and tedious dissociation process that is typically beyond the reasonable time for single-cell transcriptomics. For avoiding artificial changes in gene expression, the time elapsed from euthanizing an animal until the analysis of single cells needs to be minimized. This work presents a fast protocol enabling to obtain single-cell suspension from mouse and human teeth in an excellent quality suitable for scRNA-seq (single-cell RNA-sequencing). This protocol is based on accelerated tissue isolation steps, enzymatic digestion, and subsequent preparation of final single-cell suspension. This enables fast and gentle processing of tissues and allows using more animal or human samples for obtaining cell suspensions with high viability and minimal transcriptional changes. It is anticipated that this protocol might guide researchers interested in performing the scRNA-seq not only on the mouse or human teeth but also on other extracellular matrix-rich tissues, including cartilage, dense connective tissue, and dermis.


Subject(s)
Gene Expression Profiling , Single-Cell Analysis , Animals , Gene Expression Profiling/methods , Humans , Mice , RNA , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome
10.
Nat Genet ; 53(5): 694-706, 2021 05.
Article in English | MEDLINE | ID: mdl-33833454

ABSTRACT

Characterization of the progression of cellular states during human embryogenesis can provide insights into the origin of pediatric diseases. We examined the transcriptional states of neural crest- and mesoderm-derived lineages differentiating into adrenal glands, kidneys, endothelium and hematopoietic tissue between post-conception weeks 6 and 14 of human development. Our results reveal transitions connecting the intermediate mesoderm and progenitors of organ primordia, the hematopoietic system and endothelial subtypes. Unexpectedly, by using a combination of single-cell transcriptomics and lineage tracing, we found that intra-adrenal sympathoblasts at that stage are directly derived from nerve-associated Schwann cell precursors, similarly to local chromaffin cells, whereas the majority of extra-adrenal sympathoblasts arise from the migratory neural crest. In humans, this process persists during several weeks of development within the large intra-adrenal ganglia-like structures, which may also serve as reservoirs of originating cells in neuroblastoma.


Subject(s)
Cell Lineage , Embryo, Mammalian/metabolism , Neuroblastoma/embryology , Neuroblastoma/genetics , Single-Cell Analysis , Sympathoadrenal System/embryology , Transcriptome/genetics , Animals , Chromaffin Cells/metabolism , Chromaffin Cells/pathology , Cluster Analysis , Embryonic Development , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Infant , Mice , Neural Stem Cells/metabolism , Neuroblastoma/pathology , Schwann Cells/metabolism , Schwann Cells/pathology , Tumor Microenvironment
11.
Blood Adv ; 4(11): 2439-2450, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32492159

ABSTRACT

Pharmacological inhibitors of Bruton tyrosine kinase (BTK) have revolutionized treatment of B-lymphocyte malignancies and show great promise for dampening autoimmunity. The predominant BTK inhibitors tether irreversibly by covalently binding to cysteine 481 in the BTK catalytic domain. Substitution of cysteine 481 for serine (C481S) is the most common mechanism for acquired drug resistance. We generated a novel C481S knock-in mouse model and, using a battery of tests, no overt B-lymphocyte phenotype was found. B lymphocytes from C481S animals were resistant to irreversible, but sensitive to reversible, BTK inhibitors. In contrast, irreversible inhibitors equally impaired T-lymphocyte activation in mice, mimicking the effect of treatment in patients. This demonstrates that T-lymphocyte blockage is independent of BTK. We suggest that the C481S knock-in mouse can serve as a useful tool for the study of BTK-independent effects of irreversible inhibitors, allowing for the identification of novel therapeutic targets and pinpointing potential side effects.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , B-Lymphocytes , Protein Kinase Inhibitors , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Animals , Humans , Mice , Mice, Inbred C57BL , Mutation , Protein Kinase Inhibitors/pharmacology
12.
Leukemia ; 34(1): 271-282, 2020 01.
Article in English | MEDLINE | ID: mdl-31375745

ABSTRACT

Established cell culture systems have failed to accurately recapitulate key features of terminal erythroid maturation, hampering our ability to in vitro model and treat diseases with impaired erythropoiesis such as myelodysplastic syndromes with ring sideroblasts (MDS-RS). We developed an efficient and robust three-dimensional (3D) scaffold culture model supporting terminal erythroid differentiation from both mononuclear (MNC) or CD34+-enriched primary bone marrow cells from healthy donors and MDS-RS patients. While CD34+ cells did not proliferate beyond two weeks in 2D suspension cultures, the 3D scaffolds supported CD34+ and MNC erythroid proliferation over four weeks demonstrating the importance of the 3D environment. CD34+ cells cultured in 3D facilitated the highest expansion and maturation of erythroid cells, including generation of erythroblastic islands and enucleated erythrocytes, while MNCs supported multi-lineage hemopoietic differentiation and cytokine secretion relevant for MDS-RS. Importantly, MDS-RS 3D-cultures supported de novo generation of ring sideroblasts and maintenance of the mutated clone. The 3D cultures effectively model a clonal disease characterized by terminal erythroid failure and can be used to assess therapeutic compounds.


Subject(s)
Cell Culture Techniques , Erythropoiesis , Myelodysplastic Syndromes , Antigens, CD34 , Cell Differentiation/physiology , Cells, Cultured , Erythroid Precursor Cells/cytology , Humans , Mesenchymal Stem Cells/cytology
13.
Front Immunol ; 10: 455, 2019.
Article in English | MEDLINE | ID: mdl-30936870

ABSTRACT

The apparition of adaptive immunity in Gnathostomata correlates with the expansion of the E-protein family to encompass E2-2, HEB, and E2A. Within the family, E2-2 and HEB are more closely evolutionarily related but their concerted action in hematopoiesis remains to be explored. Here we show that the combined disruption of E2-2 and HEB results in failure to express the early lymphoid program in Common lymphoid precursors (CLPs) and a near complete block in B-cell development. In the thymus, Early T-cell progenitors (ETPs) were reduced and T-cell development perturbed, resulting in reduced CD4 T- and increased γδ T-cell numbers. In contrast, hematopoietic stem cells (HSCs), erythro-myeloid progenitors, and innate immune cells were unaffected showing that E2-2 and HEB are dispensable for the ancestral hematopoietic lineages. Taken together, this E-protein dependence suggests that the appearance of the full Gnathostomata E-protein repertoire was critical to reinforce the gene regulatory circuits that drove the emergence and expansion of the lineages constituting humoral immunity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation/physiology , Immunity, Humoral/physiology , Leukopoiesis/physiology , Lymphoid Progenitor Cells/pathology , Transcription Factor 4/physiology , Vertebrates/immunology , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/immunology , Biological Evolution , Cell Lineage , Evolution, Molecular , Gene Duplication , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Lymphocyte Subsets/pathology , Mice , Mice, Inbred C57BL , Multigene Family , Phylogeny , Sequence Alignment , Sequence Homology, Amino Acid , Spleen/pathology , Transcription Factor 4/deficiency , Transcription Factor 4/immunology
14.
Nature ; 567(7747): 234-238, 2019 03.
Article in English | MEDLINE | ID: mdl-30814736

ABSTRACT

Longitudinal bone growth in children is sustained by growth plates, narrow discs of cartilage that provide a continuous supply of chondrocytes for endochondral ossification1. However, it remains unknown how this supply is maintained throughout childhood growth. Chondroprogenitors in the resting zone are thought to be gradually consumed as they supply cells for longitudinal growth1,2, but this model has never been proved. Here, using clonal genetic tracing with multicolour reporters and functional perturbations, we demonstrate that longitudinal growth during the fetal and neonatal periods involves depletion of chondroprogenitors, whereas later in life, coinciding with the formation of the secondary ossification centre, chondroprogenitors acquire the capacity for self-renewal, resulting in the formation of large, stable monoclonal columns of chondrocytes. Simultaneously, chondroprogenitors begin to express stem cell markers and undergo symmetric cell division. Regulation of the pool of self-renewing progenitors involves the hedgehog and mammalian target of rapamycin complex 1 (mTORC1) signalling pathways. Our findings indicate that a stem cell niche develops postnatally in the epiphyseal growth plate, which provides a continuous supply of chondrocytes over a prolonged period.


Subject(s)
Chondrocytes/cytology , Clone Cells/cytology , Growth Plate/cytology , Stem Cell Niche/physiology , Aging , Animals , Cartilage/cytology , Cell Self Renewal , Clone Cells/metabolism , Female , Growth Plate/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice
15.
Blood Adv ; 2(12): 1480-1494, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29945938

ABSTRACT

Despite increasing evidence for the involvement of bone marrow (BM) hematopoietic stem cell niche in leukemogenesis, how BM mesenchymal stem and progenitor cells (MSPCs) contribute to leukemia niche formation and progression remains unclear. Using an MLL-AF9 acute myeloid leukemia (AML) mouse model, we demonstrate dynamic alterations of BM cellular niche components, including MSPCs and endothelial cells during AML development and its association with AML engraftment. Primary patient AML cells also induced similar niche alterations in xenografted mice. AML cell infiltration in BM causes an expansion of early B-cell factor 2+ (Ebf2+) MSPCs with reduced Cxcl12 expression and enhanced generation of more differentiated mesenchymal progenitor cells. Importantly, in vivo fate-mapping indicates that Ebf2+ MSPCs participated in AML niche formation. Ebf2+ cell deletion accelerated the AML development. These data suggest that native BM MSPCs may suppress AML. However, they can be remodeled by AML cells to form leukemic niche that might contribute to AML progression. AML induced dysregulation of hematopoietic niche factors like Angptl1, Cxcl12, Kitl, Il6, Nov, and Spp1 in AML BM MSPCs, which was associated with AML engraftment and partially appeared before the massive expansion of AML cells, indicating the possible involvement of the niche factors in AML progression. Our study demonstrates distinct dynamic features and roles of BM MSPCs during AML development.


Subject(s)
Carcinogenesis/pathology , Leukemia, Myeloid, Acute/pathology , Mesenchymal Stem Cells/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors , Bone Marrow/pathology , Mice , Myeloid-Lymphoid Leukemia Protein , Oncogene Proteins, Fusion , Stem Cell Niche/genetics , Transplantation, Heterologous , Tumor Burden , Tumor Microenvironment
16.
Blood Adv ; 2(5): 534-548, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29514790

ABSTRACT

Mutations of signal-induced proliferation-associated gene 1 (SIPA1), a RAP1 GTPase-activating protein, were reported in patients with juvenile myelomonocytic leukemia, a childhood myelodysplastic/myeloproliferative neoplasm (MDS/MPN). Sipa1 deficiency in mice leads to the development of age-dependent MPN. However, Sipa1 expression in bone marrow (BM) microenvironment and its effect on the pathogenesis of MPN remain unclear. We here report that Sipa1 is expressed in human and mouse BM stromal cells and downregulated in these cells from patients with MPN or MDS/MPN at diagnosis. By using the Sipa1-/- MPN mouse model, we find that Sipa1 deletion causes phenotypic and functional alterations of BM mesenchymal stem and progenitor cells prior to the initiation of the MPN. Importantly, the altered Sipa1-/- BM niche is required for the development of MDS/MPN following transplantation of normal hematopoietic cells. RNA sequencing reveals an enhanced inflammatory cytokine signaling and dysregulated Dicer1, Kitl, Angptl1, Cxcl12, and Thpo in the Sipa1-/- BM cellular niches. Our data suggest that Sipa1 expression in the BM niche is critical for maintaining BM niche homeostasis. Moreover, Sipa1 loss-induced BM niche alterations likely enable evolution of clonal hematopoiesis to the hematological malignancies. Therefore, restoring Sipa1 expression or modulating the altered signaling pathways involved might offer therapeutic potential for MPN.


Subject(s)
Bone Marrow Cells/pathology , GTPase-Activating Proteins/deficiency , Leukemia/etiology , Myeloproliferative Disorders/etiology , Nuclear Proteins/deficiency , Stem Cell Niche , Animals , Homeostasis , Humans , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL
17.
FASEB J ; 31(3): 1067-1084, 2017 03.
Article in English | MEDLINE | ID: mdl-27965322

ABSTRACT

Articular cartilage has little regenerative capacity. Recently, genetic lineage tracing experiments have revealed chondrocyte progenitors at the articular surface. We further characterized these progenitors by using in vivo genetic approaches. Histone H2B-green fluorescent protein retention revealed that superficial cells divide more slowly than underlying articular chondrocytes. Clonal genetic tracing combined with immunohistochemistry revealed that superficial cells renew their number by symmetric division, express mesenchymal stem cell markers, and generate chondrocytes via both asymmetric and symmetric differentiation. Quantitative analysis of cellular kinetics, in combination with phosphotungstic acid-enhanced micro-computed tomography, showed that superficial cells generate chondrocytes and contribute to the growth and reshaping of articular cartilage. Furthermore, we found that cartilage renewal occurs as the progeny of superficial cells fully replace fetal chondrocytes during early postnatal life. Thus, superficial cells are self-renewing progenitors that are capable of maintaining their own population and fulfilling criteria of unipotent adult stem cells. Furthermore, the progeny of these cells reconstitute adult articular cartilage de novo, entirely substituting fetal chondrocytes.-Li, L., Newton, P. T., Bouderlique, T., Sejnohova, M., Zikmund, T., Kozhemyakina, E., Xie, M., Krivanek, J., Kaiser, J., Qian, H., Dyachuk, V., Lassar, A. B., Warman, M. L., Barenius, B., Adameyko, I., Chagin, A. S. Superficial cells are self-renewing chondrocyte progenitors, which form the articular cartilage in juvenile mice.


Subject(s)
Adult Stem Cells/cytology , Cartilage, Articular/cytology , Chondrocytes/cytology , Chondrogenesis , Animals , Cartilage, Articular/physiology , Mice , Regeneration
18.
Ann Rheum Dis ; 75(3): 627-31, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26438374

ABSTRACT

OBJECTIVES: It has been suggested that the lysosomal recycling process called macro-autophagy plays a role in osteoarthritis development. We thus decided to genetically ablate the autophagy-indispensable Atg5 gene specifically in chondrocytes and analyse the development of osteoarthritis upon aging and in a post-traumatic model. METHODS: Mice lacking the Atg5 gene in their chondrocytes (Atg5cKO) were generated by crossing Atg5-floxed mice with transgenic mice that expressed cre recombinase driven by the collagen type 2 promoter. Animals were analysed at the age of 2, 6 and 12 months for age-related osteoarthritis or underwent mini-open partial medial meniscectomy at 2 months of age and were analysed 1 or 2 months after surgery. We evaluated osteoarthritis using the Osteoarthritis Research Society International (OARSI) scoring on safranin-O-stained samples. Cell death was evaluated by terminal deoxy-nucleotidyl-transferase-mediated deoxy-UTP nick end labelling (TUNEL) and by immunostaining of cleaved caspases. RESULTS: We observed the development of osteoarthritis in Atg5cKO mice with aging including fibrillation and loss of proteoglycans, which was particularly severe in males. The ablation of Atg5 was associated with an increased cell death as assessed by TUNEL, cleaved caspase 3 and cleaved caspase 9. Surprisingly, no difference in the development of post-traumatic osteoarthritis was observed between Atg5cKO and control mice. CONCLUSIONS: Autophagy protects from age-related osteoarthritis by facilitating chondrocyte survival.


Subject(s)
Autophagy/genetics , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Microtubule-Associated Proteins/genetics , Osteoarthritis/genetics , Animals , Autophagy-Related Protein 5 , Cartilage, Articular/cytology , Caspases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Osteoarthritis/metabolism , Proteoglycans/metabolism , Tibial Meniscus Injuries
19.
Autophagy ; 11(9): 1594-607, 2015.
Article in English | MEDLINE | ID: mdl-26259639

ABSTRACT

Mechanistic target of rapamycin (serine/threonine kinase) complex 1 (MTORC1) is a protein-signaling complex at the fulcrum of anabolic and catabolic processes, which acts depending on wide-ranging environmental cues. It is generally accepted that lysosomes facilitate MTORC1 activation by generating an internal pool of amino acids. Amino acids activate MTORC1 by stimulating its translocation to the lysosomal membrane where it forms a super-complex involving the lysosomal-membrane-bound vacuolar-type H(+)-ATPase (v-ATPase) proton pump. This translocation and MTORC1 activation require functional lysosomes. Here we found that, in contrast to this well-accepted concept, in epiphyseal chondrocytes inhibition of lysosomal activity by v-ATPase inhibitors bafilomycin A1 or concanamycin A potently activated MTORC1 signaling. The activity of MTORC1 was visualized by phosphorylated forms of RPS6 (ribosomal protein S6) and EIF4EBP1, 2 well-known downstream targets of MTORC1. Maximal RPS6 phosphorylation was observed at 48-h treatment and reached as high as a 12-fold increase (p < 0.018). This activation of MTORC1 was further confirmed in bone organ culture and promoted potent stimulation of longitudinal growth (p < 0.001). Importantly, the same effect was observed in ATG5 (autophagy-related 5)-deficient bones suggesting a macroautophagy-independent mechanism of MTORC1 inhibition by lysosomes. Thus, our data show that in epiphyseal chondrocytes lysosomes inhibit MTORC1 in a macroautophagy-independent manner and this inhibition likely depends on v-ATPase activity.


Subject(s)
Autophagy/drug effects , Chondrocytes/metabolism , Lysosomes/metabolism , Macrolides/pharmacology , Multiprotein Complexes/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Bone Development/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrocytes/drug effects , Chondrocytes/pathology , Hypertrophy , Lysosomes/drug effects , Mechanistic Target of Rapamycin Complex 1 , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Rats , Ribosomal Protein S6/metabolism , Tumor Suppressor Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
20.
J Bone Miner Res ; 30(12): 2249-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26077727

ABSTRACT

Longitudinal bone growth takes place in epiphyseal growth plates located in the ends of long bones. The growth plate consists of chondrocytes traversing from the undifferentiated (resting zone) to the terminally differentiated (hypertrophic zone) stage. Autophagy is an intracellular catabolic process of lysosome-dependent recycling of intracellular organelles and protein complexes. Autophagy is activated during nutritionally depleted or hypoxic conditions in order to facilitate cell survival. Chondrocytes in the middle of the growth plate are hypoxic and nutritionally depleted owing to the avascular nature of the growth plate. Accordingly, autophagy may facilitate their survival. To explore the role of autophagy in chondrocyte survival and constitutional bone growth, we generated mice with cartilage-specific ablation of either Atg5 (Atg5cKO) or Atg7 (Atg7cKO) by crossing Atg5 or Atg7 floxed mice with cartilage-specific collagen type 2 promoter-driven Cre. Both Atg5cKO and Atg7cKO mice showed growth retardation associated with enhanced chondrocyte cell death and decreased cell proliferation. Similarly, inhibition of autophagy by Bafilomycin A1 (Baf) or 3-methyladenine (3MA) promoted cell death in cultured slices of human growth plate tissue. To delineate the underlying mechanisms we employed ex vivo cultures of mouse metatarsal bones and RCJ3.IC5.18 rat chondrogenic cell line. Baf or 3MA impaired metatarsal bone growth associated with processing of caspase-3 and massive cell death. Similarly, treatment of RCJ3.IC5.18 chondrogenic cells by Baf also showed massive cell death and caspase-3 cleavage. This was associated with activation of caspase-9 and cytochrome C release. Altogether, our data suggest that autophagy is important for chondrocyte survival, and inhibition of this process leads to stunted growth and caspase-dependent death of chondrocytes.


Subject(s)
Caspases/metabolism , Chondrocytes/cytology , Gene Deletion , Microtubule-Associated Proteins/genetics , Adenine/analogs & derivatives , Adenine/metabolism , Animals , Apoptosis , Autophagy , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Cell Death , Cell Line , Cell Proliferation , Cell Survival , Collagen Type II/metabolism , Cytochromes c/metabolism , Growth Plate/growth & development , Humans , Hypoxia , Immunohistochemistry , In Situ Hybridization , Macrolides/metabolism , Metatarsal Bones/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/physiology , Proteins/genetics , Rats , Ubiquitin-Activating Enzymes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...