Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Oct 29.
Article in English | MEDLINE | ID: mdl-37961493

ABSTRACT

Haematopoietic stem cells (HSCs) reside in specialized microenvironments, also referred to as niches, and it has been widely believed that HSC numbers are determined by the niche size alone 1-5 . However, the vast excess of the number of niche cells over that of HSCs raises questions about this model. We initially established a mathematical model of niche availability and occupancy, which predicted that HSC numbers are restricted at both systemic and local levels. To address this question experimentally, we developed a femoral bone transplantation system, enabling us to increase the number of available HSC niches. We found that the addition of niches does not alter total HSC numbers in the body, regardless of whether the endogenous (host) niche is intact or defective, suggesting that HSC numbers are limited at the systemic level. Additionally, HSC numbers in transplanted wild-type femurs did not increase beyond physiological levels when HSCs were mobilized from defective endogenous niches to the periphery, indicating that HSC numbers are also constrained at the local level. Our study demonstrates that HSC numbers are not solely determined by niche availability, thereby rewriting the long-standing model for the regulation of HSC numbers.

2.
Nature ; 607(7919): 578-584, 2022 07.
Article in English | MEDLINE | ID: mdl-35636458

ABSTRACT

The nervous and immune systems are intricately linked1. Although psychological stress is known to modulate immune function, mechanistic pathways linking stress networks in the brain to peripheral leukocytes remain poorly understood2. Here we show that distinct brain regions shape leukocyte distribution and function throughout the body during acute stress in mice. Using optogenetics and chemogenetics, we demonstrate that motor circuits induce rapid neutrophil mobilization from the bone marrow to peripheral tissues through skeletal-muscle-derived neutrophil-attracting chemokines. Conversely, the paraventricular hypothalamus controls monocyte and lymphocyte egress from secondary lymphoid organs and blood to the bone marrow through direct, cell-intrinsic glucocorticoid signalling. These stress-induced, counter-directional, population-wide leukocyte shifts are associated with altered disease susceptibility. On the one hand, acute stress changes innate immunity by reprogramming neutrophils and directing their recruitment to sites of injury. On the other hand, corticotropin-releasing hormone neuron-mediated leukocyte shifts protect against the acquisition of autoimmunity, but impair immunity to SARS-CoV-2 and influenza infection. Collectively, these data show that distinct brain regions differentially and rapidly tailor the leukocyte landscape during psychological stress, therefore calibrating the ability of the immune system to respond to physical threats.


Subject(s)
Brain , Fear , Leukocytes , Motor Neurons , Neural Pathways , Stress, Psychological , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Brain/cytology , Brain/physiology , COVID-19/immunology , Chemokines/immunology , Disease Susceptibility , Fear/physiology , Glucocorticoids/metabolism , Humans , Leukocytes/cytology , Leukocytes/immunology , Lymphocytes/cytology , Lymphocytes/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Monocytes/cytology , Monocytes/immunology , Motor Neurons/cytology , Motor Neurons/physiology , Neutrophils/cytology , Neutrophils/immunology , Optogenetics , Orthomyxoviridae Infections/immunology , Paraventricular Hypothalamic Nucleus/physiology , SARS-CoV-2/immunology , Stress, Psychological/immunology , Stress, Psychological/physiopathology
3.
Oncogene ; 41(13): 1866-1881, 2022 03.
Article in English | MEDLINE | ID: mdl-35145233

ABSTRACT

Cancer treatments have been revolutionized by the emergence of immune checkpoint blockade therapies. However, only a minority of patients with various tumor types have benefited from such treatments. New strategies focusing on the immune contexture of the tumor tissue microenvironment hold great promises. Here, we created IFNα-overexpressing mesenchymal stromal cells (IFNα-MSCs). Upon direct injection into tumors, we found that these cells are powerful in eliminating several types of tumors. Interestingly, the intra-tumoral injection of IFNα-MSCs could also induce specific anti-tumor effects on distant tumors. These IFNα-MSCs promoted tumor cells to produce CXCL10, which in turn potentiates the infiltration of CD8+ T cells in the tumor site. Furthermore, IFNα-MSCs enhanced the expression of granzyme B (GZMB) in CD8+ T cells and invigorated their cytotoxicity in a Stat3-dependent manner. Genetic ablation of Stat3 in CD8+ T cells impaired the effect of IFNα-MSCs on GZMB expression. Importantly, the combination of IFNα-MSCs and PD-L1 blockade induced an even stronger anti-tumor immunity. Therefore, IFNα-MSCs represent a novel tumor immunotherapy strategy, especially when combined with PD-L1 blockade.


Subject(s)
Mesenchymal Stem Cells , Neoplasms , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Humans , Immunotherapy , Interferon-alpha , Mesenchymal Stem Cells/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Tumor Microenvironment
4.
Cell Stem Cell ; 29(2): 232-247.e7, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35065706

ABSTRACT

Host microbiota crosstalk is essential for the production and functional modulation of blood-cell lineages. Whether, and if so how, the microbiota influences hematopoietic stem cells (HSCs) is unclear. Here, we show that the microbiota regulates HSC self-renewal and differentiation under stress conditions by modulating local iron availability in the bone marrow (BM). In microbiota-depleted mice, HSC self-renewal was enhanced during regeneration, while the commitment toward differentiation was dramatically compromised. Mechanistically, microbiota depletion selectively impaired the recycling of red blood cells (RBCs) by BM macrophages, resulting in reduced local iron levels without affecting systemic iron homeostasis. Limiting iron availability in food (in vivo) or in culture (ex vivo), or by CD169+ macrophage depletion, enhanced HSC self-renewal and expansion. These results reveal an intricate interplay between the microbiota, macrophages, and iron, and their essential roles in regulating critical HSC fate decisions under stress.


Subject(s)
Bone Marrow , Microbiota , Animals , Bone Marrow/physiology , Cell Differentiation , Hematopoietic Stem Cells , Iron , Mice
5.
J Vis Exp ; (176)2021 10 21.
Article in English | MEDLINE | ID: mdl-34747411

ABSTRACT

Type 1 diabetes mellitus (T1DM) is caused by autoimmune destruction of pancreatic ß cells, which results in little or no insulin production. Islet transplantation plays an important role in the treatment of T1DM, with the improved glycometabolic control, the reduced progression of complications, the reduction of hypoglycemic episodes when compared with traditional insulin therapy. The results of phase III clinical trial also demonstrated the safety and efficacy of islet allotransplantation in T1DM. However, the shortage of pancreas donors limits its widespread use. Animals as a source of islets such as the pig offer an alternative choice. Because the architecture of the pig pancreas is different from the islets of mice or humans, the pig islet isolation procedure is still challenging. Since the translation of alternative porcine islet sources (xenogeneic) to the clinical setting for treating T1DM through cellular transplantation is of great importance, a cost-effective, standardized, and reproducible protocol for isolating porcine islets is urgently needed. This manuscript describes a simplified and cost-effective method to isolate and purify adult porcine islets based on the previous protocols that have successfully transplanted porcine islets to non-human primates. This will be a beginners guide without the use of specialized equipment such as a COBE 2991 Cell Processor.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Islets of Langerhans , Animals , Diabetes Mellitus, Type 1/surgery , Islets of Langerhans/surgery , Islets of Langerhans Transplantation/methods , Mice , Pancreas , Swine , Transplantation, Heterologous/methods
6.
Nat Commun ; 12(1): 2522, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33947846

ABSTRACT

Haematopoietic stem cells (HSCs) tightly regulate their quiescence, proliferation, and differentiation to generate blood cells during the entire lifetime. The mechanisms by which these critical activities are balanced are still unclear. Here, we report that Macrophage-Erythroblast Attacher (MAEA, also known as EMP), a receptor thus far only identified in erythroblastic island, is a membrane-associated E3 ubiquitin ligase subunit essential for HSC maintenance and lymphoid potential. Maea is highly expressed in HSCs and its deletion in mice severely impairs HSC quiescence and leads to a lethal myeloproliferative syndrome. Mechanistically, we have found that the surface expression of several haematopoietic cytokine receptors (e.g. MPL, FLT3) is stabilised in the absence of Maea, thereby prolonging their intracellular signalling. This is associated with impaired autophagy flux in HSCs but not in mature haematopoietic cells. Administration of receptor kinase inhibitor or autophagy-inducing compounds rescues the functional defects of Maea-deficient HSCs. Our results suggest that MAEA provides E3 ubiquitin ligase activity, guarding HSC function by restricting cytokine receptor signalling via autophagy.


Subject(s)
Autophagosomes/genetics , Autophagy/genetics , Cell Adhesion Molecules/metabolism , Cytoskeletal Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy/drug effects , Cell Adhesion Molecules/genetics , Cytoskeletal Proteins/genetics , Gene Expression Profiling , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Protein Stability , Receptors, Thrombopoietin/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , fms-Like Tyrosine Kinase 3/metabolism
7.
Nature ; 589(7843): 591-596, 2021 01.
Article in English | MEDLINE | ID: mdl-33361809

ABSTRACT

Haematopoietic stem cells (HSCs) reside in specialized microenvironments in the bone marrow-often referred to as 'niches'-that represent complex regulatory milieux influenced by multiple cellular constituents, including nerves1,2. Although sympathetic nerves are known to regulate the HSC niche3-6, the contribution of nociceptive neurons in the bone marrow remains unclear. Here we show that nociceptive nerves are required for enforced HSC mobilization and that they collaborate with sympathetic nerves to maintain HSCs in the bone marrow. Nociceptor neurons drive granulocyte colony-stimulating factor (G-CSF)-induced HSC mobilization via the secretion of calcitonin gene-related peptide (CGRP). Unlike sympathetic nerves, which regulate HSCs indirectly via the niche3,4,6, CGRP acts directly on HSCs via receptor activity modifying protein 1 (RAMP1) and the calcitonin receptor-like receptor (CALCRL) to promote egress by activating the Gαs/adenylyl cyclase/cAMP pathway. The ingestion of food containing capsaicin-a natural component of chili peppers that can trigger the activation of nociceptive neurons-significantly enhanced HSC mobilization in mice. Targeting the nociceptive nervous system could therefore represent a strategy to improve the yield of HSCs for stem cell-based therapeutic agents.


Subject(s)
Autonomic Pathways , Cell Movement , Hematopoietic Stem Cells/cytology , Nociception/physiology , Nociceptors/physiology , Sympathetic Nervous System/cytology , Adenylyl Cyclases/metabolism , Animals , Autonomic Pathways/drug effects , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Receptor-Like Protein/metabolism , Capsaicin/pharmacology , Cell Movement/drug effects , Cyclic AMP/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Nociception/drug effects , Nociceptors/drug effects , Receptor Activity-Modifying Protein 1/metabolism , Signal Transduction/drug effects , Stem Cell Niche , Sympathetic Nervous System/drug effects
8.
J Exp Med ; 218(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33045060

ABSTRACT

Sickle cell disease (SCD) is a common hereditary hematologic disorder. SCD patients suffer from acute vaso-occlusive episodes (VOEs), chronic organ damage, and premature death, with few therapeutic options. Although severe pain is a major clinical manifestation of SCD, it remains unknown whether nociception plays a role in SCD pathogenesis. To address this question, we generated nociceptor-deficient SCD mice and found, unexpectedly, that the absence of nociception led to more severe and more lethal VOE, indicating that somatosensory nerves protect SCD mice from VOE. Mechanistically, the beneficial effects of sensory nerves were induced by the neuropeptide calcitonin gene-related peptide (CGRP), which acted on hematopoietic cells. Additionally, oral capsaicin consumption, which can activate somatosensory nerves by binding to TRPV1, dramatically alleviated acute VOE and significantly prevented chronic liver and kidney damage in SCD mice. Thus, the manipulation of nociception may provide a promising approach to treat SCD.


Subject(s)
Anemia, Sickle Cell , Capsaicin/pharmacology , Nociceptors/metabolism , Sensory Receptor Cells/metabolism , Vascular Diseases , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Animals , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Humans , Liver Diseases/genetics , Liver Diseases/metabolism , Liver Diseases/prevention & control , Mice , Mice, Knockout , Pain/drug therapy , Pain/genetics , Pain/metabolism , Pain/pathology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/prevention & control , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Vascular Diseases/drug therapy , Vascular Diseases/genetics , Vascular Diseases/metabolism
9.
Complement Ther Med ; 52: 102481, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32951731

ABSTRACT

Sickle Cell Disease (SCD) is a chronic hemolytic disorder associated with frequent pain episodes, end organ damage and a shortened lifespan. Currently there exist no disease specific targeted therapies for the treatment of acute vaso-occlusive crisis (VOC) and management with analgesics and hydration is purely supportive. Improvement in understanding of disease pathophysiology has resulted in a great interest in disease modifying novel therapies and many are being evaluated in clinical trials. Here we report the results from the pre-specified mid-point analysis of the Phase 2 study of Intravenous Gamma Globulin (IVIG) for the treatment of acute VOC in patients with SCD and lessons learned.


Subject(s)
Anemia, Sickle Cell/drug therapy , Immunoglobulins, Intravenous/therapeutic use , Pain Management/methods , gamma-Globulins/therapeutic use , Adolescent , Adult , Anemia, Sickle Cell/complications , Child , Double-Blind Method , Female , Humans , Immunologic Factors/therapeutic use , Male , Young Adult
10.
Immunity ; 53(2): 417-428.e4, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32735844

ABSTRACT

Psychological stress has adverse effects on various human diseases, including those of the cardiovascular system. However, the mechanisms by which stress influences disease activity remain unclear. Here, using vaso-occlusive episodes (VOEs) of sickle cell disease as a vascular disease model, we show that stress promotes VOEs by eliciting a glucocorticoid hormonal response that augments gut permeability, leading to microbiota-dependent interleukin-17A (IL-17A) secretion from T helper 17 (Th17) cells of the lamina propria, followed by the expansion of the circulating pool of aged neutrophils that trigger VOEs. We identify segmented filamentous bacteria as the commensal essential for the stress-induced expansion of aged neutrophils that enhance VOEs in mice. Importantly, the inhibition of glucocorticoids synthesis, blockade of IL-17A, or depletion of the Th17 cell-inducing gut microbiota markedly reduces stress-induced VOEs. These results offer potential therapeutic targets to limit the impact of psychological stress on acute vascular occlusion.


Subject(s)
Anemia, Sickle Cell/pathology , Gastrointestinal Microbiome/immunology , Interleukin-17/metabolism , Stress, Psychological/pathology , Th17 Cells/immunology , Anemia, Sickle Cell/psychology , Animals , Bacteria/immunology , Cell Line , Germ-Free Life , Glucocorticoids/biosynthesis , Granulocyte Colony-Stimulating Factor/metabolism , HEK293 Cells , Humans , Hypothalamo-Hypophyseal System/metabolism , Inflammation/immunology , Inflammation/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology
11.
Dev Cell ; 53(5): 503-513.e5, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32413329

ABSTRACT

Bone marrow (BM) mesenchymal stem and progenitor cells (MSPCs) are a critical constituent of the hematopoietic stem cell (HSC) niche. Previous studies have suggested that the zinc-finger epithelial-mesenchymal transition transcription factor Snai2 (also known as Slug) regulated HSCs autonomously. Here, we show that Snai2 expression in the BM is restricted to the BM stromal compartment where it regulates the HSC niche. Germline or MSPC-selective Snai2 deletion reduces the functional MSPC pool and their mesenchymal lineage output and impairs HSC niche function during homeostasis and after stress. RNA sequencing analysis revealed that Spp1 (osteopontin) expression is markedly upregulated in Snai2-deficient MSPCs. Genetic deletion of Spp1 in Snai2-deficient mice rescues MSPCs' functions. Thus, SNAI2 is a critical regulator of the transcriptional network maintaining MSPCs by the suppression of osteopontin expression.


Subject(s)
Bone Marrow Cells/metabolism , Osteopontin/genetics , Snail Family Transcription Factors/metabolism , Stem Cell Niche , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Gene Deletion , Mice , Mice, Inbred C57BL , Osteopontin/metabolism , Snail Family Transcription Factors/genetics
12.
Nat Prod Res ; 34(2): 197-203, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30856343

ABSTRACT

Two new norneolignans, (7S,8R)-3-methoxy-3',4,9-trihydroxy-4',7-epoxy-8,3'-neolignane-1'-carboxylic acid (1) and (7R,8R)-3-methoxyl-4,9-dihydroxy-3':7,4':8-diepoxyneolignan-1'-carboxylic acid methyl ester (2) were isolated from Callicarpa kwangtungensis, together with ten known compounds, genistin (3), daidzin (4), silybin A (5), isosilybin A (6), isosilybin B (7), p-hydroxybenzaldehyde (8), syringic acid (9), lanceolatin A (10), icariside C5 (11), and (3S,6E,10R)-10-ß-D-glucopyranosyloxy-3,11-dihydroxy-3,7,11-trimethyldodeca-1,6-diene (12). Compounds 1 and 2 were evaluated for their effects on the inhibition of nitric oxide (NO) production in lipopolysaccharide induced RAW264.7 cells. Compounds 1 and 2 exhibited inhibitory activity with IC50 values of 31.45 ± 0.38 and 40.72 ± 0.54 µM, respectively.


Subject(s)
Callicarpa/chemistry , Lignans/isolation & purification , Nitric Oxide/antagonists & inhibitors , Animals , Inhibitory Concentration 50 , Lignans/analysis , Lignans/chemistry , Lignans/pharmacology , Lipopolysaccharides , Macrophages/metabolism , Mice , Molecular Structure , Nitric Oxide/biosynthesis , RAW 264.7 Cells
13.
Biochim Biophys Acta Mol Basis Dis ; 1865(10): 2657-2670, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31299217

ABSTRACT

Staphylococcus aureus is the most prominent cause of skin and soft tissue infections (SSTI) worldwide. Mortality associated with invasive SSTI is a major threat to public health considering the incidence of antibiotic resistant isolates in particular methicillin resistant S. aureus both in the hospital (HA-MRSA) and in the community (CA-MRSA). To overcome the increasing difficulties in the clinical management of SSTI due to MRSA, new prophylactic and therapeutic approaches are urgently needed and a preventive vaccine would be welcome. The rational design of an anti-S. aureus vaccine requires a deep knowledge of the role that the different bacterial virulence factors play according to the type of infection. In the present study, using a set of isogenic deficient mutants and their complemented strains we determined that the staphylococcal surface proteins SpA and Sbi play an important role in the induction of inflammatory cytokines and chemokines in the skin during SSTI. SpA and Sbi initiate signaling cascades that lead to the early recruitment of neutrophils, modulate their lifespan in the skin milieu and contribute to proper abscess formation and bacterial eradication. Moreover, the expression of SpA and Sbi appear critical for skin repair and wound healing. Thus, these results indicate that SpA and Sbi can promote immune responses in the skin that are beneficial for the host and therefore, should not be neutralized with vaccine formulations designed to prevent SSTI.


Subject(s)
Abscess/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Skin/immunology , Soft Tissue Infections/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , Wound Healing/physiology , Abscess/metabolism , Abscess/microbiology , Animals , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Humans , Keratinocytes , Methicillin-Resistant Staphylococcus aureus/metabolism , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin/microbiology , Skin/pathology , Soft Tissue Infections/immunology , Soft Tissue Infections/microbiology , Soft Tissue Infections/pathology , Staphylococcal Infections/immunology , Staphylococcus aureus/pathogenicity
15.
Nat Cell Biol ; 21(5): 560-567, 2019 05.
Article in English | MEDLINE | ID: mdl-30988422

ABSTRACT

Haematopoietic stem cells (HSCs) are maintained by bone marrow niches in vivo1,2, but the ability of niche cells to maintain HSCs ex vivo is markedly diminished. Expression of niche factors by Nestin-GFP+ mesenchymal-derived stromal cells (MSCs) is downregulated upon culture, suggesting that transcriptional rewiring may contribute to this reduced HSC maintenance potential. Using an RNA sequencing screen, we identified five genes encoding transcription factors (Klf7, Ostf1, Xbp1, Irf3 and Irf7) that restored HSC niche function in cultured bone marrow-derived MSCs. These revitalized MSCs (rMSCs) exhibited enhanced synthesis of HSC niche factors while retaining their mesenchymal differentiation capacity. In contrast to HSCs co-cultured with control MSCs, HSCs expanded with rMSCs showed higher repopulation capacity and protected lethally irradiated recipient mice. Competitive reconstitution assays revealed an approximately sevenfold expansion of functional HSCs by rMSCs. rMSCs prevented the accumulation of DNA damage in cultured HSCs, a hallmark of ageing and replication stress. Analysis of the reprogramming mechanisms uncovered a role for myocyte enhancer factor 2c (Mef2c) in the revitalization of MSCs. These results provide insight into the transcriptional regulation of the niche with implications for stem cell-based therapies.


Subject(s)
Cell Differentiation/genetics , Cell Engineering/methods , Hematopoietic Stem Cells/cytology , Stem Cell Niche/genetics , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/metabolism , Humans , Interferon Regulatory Factor-3/genetics , Intracellular Signaling Peptides and Proteins , Kruppel-Like Transcription Factors/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Transgenic , Nestin/genetics , Peptides/genetics , Sequence Analysis, RNA/methods , X-Box Binding Protein 1/genetics
16.
Nat Commun ; 9(1): 2449, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29934585

ABSTRACT

Endothelial cells (ECs) contribute to haematopoietic stem cell (HSC) maintenance in bone marrow, but the differential contributions of EC subtypes remain unknown, owing to the lack of methods to separate with high purity arterial endothelial cells (AECs) from sinusoidal endothelial cells (SECs). Here we show that the combination of podoplanin (PDPN) and Sca-1 expression distinguishes AECs (CD45- Ter119- Sca-1bright PDPN-) from SECs (CD45- Ter119- Sca-1dim PDPN+). PDPN can be substituted for antibodies against the adhesion molecules ICAM1 or E-selectin. Unexpectedly, prospective isolation reveals that AECs secrete nearly all detectable EC-derived stem cell factors (SCF). Genetic deletion of Scf in AECs, but not SECs, significantly reduced functional HSCs. Lineage-tracing analyses suggest that AECs and SECs self-regenerate independently after severe genotoxic insults, indicating the persistence of, and recovery from, radio-resistant pre-specified EC precursors. AEC-derived SCF also promotes HSC recovery after myeloablation. These results thus uncover heterogeneity in the contribution of ECs in stem cell niches.


Subject(s)
Bone Marrow Cells/metabolism , Endothelial Cells/metabolism , Stem Cell Factor/metabolism , Animals , Antigens, Ly/metabolism , Arteries/cytology , Bone Marrow/blood supply , Bone Marrow Transplantation , Capillaries/cytology , Cell Differentiation/physiology , Cells, Cultured , Female , Hematopoietic Stem Cells/physiology , Male , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Stem Cell Factor/genetics , Stem Cell Niche/physiology , Transplantation Chimera
17.
Development ; 145(2)2018 01 22.
Article in English | MEDLINE | ID: mdl-29358215

ABSTRACT

Hematopoietic stem cells (HSCs) develop in discrete anatomical niches, migrating during embryogenesis from the aorta-gonad-mesonephros (AGM) region to the fetal liver, and finally to the bone marrow, where most HSCs reside throughout adult life. These niches provide supportive microenvironments that specify, expand and maintain HSCs. Understanding the constituents and molecular regulation of HSC niches is of considerable importance as it could shed new light on the mechanistic principles of HSC emergence and maintenance, and provide novel strategies for regenerative medicine. However, controversy exists concerning the cellular complexity of the bone marrow niche, and our understanding of the different HSC niches during development remains limited. In this Review, we summarize and discuss what is known about the heterogeneity of the HSC niches at distinct stages of their ontogeny, from the embryo to the adult bone marrow, drawing predominantly on data from mouse studies.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Stem Cell Niche/physiology , Aging/pathology , Aging/physiology , Animals , Aorta/embryology , Cell Lineage , Female , Gonads/embryology , Hematologic Neoplasms/pathology , Hematopoietic System/embryology , Humans , Male , Mesonephros/embryology , Mice , Placenta/cytology , Placenta/physiology , Pregnancy , Stromal Cells/cytology , Stromal Cells/physiology , Sympathetic Nervous System/embryology , Sympathetic Nervous System/physiology
18.
Mol Ther Methods Clin Dev ; 4: 27-38, 2017 Mar 17.
Article in English | MEDLINE | ID: mdl-28344989

ABSTRACT

Antigen-specific immunotherapy of type 1 diabetes, typically via delivery of a single native ß cell antigen, has had little clinical benefit to date. With increasing evidence that diabetogenic T cells react against multiple ß cell antigens, including previously unappreciated neo-antigens that can be emulated by mimotopes, a shift from protein- to epitope-based therapy is warranted. To this end, we aimed to achieve efficient co-presentation of multiple major epitopes targeting both CD4+ and CD8+ diabetogenic T cells. We have compared native epitopes versus mimotopes as well as various targeting signals in an effort to optimize recognition by both types of T cells in vitro. Optimal engagement of all T cells was achieved with segregation of CD8 and CD4 epitopes, the latter containing mimotopes and driven by endosome-targeting signals, after delivery into either dendritic or stromal cells. The CD4+ T cell responses elicited by the endogenously delivered epitopes were comparable with high concentrations of soluble peptide and included functional regulatory T cells. This work has important implications for the improvement of antigen-specific therapies using an epitope-based approach to restore tolerance in type 1 diabetes and in a variety of other diseases requiring concomitant targeting of CD4+ and CD8+ T cells.

20.
Cell Rep ; 16(3): 769-80, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27396328

ABSTRACT

Osteoblasts and adipocytes are derived from a common precursor, mesenchymal stem cells (MSCs). Alterations in the normal fate of differentiating MSCs are involved in the development of obesity and osteoporosis. Here, we report that viable motheaten (me(v)) mice, which are deficient in the SH2-domain-containing phosphatase-1 (SHP1), develop osteoporosis spontaneously. Consistently, MSCs from me(v)/me(v) mice exhibit significantly reduced osteogenic potential and greatly increased adipogenic potential. When MSCs were transplanted into nude mice, SHP1-deficient MSCs resulted in diminished bone formation compared with wild-type MSCs. SHP1 was found to bind to GSK3ß and suppress its kinase activity by dephosphorylating pY216, thus resulting in ß-catenin stabilization. Mice, in which SHP1 was deleted in MSCs using SHP1(fl/fl)Dermo1-cre, displayed significantly decreased bone mass and increased adipose tissue. Taken together, these results suggest a possible role for SHP1 in controlling tissue homeostasis through modulation of MSC differentiation via Wnt signaling regulation.


Subject(s)
Bone Density/physiology , Cell Differentiation/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Adipocytes/metabolism , Adipogenesis/physiology , Animals , Cells, Cultured , Female , Glycogen Synthase Kinase 3 beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Osteoblasts/metabolism , Osteoblasts/physiology , Osteogenesis/physiology , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...