Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nat Aging ; 4(5): 727-744, 2024 May.
Article in English | MEDLINE | ID: mdl-38622407

ABSTRACT

Skeletal muscle aging is a key contributor to age-related frailty and sarcopenia with substantial implications for global health. Here we profiled 90,902 single cells and 92,259 single nuclei from 17 donors to map the aging process in the adult human intercostal muscle, identifying cellular changes in each muscle compartment. We found that distinct subsets of muscle stem cells exhibit decreased ribosome biogenesis genes and increased CCL2 expression, causing different aging phenotypes. Our atlas also highlights an expansion of nuclei associated with the neuromuscular junction, which may reflect re-innervation, and outlines how the loss of fast-twitch myofibers is mitigated through regeneration and upregulation of fast-type markers in slow-twitch myofibers with age. Furthermore, we document the function of aging muscle microenvironment in immune cell attraction. Overall, we present a comprehensive human skeletal muscle aging resource ( https://www.muscleageingcellatlas.org/ ) together with an in-house mouse muscle atlas to study common features of muscle aging across species.


Subject(s)
Aging , Muscle, Skeletal , Humans , Aging/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Animals , Mice , Adult , Aged , Sarcopenia/pathology , Sarcopenia/metabolism , Male , Neuromuscular Junction/metabolism , Middle Aged , Female
2.
Kidney Int ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38431215

ABSTRACT

Despite the recent advances in our understanding of the role of lipids, metabolites, and related enzymes in mediating kidney injury, there is limited integrated multi-omics data identifying potential metabolic pathways driving impaired kidney function. The limited availability of kidney biopsies from living donors with acute kidney injury has remained a major constraint. Here, we validated the use of deceased transplant donor kidneys as a good model to study acute kidney injury in humans and characterized these kidneys using imaging and multi-omics approaches. We noted consistent changes in kidney injury and inflammatory markers in donors with reduced kidney function. Neighborhood and correlation analyses of imaging mass cytometry data showed that subsets of kidney cells (proximal tubular cells and fibroblasts) are associated with the expression profile of kidney immune cells, potentially linking these cells to kidney inflammation. Integrated transcriptomic and metabolomic analysis of human kidneys showed that kidney arachidonic acid metabolism and seven other metabolic pathways were upregulated following diminished kidney function. To validate the arachidonic acid pathway in impaired kidney function we demonstrated increased levels of cytosolic phospholipase A2 protein and related lipid mediators (prostaglandin E2) in the injured kidneys. Further, inhibition of cytosolic phospholipase A2 reduced injury and inflammation in human kidney proximal tubular epithelial cells in vitro. Thus, our study identified cell types and metabolic pathways that may be critical for controlling inflammation associated with impaired kidney function in humans.

3.
Cell Rep Methods ; 3(10): 100598, 2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37776856

ABSTRACT

Spatially resolved omics technologies reveal context-dependent cellular regulatory networks in tissues of interest. Beyond transcriptome analysis, information on epigenetic traits and chromatin accessibility can provide further insights on gene regulation in health and disease. Nevertheless, compared to the enormous advancements in spatial transcriptomics technologies, the field of spatial epigenomics is much younger and still underexplored. In this study, we report laser capture microdissection coupled to ATAC-seq (LCM-ATAC-seq) applied to fresh frozen samples for the spatial characterization of chromatin accessibility. We first demonstrate the efficient use of LCM coupled to in situ tagmentation and evaluate its performance as a function of cell number, microdissected areas, and tissue type. Further, we demonstrate its use for the targeted chromatin accessibility analysis of discrete contiguous or scattered cell populations in tissues via single-nuclei capture based on immunostaining for specific cellular markers.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Chromatin , Chromatin/genetics , Laser Capture Microdissection , Gene Expression Profiling , Freezing
4.
Nature ; 619(7971): 801-810, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37438528

ABSTRACT

The function of a cell is defined by its intrinsic characteristics and its niche: the tissue microenvironment in which it dwells. Here we combine single-cell and spatial transcriptomics data to discover cellular niches within eight regions of the human heart. We map cells to microanatomical locations and integrate knowledge-based and unsupervised structural annotations. We also profile the cells of the human cardiac conduction system1. The results revealed their distinctive repertoire of ion channels, G-protein-coupled receptors (GPCRs) and regulatory networks, and implicated FOXP2 in the pacemaker phenotype. We show that the sinoatrial node is compartmentalized, with a core of pacemaker cells, fibroblasts and glial cells supporting glutamatergic signalling. Using a custom CellPhoneDB.org module, we identify trans-synaptic pacemaker cell interactions with glia. We introduce a druggable target prediction tool, drug2cell, which leverages single-cell profiles and drug-target interactions to provide mechanistic insights into the chronotropic effects of drugs, including GLP-1 analogues. In the epicardium, we show enrichment of both IgG+ and IgA+ plasma cells forming immune niches that may contribute to infection defence. Overall, we provide new clarity to cardiac electro-anatomy and immunology, and our suite of computational approaches can be applied to other tissues and organs.


Subject(s)
Cellular Microenvironment , Heart , Multiomics , Myocardium , Humans , Cell Communication , Fibroblasts/cytology , Glutamic Acid/metabolism , Heart/anatomy & histology , Heart/innervation , Ion Channels/metabolism , Myocardium/cytology , Myocardium/immunology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Neuroglia/cytology , Pericardium/cytology , Pericardium/immunology , Plasma Cells/immunology , Receptors, G-Protein-Coupled/metabolism , Sinoatrial Node/anatomy & histology , Sinoatrial Node/cytology , Sinoatrial Node/physiology , Heart Conduction System/anatomy & histology , Heart Conduction System/cytology , Heart Conduction System/metabolism
5.
Blood ; 141(19): 2343-2358, 2023 05 11.
Article in English | MEDLINE | ID: mdl-36758207

ABSTRACT

Classic Hodgkin lymphoma (cHL) has a rich immune infiltrate, which is an intrinsic component of the neoplastic process. Malignant Hodgkin Reed-Sternberg cells (HRSCs) create an immunosuppressive microenvironment by the expression of regulatory molecules, preventing T-cell activation. It has also been demonstrated that mononuclear phagocytes (MNPs) in the vicinity of HRSCs express similar regulatory mechanisms in parallel, and their presence in tissue is associated with inferior patient outcomes. MNPs in cHL have hitherto been identified by a small number of canonical markers and are usually described as tumor-associated macrophages. The organization of MNP networks and interactions with HRSCs remains unexplored at high resolution. Here, we defined the global immune-cell composition of cHL and nonlymphoma lymph nodes, integrating data across single-cell RNA sequencing, spatial transcriptomics, and multiplexed immunofluorescence. We observed that MNPs comprise multiple subsets of monocytes, macrophages, and dendritic cells (DCs). Classical monocytes, macrophages and conventional DC2s were enriched in the vicinity of HRSCs, but plasmacytoid DCs and activated DCs were excluded. Unexpectedly, cDCs and monocytes expressed immunoregulatory checkpoints PD-L1, TIM-3, and the tryptophan-catabolizing protein IDO, at the same level as macrophages. Expression of these molecules increased with age. We also found that classical monocytes are important signaling hubs, potentially controlling the retention of cDC2 and ThExh via CCR1-, CCR4-, CCR5-, and CXCR3-dependent signaling. Enrichment of the cDC2-monocyte-macrophage network in diagnostic biopsies is associated with early treatment failure. These results reveal unanticipated complexity and spatial polarization within the MNP compartment, further demonstrating their potential roles in immune evasion by cHL.


Subject(s)
Hodgkin Disease , Humans , Hodgkin Disease/diagnosis , Reed-Sternberg Cells/metabolism , Macrophages/metabolism , Monocytes/metabolism , Immunosuppressive Agents , Tumor Microenvironment
7.
Nat Genet ; 55(1): 66-77, 2023 01.
Article in English | MEDLINE | ID: mdl-36543915

ABSTRACT

Single-cell transcriptomics has allowed unprecedented resolution of cell types/states in the human lung, but their spatial context is less well defined. To (re)define tissue architecture of lung and airways, we profiled five proximal-to-distal locations of healthy human lungs in depth using multi-omic single cell/nuclei and spatial transcriptomics (queryable at lungcellatlas.org ). Using computational data integration and analysis, we extend beyond the suspension cell paradigm and discover macro and micro-anatomical tissue compartments including previously unannotated cell types in the epithelial, vascular, stromal and nerve bundle micro-environments. We identify and implicate peribronchial fibroblasts in lung disease. Importantly, we discover and validate a survival niche for IgA plasma cells in the airway submucosal glands (SMG). We show that gland epithelial cells recruit B cells and IgA plasma cells, and promote longevity and antibody secretion locally through expression of CCL28, APRIL and IL-6. This new 'gland-associated immune niche' has implications for respiratory health.


Subject(s)
Lung , Respiratory Mucosa , Humans , Respiratory Mucosa/metabolism , Epithelial Cells/metabolism , B-Lymphocytes , Immunoglobulin A/metabolism
8.
Nat Cell Biol ; 24(10): 1487-1498, 2022 10.
Article in English | MEDLINE | ID: mdl-36109670

ABSTRACT

The liver has been studied extensively due to the broad number of diseases affecting its vital functions. However, therapeutic advances have been hampered by the lack of knowledge concerning human hepatic development. Here, we addressed this limitation by describing the developmental trajectories of different cell types that make up the human liver at single-cell resolution. These transcriptomic analyses revealed that sequential cell-to-cell interactions direct functional maturation of hepatocytes, with non-parenchymal cells playing essential roles during organogenesis. We utilized this information to derive bipotential hepatoblast organoids and then exploited this model system to validate the importance of signalling pathways in hepatocyte and cholangiocyte specification. Further insights into hepatic maturation also enabled the identification of stage-specific transcription factors to improve the functionality of hepatocyte-like cells generated from human pluripotent stem cells. Thus, our study establishes a platform to investigate the basic mechanisms directing human liver development and to produce cell types for clinical applications.


Subject(s)
Hepatocytes , Liver , Humans , Liver/metabolism , Hepatocytes/metabolism , Cell Differentiation , Organoids , Transcription Factors/genetics , Transcription Factors/metabolism
9.
iScience ; 25(7): 104660, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35845169

ABSTRACT

Bladder infection affects a hundred million people annually, but our understanding of bladder immunity is incomplete. We found type 17 immune response genes among the most up-regulated networks in mouse bladder following uropathogenic Escherichia coli (UPEC) challenge. Intravital imaging revealed submucosal Rorc+ cells responsive to UPEC challenge, and we found increased Il17 and IL22 transcripts in wild-type and Rag2 -/- mice, implicating group 3 innate lymphoid cells (ILC3s) as a source of these cytokines. NCR-positive and negative ILC3 subsets were identified in murine and human bladders, with local proliferation increasing IL17-producing ILC3s post infection. ILC3s made a more limited contribution to bladder IL22, with prominent early induction of IL22 evident in Th17 cells. Single-cell RNA sequencing revealed bladder NCR-negative ILC3s as the source of IL17 and identified putative ILC3-myeloid cell interactions, including via lymphotoxin-ß-LTBR. Altogether, our data provide important insights into the orchestration and execution of type 17 immunity in bladder defense.

10.
Sci Immunol ; 7(69): eabm9060, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35302862

ABSTRACT

B cells generate antibodies that are essential for immune protection, but their subgroups are poorly defined. Here, we perform undirected deep profiling of B cells in matched human lymphoid tissues from deceased transplant organ donors and blood. In addition to identifying unanticipated features of tissue-based B cell differentiation, we resolve two subsets of marginal zone B (MZB) cells differing in cell surface and transcriptomic profiles, clonal relationships to other subsets, enrichment of genes in the NOTCH pathway, distribution bias within splenic marginal zone microenvironment, and immunoglobulin repertoire diversity and hypermutation frequency. Each subset is present in spleen, gut-associated lymphoid tissue, mesenteric lymph nodes, and blood. MZB cells and the lineage from which they are derived are depleted in lupus nephritis. Here, we show that this depletion is of only one MZB subset. The other remains unchanged as a proportion of total B cells compared with health. Thus, it is important to factor MZB cell heterogeneity into studies of human B cell responses and pathology.


Subject(s)
B-Lymphocytes , Lymphoid Tissue , Humans , Lymphocyte Activation , Lymphocyte Count , Spleen
11.
Injury ; 53(4): 1532-1538, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35168759

ABSTRACT

BACKGROUND: Tibial shaft fractures are the commonest long bone fracture, with early weight-bearing improving the rate of bony union. However, an intact fibula can act as a strut that splints the tibial segments and holds them apart. A fibular osteotomy, in which a 2.5 cm length of fibula is removed, has been used to treat delayed and hypertrophic non-union by increasing axial tibial loading. However, there is no consensus on the optimal site for the partial fibulectomy. METHODS: Nine leg specimens were obtained from formalin-embalmed cadavers. Transverse mid-shaft tibial fractures were created using an oscillating saw. A rig was designed to compress the legs with an adjustable axial load and measure the force within the fracture site in order to ascertain load transmission through the tibia over a range of weights. After 2.5cm-long fibulectomies were performed at one of three levels on each specimen, load transmission through the tibia was re-assessed. A beam structure model of the intact leg was designed to explain the findings. RESULTS: With an intact fibula, mean tibial loading at 34 kg was 15.52 ± 3.26 kg, increasing to 17.42 ± 4.13 kg after fibular osteotomy. This increase was only significant where the osteotomy was performed proximal to or at the level of the tibial fracture. Modelling midshaft tibial loading using the Euler-Bernoulli beam theory showed that 80.5% of the original force was transmitted through the tibia with an intact fibula, rising to 81.1% after a distal fibulectomy, and 100% with a proximal fibulectomy. CONCLUSION: This study describes a novel method of measuring axial tibial forces. We demonstrated that a fibular osteotomy increases axial tibial loading regardless of location, with the greatest increase after proximal fibular osteotomy. A contributing factor for this can be explained by a simple beam model. We therefore recommend a proximal fibular osteotomy when it is performed in the treatment of delayed and non-union of tibial midshaft fractures.


Subject(s)
Fibula , Tibial Fractures , Diaphyses , Fibula/surgery , Humans , Osteotomy , Tibia/surgery , Tibial Fractures/surgery
12.
Blood ; 139(23): 3387-3401, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35073399

ABSTRACT

Rare hematopoietic stem and progenitor cell (HSPC) pools outside the bone marrow (BM) contribute to blood production in stress and disease but remain ill-defined. Although nonmobilized peripheral blood (PB) is routinely sampled for clinical management, the diagnosis and monitoring potential of PB HSPCs remain untapped, as no healthy PB HSPC baseline has been reported. Here we comprehensively delineate human extramedullary HSPC compartments comparing spleen, PB, and mobilized PB to BM using single-cell RNA-sequencing and/or functional assays. We uncovered HSPC features shared by extramedullary tissues and others unique to PB. First, in contrast to actively dividing BM HSPCs, we found no evidence of substantial ongoing hematopoiesis in extramedullary tissues at steady state but report increased splenic HSPC proliferative output during stress erythropoiesis. Second, extramedullary hematopoietic stem cells/multipotent progenitors (HSCs/MPPs) from spleen, PB, and mobilized PB share a common transcriptional signature and increased abundance of lineage-primed subsets compared with BM. Third, healthy PB HSPCs display a unique bias toward erythroid-megakaryocytic differentiation. At the HSC/MPP level, this is functionally imparted by a subset of phenotypic CD71+ HSCs/MPPs, exclusively producing erythrocytes and megakaryocytes, highly abundant in PB but rare in other adult tissues. Finally, the unique erythroid-megakaryocytic-skewing of PB is perturbed with age in essential thrombocythemia and ß-thalassemia. Collectively, we identify extramedullary lineage-primed HSPC reservoirs that are nonproliferative in situ and report involvement of splenic HSPCs during demand-adapted hematopoiesis. Our data also establish aberrant composition and function of circulating HSPCs as potential clinical indicators of BM dysfunction.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells , Adult , Bone Marrow , Bone Marrow Cells/physiology , Erythropoiesis , Humans , Megakaryocytes
13.
J Pediatr Surg ; 57(2): 245-249, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34952694

ABSTRACT

BACKGROUND: Intra-abdominal collections (IAC) are a common complication following appendicectomy, one of the most commonly performed emergency abdominal procedures in childhood. The option to drain a collection is frequently available but not always required. AIM: The aim of this study was to compare the outcomes of medically and procedurally-managed post appendicectomy IACs and suggest a method of standardising the need for intervention. METHODS: A single centre, retrospective review of children aged ≤ 16 years presenting between 2014 and 2019 was performed. Patient demographics, management, and outcome data were collected. IAC volume and surface area were calculated assuming a prolate spheroid or true ellipsoid depending on the number of dimensions reported. RESULTS: 60 patients (18%) of 334 patients developed an IAC post appendicectomy. Medical management was undertaken in 44 (73%), drainage in 12 (20%), and surgical washout in 4 (7%). Collection size was associated with failure of medical management: maximum diameter (p = 0.028), volume (p = 0.002), and surface area (p = 0.001). Collections with a volume of 2 ml/kg were significantly less likely to fail medical management than larger collections (0/33 vs 6/11; p < 0.0001). DISCUSSION: Not all post appendicectomy IACs require drainage. The relationship between collection volume and need for drainage is more closely assessed using a volume calculation rather than a single dimension measurement, particularly when adjusted for weight of the child. A cut off of 2 ml/kg appears to be a good objective measure for intervention and provides a communication tool for discussion amongst the multidisciplinary team. Prospectively collected multicentre data on this subject would be timely. LEVEL OF EVIDENCE: III.


Subject(s)
Appendicitis , Abdominal Muscles , Adolescent , Appendectomy , Appendicitis/surgery , Child , Drainage , Humans , Retrospective Studies
14.
Nat Genet ; 53(12): 1698-1711, 2021 12.
Article in English | MEDLINE | ID: mdl-34857954

ABSTRACT

The endometrium, the mucosal lining of the uterus, undergoes dynamic changes throughout the menstrual cycle in response to ovarian hormones. We have generated dense single-cell and spatial reference maps of the human uterus and three-dimensional endometrial organoid cultures. We dissect the signaling pathways that determine cell fate of the epithelial lineages in the lumenal and glandular microenvironments. Our benchmark of the endometrial organoids reveals the pathways and cell states regulating differentiation of the secretory and ciliated lineages both in vivo and in vitro. In vitro downregulation of WNT or NOTCH pathways increases the differentiation efficiency along the secretory and ciliated lineages, respectively. We utilize our cellular maps to deconvolute bulk data from endometrial cancers and endometriotic lesions, illuminating the cell types dominating in each of these disorders. These mechanistic insights provide a platform for future development of treatments for common conditions including endometriosis and endometrial carcinoma.


Subject(s)
Endometrium/physiology , Menstrual Cycle , Cell Differentiation , Cell Lineage , Cellular Microenvironment , Endometrial Neoplasms/pathology , Endometrium/embryology , Endometrium/pathology , Female , Gonadal Steroid Hormones/metabolism , Humans , In Vitro Techniques , Organoids , Receptors, Notch/metabolism , Signal Transduction , Spatio-Temporal Analysis , Tissue Culture Techniques , Transcriptome , Uterus/pathology , Wnt Proteins/metabolism
15.
Commun Biol ; 4(1): 1186, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34650224

ABSTRACT

The adoptive transfer of regulatory T-cells (Tregs) is a promising therapeutic approach in transplantation and autoimmunity. However, because large cell numbers are needed to achieve a therapeutic effect, in vitro expansion is required. By comparing their function, phenotype and transcriptomic profile against ex vivo Tregs, we demonstrate that expanded human Tregs switch their metabolism to aerobic glycolysis and show enhanced suppressive function through hypoxia-inducible factor 1-alpha (HIF1A) driven acquisition of CD73 expression. In conjunction with CD39, CD73 expression enables expanded Tregs to convert ATP to immunosuppressive adenosine. We conclude that for maximum therapeutic benefit, Treg expansion protocols should be optimised for CD39/CD73 co-expression.


Subject(s)
5'-Nucleotidase/genetics , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , T-Lymphocytes, Regulatory/metabolism , 5'-Nucleotidase/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male
16.
Nature ; 597(7875): 250-255, 2021 09.
Article in English | MEDLINE | ID: mdl-34497389

ABSTRACT

The cellular landscape of the human intestinal tract is dynamic throughout life, developing in utero and changing in response to functional requirements and environmental exposures. Here, to comprehensively map cell lineages, we use single-cell RNA sequencing and antigen receptor analysis of almost half a million cells from up to 5 anatomical regions in the developing and up to 11 distinct anatomical regions in the healthy paediatric and adult human gut. This reveals the existence of transcriptionally distinct BEST4 epithelial cells throughout the human intestinal tract. Furthermore, we implicate IgG sensing as a function of intestinal tuft cells. We describe neural cell populations in the developing enteric nervous system, and predict cell-type-specific expression of genes associated with Hirschsprung's disease. Finally, using a systems approach, we identify key cell players that drive the formation of secondary lymphoid tissue in early human development. We show that these programs are adopted in inflammatory bowel disease to recruit and retain immune cells at the site of inflammation. This catalogue of intestinal cells will provide new insights into cellular programs in development, homeostasis and disease.


Subject(s)
Aging , Enteric Nervous System/cytology , Fetus/cytology , Health , Intestines/cytology , Intestines/growth & development , Lymph Nodes/cytology , Lymph Nodes/growth & development , Adult , Animals , Child , Crohn Disease/pathology , Datasets as Topic , Enteric Nervous System/anatomy & histology , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Epithelial Cells/cytology , Female , Fetus/anatomy & histology , Fetus/embryology , Humans , Intestines/embryology , Intestines/innervation , Lymph Nodes/embryology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Organogenesis , Receptors, IgG/metabolism , Signal Transduction , Spatio-Temporal Analysis , Time Factors
17.
Science ; 373(6556): 760-767, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34385390

ABSTRACT

The origin of human metaplastic states and their propensity for cancer is poorly understood. Barrett's esophagus is a common metaplastic condition that increases the risk for esophageal adenocarcinoma, and its cellular origin is enigmatic. To address this, we harvested tissues spanning the gastroesophageal junction from healthy and diseased donors, including isolation of esophageal submucosal glands. A combination of single-cell transcriptomic profiling, in silico lineage tracing from methylation, open chromatin and somatic mutation analyses, and functional studies in organoid models showed that Barrett's esophagus originates from gastric cardia through c-MYC and HNF4A-driven transcriptional programs. Furthermore, our data indicate that esophageal adenocarcinoma likely arises from undifferentiated Barrett's esophagus cell types even in the absence of a pathologically identifiable metaplastic precursor, illuminating early detection strategies.


Subject(s)
Adenocarcinoma/pathology , Barrett Esophagus/pathology , Cardia/cytology , Esophageal Neoplasms/pathology , Esophagus/pathology , Barrett Esophagus/genetics , Barrett Esophagus/metabolism , Cardia/chemistry , Cell Differentiation , Cell Lineage , Cell Transformation, Neoplastic , Epigenesis, Genetic , Epithelial Cells/cytology , Epithelial Cells/metabolism , Esophagus/cytology , Esophagus/metabolism , Exocrine Glands/chemistry , Exocrine Glands/cytology , Hepatocyte Nuclear Factor 4/metabolism , Humans , Keratin-7/analysis , Metaplasia , Phenotype , Proto-Oncogene Proteins c-myc/metabolism , RNA-Seq , Single-Cell Analysis , Transcription, Genetic , Transcriptome
18.
Sci Rep ; 11(1): 10418, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001961

ABSTRACT

Cryopreservation offers the potential to increase the availability of pancreatic islets for treatment of diabetic patients. However, current protocols, which use dimethyl sulfoxide (DMSO), lead to poor cryosurvival of islets. We demonstrate that equilibration of mouse islets with small molecules in aqueous solutions can be accelerated from > 24 to 6 h by increasing incubation temperature to 37 °C. We utilize this finding to demonstrate that current viability staining protocols are inaccurate and to develop a novel cryopreservation method combining DMSO with trehalose pre-incubation to achieve improved cryosurvival. This protocol resulted in improved ATP/ADP ratios and peptide secretion from ß-cells, preserved cAMP response, and a gene expression profile consistent with improved cryoprotection. Our findings have potential to increase the availability of islets for transplantation and to inform the design of cryopreservation protocols for other multicellular aggregates, including organoids and bioengineered tissues.


Subject(s)
Cryopreservation/methods , Cryoprotective Agents/pharmacokinetics , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation/methods , Islets of Langerhans , Animals , Cell Survival , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/chemically induced , Humans , Male , Mice , Models, Animal , Primary Cell Culture , Streptozocin/administration & dosage , Streptozocin/toxicity
19.
Clin Transl Immunology ; 9(11): e1202, 2020.
Article in English | MEDLINE | ID: mdl-33173582

ABSTRACT

OBJECTIVES: Humanised mice have emerged as valuable models for pre-clinical testing of the safety and efficacy of immunotherapies. Given the variety of models available, selection of the most appropriate humanised mouse model is critical in study design. Here, we aimed to develop a model for predicting cytokine release syndrome (CRS) while minimising graft-versus-host disease (GvHD). METHODS: To overcome donor-induced variation, we directly compared the in vitro and in vivo immune phenotype of immunodeficient NSG mice reconstituted with human bone marrow (BM) CD34+ haematopoietic stem cells (HSCs), peripheral blood mononuclear cells (PBMCs) or spleen mononuclear cells (SPMCs) from the same human donors. SPMC engraftment in NSG-dKO mice, which lack MHC class I and II, was also evaluated as a strategy to limit GvHD. Another group of mice was engrafted with umbilical cord blood (UCB) CD34+ HSCs. Induction of CRS in vivo was investigated upon administration of the anti-CD3 monoclonal antibody OKT3. RESULTS: PBMC- and SPMC-reconstituted NSG mice showed short-term survival, with engrafted human T cells exhibiting mostly an effector memory phenotype. Survival in SPMC-reconstituted NSG-dKO mice was significantly longer. Conversely, both BM and UCB-HSC models showed longer survival, without demonstrable GvHD and a more naïve T-cell phenotype. PBMC- and SPMC-reconstituted mice, but not BM-HSC or UCB-HSC mice, experienced severe clinical signs of CRS upon administration of OKT3. CONCLUSION: PBMC- and SPMC-reconstituted NSG mice better predict OKT3-mediated CRS. The SPMC model allows generation of large experimental groups, and the use of NSG-dKO mice mitigates the limitation of early GvHD.

20.
Cell Stem Cell ; 27(6): 905-919.e10, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33142113

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is the cause of a present pandemic, infects human lung alveolar type 2 (hAT2) cells. Characterizing pathogenesis is crucial for developing vaccines and therapeutics. However, the lack of models mirroring the cellular physiology and pathology of hAT2 cells limits the study. Here, we develop a feeder-free, long-term, three-dimensional (3D) culture technique for hAT2 cells derived from primary human lung tissue and investigate infection response to SARS-CoV-2. By imaging-based analysis and single-cell transcriptome profiling, we reveal rapid viral replication and the increased expression of interferon-associated genes and proinflammatory genes in infected hAT2 cells, indicating a robust endogenous innate immune response. Further tracing of viral mutations acquired during transmission identifies full infection of individual cells effectively from a single viral entry. Our study provides deep insights into the pathogenesis of SARS-CoV-2 and the application of defined 3D hAT2 cultures as models for respiratory diseases.


Subject(s)
COVID-19 , Pulmonary Alveoli/virology , SARS-CoV-2/physiology , Stem Cells/virology , COVID-19/virology , Cell Culture Techniques , Culture Media , Humans , Interferons/metabolism , Models, Biological , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/ultrastructure , SARS-CoV-2/ultrastructure , Transcriptome , Virus Internalization , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...