Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nature ; 608(7924): 766-777, 2022 08.
Article in English | MEDLINE | ID: mdl-35948637

ABSTRACT

Myocardial infarction is a leading cause of death worldwide1. Although advances have been made in acute treatment, an incomplete understanding of remodelling processes has limited the effectiveness of therapies to reduce late-stage mortality2. Here we generate an integrative high-resolution map of human cardiac remodelling after myocardial infarction using single-cell gene expression, chromatin accessibility and spatial transcriptomic profiling of multiple physiological zones at distinct time points in myocardium from patients with myocardial infarction and controls. Multi-modal data integration enabled us to evaluate cardiac cell-type compositions at increased resolution, yielding insights into changes of the cardiac transcriptome and epigenome through the identification of distinct tissue structures of injury, repair and remodelling. We identified and validated disease-specific cardiac cell states of major cell types and analysed them in their spatial context, evaluating their dependency on other cell types. Our data elucidate the molecular principles of human myocardial tissue organization, recapitulating a gradual cardiomyocyte and myeloid continuum following ischaemic injury. In sum, our study provides an integrative molecular map of human myocardial infarction, represents an essential reference for the field and paves the way for advanced mechanistic and therapeutic studies of cardiac disease.


Subject(s)
Atrial Remodeling , Chromatin Assembly and Disassembly , Gene Expression Profiling , Myocardial Infarction , Single-Cell Analysis , Ventricular Remodeling , Atrial Remodeling/genetics , Case-Control Studies , Chromatin/genetics , Epigenome , Humans , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Time Factors , Ventricular Remodeling/genetics
2.
Nature ; 589(7841): 281-286, 2021 01.
Article in English | MEDLINE | ID: mdl-33176333

ABSTRACT

Kidney fibrosis is the hallmark of chronic kidney disease progression; however, at present no antifibrotic therapies exist1-3. The origin, functional heterogeneity and regulation of scar-forming cells that occur during human kidney fibrosis remain poorly understood1,2,4. Here, using single-cell RNA sequencing, we profiled the transcriptomes of cells from the proximal and non-proximal tubules of healthy and fibrotic human kidneys to map the entire human kidney. This analysis enabled us to map all matrix-producing cells at high resolution, and to identify distinct subpopulations of pericytes and fibroblasts as the main cellular sources of scar-forming myofibroblasts during human kidney fibrosis. We used genetic fate-tracing, time-course single-cell RNA sequencing and ATAC-seq (assay for transposase-accessible chromatin using sequencing) experiments in mice, and spatial transcriptomics in human kidney fibrosis, to shed light on the cellular origins and differentiation of human kidney myofibroblasts and their precursors at high resolution. Finally, we used this strategy to detect potential therapeutic targets, and identified NKD2 as a myofibroblast-specific target in human kidney fibrosis.


Subject(s)
Cell Lineage , Fibrosis/pathology , Kidney Tubules/pathology , Myofibroblasts/pathology , Renal Insufficiency, Chronic/pathology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/metabolism , Case-Control Studies , Cell Differentiation , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Male , Mesoderm/cytology , Mesoderm/pathology , Mice , Myofibroblasts/metabolism , Pericytes/cytology , Pericytes/pathology , RNA-Seq , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Single-Cell Analysis , Transcriptome
3.
Blood ; 136(18): 2051-2064, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32726410

ABSTRACT

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) that leads to progressive bone marrow (BM) fibrosis. Although the cellular mutations involved in the pathogenesis of PMF have been extensively investigated, the sequential events that drive stromal activation and fibrosis by hematopoietic-stromal cross-talk remain elusive. Using an unbiased approach and validation in patients with MPN, we determined that the differential spatial expression of the chemokine CXCL4/platelet factor-4 marks the progression of fibrosis. We show that the absence of hematopoietic CXCL4 ameliorates the MPN phenotype, reduces stromal cell activation and BM fibrosis, and decreases the activation of profibrotic pathways in megakaryocytes, inflammation in fibrosis-driving cells, and JAK/STAT activation in both megakaryocytes and stromal cells in 3 murine PMF models. Our data indicate that higher CXCL4 expression in MPN has profibrotic effects and is a mediator of the characteristic inflammation. Therefore, targeting CXCL4 might be a promising strategy to reduce inflammation in PMF.


Subject(s)
Bone Marrow/pathology , Fibrosis/pathology , Inflammation/pathology , Myeloproliferative Disorders/complications , Platelet Factor 4/metabolism , Primary Myelofibrosis/pathology , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Proliferation , Disease Progression , Fibrosis/etiology , Fibrosis/immunology , Fibrosis/metabolism , Humans , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Megakaryocytes , Mice , Mice, Knockout , Mutation , Platelet Factor 4/genetics , Primary Myelofibrosis/etiology , Primary Myelofibrosis/immunology , Primary Myelofibrosis/metabolism
4.
J Am Soc Nephrol ; 32(1): 52-68, 2021 01.
Article in English | MEDLINE | ID: mdl-33154175

ABSTRACT

BACKGROUND: Nephropathologic analyses provide important outcomes-related data in experiments with the animal models that are essential for understanding kidney disease pathophysiology. Precision medicine increases the demand for quantitative, unbiased, reproducible, and efficient histopathologic analyses, which will require novel high-throughput tools. A deep learning technique, the convolutional neural network, is increasingly applied in pathology because of its high performance in tasks like histology segmentation. METHODS: We investigated use of a convolutional neural network architecture for accurate segmentation of periodic acid-Schiff-stained kidney tissue from healthy mice and five murine disease models and from other species used in preclinical research. We trained the convolutional neural network to segment six major renal structures: glomerular tuft, glomerulus including Bowman's capsule, tubules, arteries, arterial lumina, and veins. To achieve high accuracy, we performed a large number of expert-based annotations, 72,722 in total. RESULTS: Multiclass segmentation performance was very high in all disease models. The convolutional neural network allowed high-throughput and large-scale, quantitative and comparative analyses of various models. In disease models, computational feature extraction revealed interstitial expansion, tubular dilation and atrophy, and glomerular size variability. Validation showed a high correlation of findings with current standard morphometric analysis. The convolutional neural network also showed high performance in other species used in research-including rats, pigs, bears, and marmosets-as well as in humans, providing a translational bridge between preclinical and clinical studies. CONCLUSIONS: We developed a deep learning algorithm for accurate multiclass segmentation of digital whole-slide images of periodic acid-Schiff-stained kidneys from various species and renal disease models. This enables reproducible quantitative histopathologic analyses in preclinical models that also might be applicable to clinical studies.


Subject(s)
Deep Learning , Diagnosis, Computer-Assisted , Kidney/physiopathology , Pattern Recognition, Automated , Algorithms , Animals , Disease Models, Animal , Image Processing, Computer-Assisted/methods , Kidney Diseases/pathology , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Neural Networks, Computer , Periodic Acid/chemistry , Reproducibility of Results , Schiff Bases , Translational Research, Biomedical
5.
Kidney Int ; 96(2): 505-516, 2019 08.
Article in English | MEDLINE | ID: mdl-31155155

ABSTRACT

Recent developments in optical tissue clearing have been difficult to apply for the morphometric analysis of organs with high cellular content and small functional structures, such as the kidney. Here, we establish combinations of genetic and immuno-labelling for single cell identification, tissue clearing and subsequent de-clarification for histoimmunopathology and transmission electron microscopy. Using advanced light microscopy and computational analyses, we investigated a murine model of crescentic nephritis, an inflammatory kidney disease typified by immune-mediated damage to glomeruli leading to the formation of hypercellular lesions and the rapid loss of kidney function induced by nephrotoxic serum. Results show a graded susceptibility of the glomeruli, significant podocyte loss and capillary injury. These effects are associated with activation of parietal epithelial cells and formation of glomerular lesions that may evolve and obstruct the kidney tubule, thereby explaining the loss of kidney function. Thus, our work provides new high-throughput endpoints for the analysis of complex tissues with single-cell resolution.


Subject(s)
Glomerulonephritis/pathology , Histocytological Preparation Techniques/methods , Imaging, Three-Dimensional , Podocytes/physiology , Single-Cell Analysis/methods , Animals , Capillaries , Disease Models, Animal , Disease Progression , Fluorescence , Fluorescent Dyes/chemistry , Genes, Reporter/genetics , Glomerulonephritis/immunology , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Humans , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Podocytes/ultrastructure
6.
J Am Soc Nephrol ; 24(12): 1966-80, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23970123

ABSTRACT

Under physiologic conditions, significant amounts of plasma protein pass the renal filter and are reabsorbed by proximal tubular cells, but it is not clear whether the endocytosed protein, particularly albumin, is degraded in lysosomes or returned to the circulatory system intact. To resolve this question, a transgenic mouse with podocyte-specific expression of doxycycline-inducible tagged murine albumin was developed. To assess potential glomerular backfiltration, two types of albumin with different charges were expressed. On administration of doxycycline, podocytes expressed either of the two types of transgenic albumin, which were secreted into the primary filtrate and reabsorbed by proximal tubular cells, resulting in serum accumulation. Renal transplantation experiments confirmed that extrarenal transcription of transgenic albumin was unlikely to account for these results. Genetic deletion of the neonatal Fc receptor (FcRn), which rescues albumin and IgG from lysosomal degradation, abolished transcytosis of both types of transgenic albumin and IgG in proximal tubular cells. In summary, we provide evidence of a transcytosis within the kidney tubular system that protects albumin and IgG from lysosomal degradation, allowing these proteins to be recycled intact.


Subject(s)
Albuminuria/metabolism , Kidney Tubules, Proximal/metabolism , Models, Biological , Serum Albumin/metabolism , Transcytosis/physiology , Animals , Anti-Bacterial Agents/pharmacology , Doxycycline/pharmacology , Endocytosis/physiology , Gene Expression/drug effects , Humans , Immunoglobulin G/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Kidney Transplantation , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Mice, Transgenic , Podocytes/metabolism , Protein Structure, Tertiary , Rats , Rats, Transgenic , Serum Albumin/chemistry , Serum Albumin/genetics
7.
Cell Rep ; 42(2): 112131, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36807143

ABSTRACT

Fibrosis represents the common end stage of chronic organ injury independent of the initial insult, destroying tissue architecture and driving organ failure. Here we discover a population of profibrotic macrophages marked by expression of Spp1, Fn1, and Arg1 (termed Spp1 macrophages), which expands after organ injury. Using an unbiased approach, we identify the chemokine (C-X-C motif) ligand 4 (CXCL4) to be among the top upregulated genes during profibrotic Spp1 macrophage differentiation. In vitro and in vivo studies show that loss of Cxcl4 abrogates profibrotic Spp1 macrophage differentiation and ameliorates fibrosis after both heart and kidney injury. Moreover, we find that platelets, the most abundant source of CXCL4 in vivo, drive profibrotic Spp1 macrophage differentiation. Single nuclear RNA sequencing with ligand-receptor interaction analysis reveals that macrophages orchestrate fibroblast activation via Spp1, Fn1, and Sema3 crosstalk. Finally, we confirm that Spp1 macrophages expand in both human chronic kidney disease and heart failure.


Subject(s)
Macrophages , Myofibroblasts , Humans , Fibrosis , Ligands , Macrophages/metabolism , Myofibroblasts/metabolism , Osteopontin , Platelet Factor 4/genetics , Platelet Factor 4/metabolism
8.
Nat Genet ; 54(11): 1690-1701, 2022 11.
Article in English | MEDLINE | ID: mdl-36303074

ABSTRACT

Adult kidney organoids have been described as strictly tubular epithelia and termed tubuloids. While the cellular origin of tubuloids has remained elusive, here we report that they originate from a distinct CD24+ epithelial subpopulation. Long-term-cultured CD24+ cell-derived tubuloids represent a functional human kidney tubule. We show that kidney tubuloids can be used to model the most common inherited kidney disease, namely autosomal dominant polycystic kidney disease (ADPKD), reconstituting the phenotypic hallmark of this disease with cyst formation. Single-cell RNA sequencing of CRISPR-Cas9 gene-edited PKD1- and PKD2-knockout tubuloids and human ADPKD and control tissue shows similarities in upregulation of disease-driving genes. Furthermore, in a proof of concept, we demonstrate that tolvaptan, the only approved drug for ADPKD, has a significant effect on cyst size in tubuloids but no effect on a pluripotent stem cell-derived model. Thus, tubuloids are derived from a tubular epithelial subpopulation and represent an advanced system for ADPKD disease modeling.


Subject(s)
Cysts , Polycystic Kidney, Autosomal Dominant , Adult , Humans , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Organoids , Kidney , CD24 Antigen/genetics
9.
Pediatr Nephrol ; 26(10): 1775-80, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21184239

ABSTRACT

In recent years, the podocyte, with its elaborate cytoarchitecture and slit diaphragm, has been the focus of extensive research, yet its precise role in the glomerular filtration barrier is still debated. There are puzzling observations indicating that a comprehensive mechanistic model for glomerular filtration is still necessary. There is no doubt that podocytes are essential for glomerular filtration barrier integrity. However, most albumin never reaches the podocyte because it is prevented from entering the glomerular filter at the endothelium level. Another puzzling observation is that the glomerular filter never clogs despite its high load of several kilograms of plasma proteins per day. Recently, we proposed a novel model in which an electrical potential difference is generated across the glomerular filtration barrier by filtration. The model offers novel potential solutions to some of the riddles regarding the glomerular filter.


Subject(s)
Kidney Glomerulus/pathology , Podocytes/pathology , Proteinuria/pathology , Animals , Endothelium/metabolism , Endothelium/pathology , Endothelium/physiopathology , Glomerular Filtration Barrier , Humans , Kidney/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/physiopathology , Proteinuria/metabolism , Proteinuria/physiopathology , Serum Albumin/metabolism
10.
J Am Soc Nephrol ; 21(12): 2053-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20947631

ABSTRACT

There is ongoing controversy about the mechanisms that determine the characteristics of the glomerular filter. Here, we tested whether flow across the glomerular filter generates extracellular electrical potential differences, which could be an important determinant of glomerular filtration. In micropuncture experiments in Necturus maculosus, we measured a potential difference across the glomerular filtration barrier that was proportional to filtration pressure (-0.045 mV/10 cm H2O). The filtration-dependent potential was generated without temporal delay and was negative within Bowman's space. Perfusion with the cationic polymer protamine abolished the potential difference. We propose a mathematical model that considers the relative contributions of diffusion, convection, and electrophoretic effects on the total flux of albumin across the filter. According to this model, potential differences of -0.02 to -0.05 mV can induce electrophoretic effects that significantly influence the glomerular sieving coefficient of albumin. This model of glomerular filtration has the potential to provide a mechanistic theory, based on experimental data, about the filtration characteristics of the glomerular filtration barrier. It provides a unique approach to the microanatomy of the glomerulus, renal autoregulation, and the pathogenesis of proteinuria.


Subject(s)
Cell Membrane Permeability/physiology , Glomerular Basement Membrane/physiology , Kidney Glomerulus/physiology , Membrane Potentials/physiology , Animals , Biological Transport, Active , Disease Models, Animal , Electric Impedance , Glomerular Basement Membrane/metabolism , Glomerular Filtration Rate , Humans , Kidney Diseases/physiopathology , Kidney Glomerulus/blood supply , Necturus maculosus , Renal Blood Flow, Effective/physiology
11.
Methods Mol Biol ; 2299: 323-338, 2021.
Article in English | MEDLINE | ID: mdl-34028752

ABSTRACT

Chronic kidney disease (CKD) affects over 10% of the worldwide population and kidney fibrosis is a main driver of CKD and considered a therapeutic target. The mechanisms leading to kidney fibrosis are highly complexed and can be best studied in rodent models. Here we describe the most commonly used kidney fibrosis models in mice, the unilateral ureteral obstruction (UUO) model and the ischemia reperfusion injury (IRI) model. Both models are easy to learn and can be applied in animals of different age, sex, and strain.


Subject(s)
Kidney/pathology , Reperfusion Injury/pathology , Ureteral Obstruction/pathology , Animals , Biomarkers/metabolism , Disease Models, Animal , Female , Fibrosis , Humans , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism , Ureteral Obstruction/metabolism
12.
Nat Commun ; 12(1): 6386, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34737275

ABSTRACT

A major drawback of single-cell ATAC-seq (scATAC-seq) is its sparsity, i.e., open chromatin regions with no reads due to loss of DNA material during the scATAC-seq protocol. Here, we propose scOpen, a computational method based on regularized non-negative matrix factorization for imputing and quantifying the open chromatin status of regulatory regions from sparse scATAC-seq experiments. We show that scOpen improves crucial downstream analysis steps of scATAC-seq data as clustering, visualization, cis-regulatory DNA interactions, and delineation of regulatory features. We demonstrate the power of scOpen to dissect regulatory changes in the development of fibrosis in the kidney. This identifies a role of Runx1 and target genes by promoting fibroblast to myofibroblast differentiation driving kidney fibrosis.


Subject(s)
Chromatin/metabolism , DNA/metabolism , Sequence Analysis, DNA/standards , Sequence Analysis, DNA/methods , Single-Cell Analysis/methods
SELECTION OF CITATIONS
SEARCH DETAIL