Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Am Soc Nephrol ; 34(11): 1843-1862, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37639336

ABSTRACT

SIGNIFICANCE STATEMENT: Mouse models have been widely used to understand kidney disease pathomechanisms and play an important role in drug discovery. However, these models have not been systematically analyzed and compared. The authors characterized 18 different mouse kidney disease models at both bulk and single-cell gene expression levels and compared single-cell gene expression data from diabetic kidney disease (DKD) mice and from patients with DKD. Although single cell-level gene expression changes were mostly model-specific, different disease models showed similar changes when compared at a pathway level. The authors also found that changes in fractions of cell types are major drivers of bulk gene expression differences. Although the authors found only a small overlap of single cell-level gene expression changes between the mouse DKD model and patients, they observed consistent pathway-level changes. BACKGROUND: Mouse models have been widely used to understand kidney disease pathomechanisms and play an important role in drug discovery. However, these models have not been systematically analyzed and compared. METHODS: We analyzed single-cell RNA sequencing data (36 samples) and bulk gene expression data (42 samples) from 18 commonly used mouse kidney disease models. We compared single-nucleus RNA sequencing data from a mouse diabetic kidney disease model with data from patients with diabetic kidney disease and healthy controls. RESULTS: We generated a uniformly processed mouse single-cell atlas containing information for nearly 300,000 cells, identifying all major kidney cell types and states. Our analysis revealed that changes in fractions of cell types are major drivers of differences in bulk gene expression. Although gene expression changes at the single-cell level were mostly model-specific, different disease models showed similar changes when compared at a pathway level. Tensor decomposition analysis highlighted the important changes in proximal tubule cells in disease states. Specifically, we identified important alterations in expression of metabolic and inflammation-associated pathways. The mouse diabetic kidney disease model and patients with diabetic kidney disease shared only a small number of conserved cell type-specific differentially expressed genes, but we observed pathway-level activation patterns conserved between mouse and human diabetic kidney disease samples. CONCLUSIONS: This study provides a comprehensive mouse kidney single-cell atlas and defines gene expression commonalities and differences in disease states in mice. The results highlight the key role of cell heterogeneity in driving changes in bulk gene expression and the limited overlap of single-cell gene expression changes between animal models and patients, but they also reveal consistent pathway-level changes.


Subject(s)
Diabetic Nephropathies , Humans , Mice , Animals , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Kidney/metabolism , Kidney Tubules, Proximal/metabolism , Disease Models, Animal , Epithelial Cells/metabolism
2.
Mol Ther ; 30(7): 2491-2504, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35450819

ABSTRACT

Coding variants (named G1 and G2) in Apolipoprotein L1 (APOL1) can explain most excess risk of kidney disease observed in African American individuals. It has been proposed that risk variant APOL1 dose, such as increased risk variant APOL1 level serves as a trigger (second hit) for disease development. The goal of this study was to determine whether lowering risk variant APOL1 levels protects from disease development in a podocyte-specific transgenic mouse disease model. We administered antisense oligonucleotides (ASO) targeting APOL1 to podocyte-specific G2APOL1 mice and observed efficient reduction of APOL1 levels. APOL1 ASO1, which more efficiently lowered APOL1 transcript levels, protected mice from albuminuria, glomerulosclerosis, tubulointerstitial fibrosis, and renal failure. Administration of APOL1 ASO1 was effective even for established disease in the NEFTA-rtTA/TRE-G2APOL1 (NEFTA/G2APOL1) mice. We observed a strong correlation between APOL1 transcript level and disease severity. We concluded that APOL1 ASO1 may be an effective therapeutic approach for APOL1-associated glomerular disease.


Subject(s)
Kidney Diseases , Podocytes , Renal Insufficiency , Animals , Apolipoprotein L1/genetics , Apolipoproteins/genetics , Genetic Variation , Kidney Diseases/genetics , Kidney Diseases/therapy , Mice , Mice, Transgenic , Oligonucleotides, Antisense/genetics
3.
J Am Soc Nephrol ; 32(3): 614-627, 2021 03.
Article in English | MEDLINE | ID: mdl-33531352

ABSTRACT

BACKGROUND: Microscopic analysis of urine sediment is probably the most commonly used diagnostic procedure in nephrology. The urinary cells, however, have not yet undergone careful unbiased characterization. METHODS: Single-cell transcriptomic analysis was performed on 17 urine samples obtained from five subjects at two different occasions, using both spot and 24-hour urine collection. A pooled urine sample from multiple healthy individuals served as a reference control. In total 23,082 cells were analyzed. Urinary cells were compared with human kidney and human bladder datasets to understand similarities and differences among the observed cell types. RESULTS: Almost all kidney cell types can be identified in urine, such as podocyte, proximal tubule, loop of Henle, and collecting duct, in addition to macrophages, lymphocytes, and bladder cells. The urinary cell-type composition was subject specific and reasonably stable using different collection methods and over time. Urinary cells clustered with kidney and bladder cells, such as urinary podocytes with kidney podocytes, and principal cells of the kidney and urine, indicating their similarities in gene expression. CONCLUSIONS: A reference dataset for cells in human urine was generated. Single-cell transcriptomics enables detection and quantification of almost all types of cells in the kidney and urinary tract.


Subject(s)
Kidney/cytology , Aged , DNA Barcoding, Taxonomic , Female , Gene Library , Humans , Kidney/metabolism , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/urine , Male , Middle Aged , Podocytes/cytology , Podocytes/metabolism , RNA-Seq , Single-Cell Analysis/methods , Single-Cell Analysis/statistics & numerical data , Transcriptome , Urinary Bladder/cytology , Urinary Bladder/metabolism , Urine/cytology
4.
J Am Soc Nephrol ; 30(4): 594-609, 2019 04.
Article in English | MEDLINE | ID: mdl-30850438

ABSTRACT

BACKGROUND: Cytosine methylation of regulatory regions, such as promoters and enhancers, plays a key role in regulating gene expression, however, its role in kidney development has not been analyzed. METHODS: To identify functionally important epigenome-modifying enzymes and genome regions where methylation modifications are functionally important for kidney development, we performed genome-wide methylation analysis, expression profiling, and systematic genetic targeting of DNA methyltransferases (Dnmt1, Dnmt3a, and Dnmt3b) and Ten-eleven translocation methylcytosine hydroxylases (Tet2) in nephron progenitor cells (Six2Cre) in mice. RESULTS: Genome-wide methylome analysis indicated dynamic changes on promoters and enhancers during development. Six2CreDnmt3af/f, Six2CreDnmt3bf/f, and Six2CreTet2f/f mice showed no significant structural or functional renal abnormalities. In contrast, Six2CreDnmt1f/f mice died within 24 hours of birth, from a severe kidney developmental defect. Genome-wide methylation analysis indicated a marked loss of methylation of transposable elements. RNA sequencing detected endogenous retroviral transcripts. Expression of intracellular viral sensing pathways (RIG-I), early embryonic, nonrenal lineage genes and increased cell death contributed to the phenotype development. In podocytes, loss of Dnmt1, Dnmt3a, Dnmt3b, or Tet2 did not lead to functional or structural differences at baseline or after toxic injury. CONCLUSIONS: Genome-wide cytosine methylation and gene expression profiling showed that by silencing embryonic, nonrenal lineage genes and transposable elements, DNMT1-mediated cytosine methylation is essential for kidney development.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methylation , Epigenome/genetics , Homeodomain Proteins/genetics , Kidney/growth & development , Stem Cells/physiology , Transcription Factors/genetics , Animals , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA Transposable Elements , DNA-Binding Proteins/genetics , Dioxygenases , Enhancer Elements, Genetic , Gene Expression , Gene Silencing , Kidney/enzymology , Male , Mice , Podocytes/cytology , Podocytes/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , RNA, Viral/analysis , Sequence Analysis, RNA , Stem Cells/cytology , Transcriptome , DNA Methyltransferase 3B
5.
Nat Commun ; 15(1): 4325, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773071

ABSTRACT

Hematopoietic stem cell (HSC) mutations can result in clonal hematopoiesis (CH) with heterogeneous clinical outcomes. Here, we investigate how the cell state preceding Tet2 mutation impacts the pre-malignant phenotype. Using an inducible system for clonal analysis of myeloid progenitors, we find that the epigenetic features of clones at similar differentiation status are highly heterogeneous and functionally respond differently to Tet2 mutation. Cell differentiation stage also influences Tet2 mutation response indicating that the cell of origin's epigenome modulates clone-specific behaviors in CH. Molecular features associated with higher risk outcomes include Sox4 that sensitizes cells to Tet2 inactivation, inducing dedifferentiation, altered metabolism and increasing the in vivo clonal output of mutant cells, as confirmed in primary GMP and HSC models. Our findings validate the hypothesis that epigenetic features can predispose specific clones for dominance, explaining why identical genetic mutations can result in different phenotypes.


Subject(s)
DNA-Binding Proteins , Dioxygenases , Epigenesis, Genetic , Hematopoietic Stem Cells , Mutation , Proto-Oncogene Proteins , Dioxygenases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Animals , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Humans , Hematopoiesis/genetics , Mice , Cell Differentiation/genetics
6.
bioRxiv ; 2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37034577

ABSTRACT

Cis-regulatory elements control gene expression and are dynamic in their structure, reflecting changes to the composition of diverse effector proteins over time1-3. Here we sought to connect the structural changes at cis-regulatory elements to alterations in cellular fate and function. To do this we developed PRINT, a computational method that uses deep learning to correct sequence bias in chromatin accessibility data and identifies multi-scale footprints of DNA-protein interactions. We find that multi-scale footprints enable more accurate inference of TF and nucleosome binding. Using PRINT with single-cell multi-omics, we discover wide-spread changes to the structure and function of candidate cis-regulatory elements (cCREs) across hematopoiesis, wherein nucleosomes slide, expose DNA for TF binding, and promote gene expression. Activity segmentation using the co-variance across cell states identifies "sub-cCREs" as modular cCRE subunits of regulatory DNA. We apply this single-cell and PRINT approach to characterize the age-associated alterations to cCREs within hematopoietic stem cells (HSCs). Remarkably, we find a spectrum of aging alterations among HSCs corresponding to a global gain of sub-cCRE activity while preserving cCRE accessibility. Collectively, we reveal the functional importance of cCRE structure across cell states, highlighting changes to gene regulation at single-cell and single-base-pair resolution.

7.
Nat Biotechnol ; 2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37537502

ABSTRACT

Single-cell assay for transposase-accessible chromatin by sequencing (scATAC-seq) has emerged as a powerful tool for dissecting regulatory landscapes and cellular heterogeneity. However, an exploration of systemic biases among scATAC-seq technologies has remained absent. In this study, we benchmark the performance of eight scATAC-seq methods across 47 experiments using human peripheral blood mononuclear cells (PBMCs) as a reference sample and develop PUMATAC, a universal preprocessing pipeline, to handle the various sequencing data formats. Our analyses reveal significant differences in sequencing library complexity and tagmentation specificity, which impact cell-type annotation, genotype demultiplexing, peak calling, differential region accessibility and transcription factor motif enrichment. Our findings underscore the importance of sample extraction, method selection, data processing and total cost of experiments, offering valuable guidance for future research. Finally, our data and analysis pipeline encompasses 169,000 PBMC scATAC-seq profiles and a best practices code repository for scATAC-seq data analysis, which are freely available to extend this benchmarking effort to future protocols.

8.
Nat Commun ; 13(1): 4018, 2022 07 11.
Article in English | MEDLINE | ID: mdl-35821371

ABSTRACT

The kidney has tremendous capacity to repair after acute injury, however, pathways guiding adaptive and fibrotic repair are poorly understood. We developed a model of adaptive and fibrotic kidney regeneration by titrating ischemic injury dose. We performed detailed biochemical and histological analysis and profiled transcriptomic changes at bulk and single-cell level (> 110,000 cells) over time. Our analysis highlights kidney proximal tubule cells as key susceptible cells to injury. Adaptive proximal tubule repair correlated with fatty acid oxidation and oxidative phosphorylation. We identify a specific maladaptive/profibrotic proximal tubule cluster after long ischemia, which expresses proinflammatory and profibrotic cytokines and myeloid cell chemotactic factors. Druggability analysis highlights pyroptosis/ferroptosis as vulnerable pathways in these profibrotic cells. Pharmacological targeting of pyroptosis/ferroptosis in vivo pushed cells towards adaptive repair and ameliorates fibrosis. In summary, our single-cell analysis defines key differences in adaptive and fibrotic repair and identifies druggable pathways for pharmacological intervention to prevent kidney fibrosis.


Subject(s)
Acute Kidney Injury , Kidney , Acute Kidney Injury/drug therapy , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Fibrosis , Humans , Kidney/metabolism , Regeneration , Single-Cell Analysis
9.
Nat Metab ; 3(4): 469-484, 2021 04.
Article in English | MEDLINE | ID: mdl-33846639

ABSTRACT

Brown adipose tissue can expend large amounts of energy, and therefore increasing its size or activity is a promising therapeutic approach to combat metabolic disease. In humans, major deposits of brown fat cells are found intimately associated with large blood vessels, corresponding to perivascular adipose tissue (PVAT). However, the cellular origins of PVAT are poorly understood. Here, we determine the identity of perivascular adipocyte progenitors in mice and humans. In mice, thoracic PVAT develops from a fibroblastic lineage, consisting of progenitor cells (Pdgfra+, Ly6a+ and Pparg-) and preadipocytes (Pdgfra+, Ly6a+ and Pparg+), which share transcriptional similarity with analogous cell types in white adipose tissue. Interestingly, the aortic adventitia of adult animals contains a population of adipogenic smooth muscle cells (Myh11+, Pdgfra- and Pparg+) that contribute to perivascular adipocyte formation. Similarly, human PVAT contains presumptive fibroblastic and smooth muscle-like adipocyte progenitor cells, as revealed by single-nucleus RNA sequencing. Together, these studies define distinct populations of progenitor cells for thermogenic PVAT, providing a foundation for developing strategies to augment brown fat activity.


Subject(s)
Adipocytes, Brown/physiology , Adipose Tissue, Brown/physiology , Cell Lineage/physiology , Thermogenesis/physiology , Adipocytes, White/physiology , Adipogenesis/physiology , Adipose Tissue, Brown/growth & development , Animals , Animals, Newborn , Aorta/cytology , Aorta/physiology , Blood Vessels/physiology , Cell Lineage/genetics , Fibroblasts/physiology , Gene Expression Regulation/physiology , Humans , Infant, Newborn , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/physiology , Stem Cells/physiology , Thermogenesis/genetics
10.
Nat Commun ; 12(1): 2277, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859189

ABSTRACT

Determining the epigenetic program that generates unique cell types in the kidney is critical for understanding cell-type heterogeneity during tissue homeostasis and injury response. Here, we profile open chromatin and gene expression in developing and adult mouse kidneys at single cell resolution. We show critical reliance of gene expression on distal regulatory elements (enhancers). We reveal key cell type-specific transcription factors and major gene-regulatory circuits for kidney cells. Dynamic chromatin and expression changes during nephron progenitor differentiation demonstrates that podocyte commitment occurs early and is associated with sustained Foxl1 expression. Renal tubule cells follow a more complex differentiation, where Hfn4a is associated with proximal and Tfap2b with distal fate. Mapping single nucleotide variants associated with human kidney disease implicates critical cell types, developmental stages, genes, and regulatory mechanisms. The single cell multi-omics atlas reveals key chromatin remodeling events and gene expression dynamics associated with kidney development.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Nephrons/growth & development , Organogenesis/genetics , Renal Insufficiency, Chronic/genetics , Animals , Cell Communication , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Epigenomics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genetic Loci/genetics , Genome-Wide Association Study , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Humans , Mice , Nephrons/cytology , Podocytes/physiology , Polymorphism, Single Nucleotide , RNA-Seq , Renal Insufficiency, Chronic/pathology , Single-Cell Analysis , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism
11.
Commun Biol ; 3(1): 306, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32533049

ABSTRACT

Melanoma represents the most serious type of skin cancer. Although recent years have seen advances using targeted and immunotherapies, most patients remain at high risk for tumor recurrence. Here we show that IRAK-M, a negative regulator of MyD88 signaling, is deficient or low in melanoma and expression levels correlate with patient survival. Inducing IRAK-M expression using genetic approaches or epigenetic modifiers initiates apoptosis by prompting its interaction with TRAF6 via IRAK-M's C-terminal domain. This complex recruits and degrades calpastatin which stimulates calpain activity and triggers caspase-3-dependent but caspase-8,-9-independent apoptosis. Using a drug screen, we identified compounds that induced IRAK-M expression. Administration of IRAK-M-inducing drugs reduced tumor growth in mice but was ineffective against IRAK-M knock-down tumors. These results uncover a previously uncharacterized apoptosis pathway, emphasize IRAK-M as a potential therapeutic target and suggest that the anticancer activity of certain drugs could do so through their ability to induce IRAK-M expression.


Subject(s)
Apoptosis , Calcium-Binding Proteins/antagonists & inhibitors , Caspase 3/metabolism , Gene Expression Regulation, Neoplastic , Interleukin-1 Receptor-Associated Kinases/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Melanoma/pathology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caspase 3/genetics , Cell Proliferation , Female , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Elife ; 92020 04 14.
Article in English | MEDLINE | ID: mdl-32286228

ABSTRACT

Bone marrow mesenchymal lineage cells are a heterogeneous cell population involved in bone homeostasis and diseases such as osteoporosis. While it is long postulated that they originate from mesenchymal stem cells, the true identity of progenitors and their in vivo bifurcated differentiation routes into osteoblasts and adipocytes remain poorly understood. Here, by employing large scale single cell transcriptome analysis, we computationally defined mesenchymal progenitors at different stages and delineated their bi-lineage differentiation paths in young, adult and aging mice. One identified subpopulation is a unique cell type that expresses adipocyte markers but contains no lipid droplets. As non-proliferative precursors for adipocytes, they exist abundantly as pericytes and stromal cells that form a ubiquitous 3D network inside the marrow cavity. Functionally they play critical roles in maintaining marrow vasculature and suppressing bone formation. Therefore, we name them marrow adipogenic lineage precursors (MALPs) and conclude that they are a newly identified component of marrow adipose tissue.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Lineage , Mesenchymal Stem Cells/cytology , Animals , Genomics/methods , Mice , Transcriptome
13.
Cell Metab ; 30(4): 784-799.e5, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31474566

ABSTRACT

Fibrosis is the final common pathway leading to end-stage renal failure. By analyzing the kidneys of patients and animal models with fibrosis, we observed a significant mitochondrial defect, including the loss of the mitochondrial transcription factor A (TFAM) in kidney tubule cells. Here, we generated mice with tubule-specific deletion of TFAM (Ksp-Cre/Tfamflox/flox). While these mice developed severe mitochondrial loss and energetic deficit by 6 weeks of age, kidney fibrosis, immune cell infiltration, and progressive azotemia causing death were only observed around 12 weeks of age. In renal cells of TFAM KO (knockout) mice, aberrant packaging of the mitochondrial DNA (mtDNA) resulted in its cytosolic translocation, activation of the cytosolic cGAS-stimulator of interferon genes (STING) DNA sensing pathway, and thus cytokine expression and immune cell recruitment. Ablation of STING ameliorated kidney fibrosis in mouse models of chronic kidney disease, demonstrating how TFAM sequesters mtDNA to limit the inflammation leading to fibrosis.


Subject(s)
DNA, Mitochondrial/metabolism , Kidney Tubules/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Renal Insufficiency, Chronic/pathology , Animals , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Humans , Inflammation/pathology , Kidney Tubules/pathology , Male , Mice , Mitochondrial Proteins/metabolism , RAW 264.7 Cells , Transcription Factors/metabolism
14.
Nat Genet ; 51(1): 51-62, 2019 01.
Article in English | MEDLINE | ID: mdl-30578418

ABSTRACT

In this trans-ethnic multi-omic study, we reinterpret the genetic architecture of blood pressure to identify genes, tissues, phenomes and medication contexts of blood pressure homeostasis. We discovered 208 novel common blood pressure SNPs and 53 rare variants in genome-wide association studies of systolic, diastolic and pulse pressure in up to 776,078 participants from the Million Veteran Program (MVP) and collaborating studies, with analysis of the blood pressure clinical phenome in MVP. Our transcriptome-wide association study detected 4,043 blood pressure associations with genetically predicted gene expression of 840 genes in 45 tissues, and mouse renal single-cell RNA sequencing identified upregulated blood pressure genes in kidney tubule cells.


Subject(s)
Blood Pressure/genetics , Ethnicity/genetics , Adolescent , Animals , Female , Gene Expression/genetics , Genome-Wide Association Study/methods , Humans , Kidney Tubules/physiology , Male , Mice , Middle Aged , Polymorphism, Single Nucleotide/genetics , Transcriptome/genetics , Up-Regulation/genetics
15.
Science ; 360(6390): 758-763, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29622724

ABSTRACT

Our understanding of kidney disease pathogenesis is limited by an incomplete molecular characterization of the cell types responsible for the organ's multiple homeostatic functions. To help fill this knowledge gap, we characterized 57,979 cells from healthy mouse kidneys by using unbiased single-cell RNA sequencing. On the basis of gene expression patterns, we infer that inherited kidney diseases that arise from distinct genetic mutations but share the same phenotypic manifestation originate from the same differentiated cell type. We also found that the collecting duct in kidneys of adult mice generates a spectrum of cell types through a newly identified transitional cell. Computational cell trajectory analysis and in vivo lineage tracing revealed that intercalated cells and principal cells undergo transitions mediated by the Notch signaling pathway. In mouse and human kidney disease, these transitions were shifted toward a principal cell fate and were associated with metabolic acidosis.


Subject(s)
Cell Tracking/methods , Gene Expression Profiling/methods , Kidney Diseases/genetics , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Single-Cell Analysis/methods , Animals , Cell Plasticity , Genetic Markers , Humans , Mice , Receptors, Notch/metabolism , Sequence Analysis, RNA/methods , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL