Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Nat Commun ; 15(1): 5956, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39009581

ABSTRACT

DNA methylation (DNAm) is one of the most reliable biomarkers of aging across mammalian tissues. While the age-dependent global loss of DNAm has been well characterized, DNAm gain is less characterized. Studies have demonstrated that CpGs which gain methylation with age are enriched in Polycomb Repressive Complex 2 (PRC2) targets. However, whole-genome examination of all PRC2 targets as well as determination of the pan-tissue or tissue-specific nature of these associations is lacking. Here, we show that low-methylated regions (LMRs) which are highly bound by PRC2 in embryonic stem cells (PRC2 LMRs) gain methylation with age in all examined somatic mitotic cells. We estimated that this epigenetic change represents around 90% of the age-dependent DNAm gain genome-wide. Therefore, we propose the "PRC2-AgeIndex," defined as the average DNAm in PRC2 LMRs, as a universal biomarker of cellular aging in somatic cells which can distinguish the effect of different anti-aging interventions.


Subject(s)
Aging , Biomarkers , DNA Methylation , Epigenesis, Genetic , Polycomb Repressive Complex 2 , Rejuvenation , Animals , Aging/metabolism , Aging/genetics , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/genetics , Rejuvenation/physiology , Biomarkers/metabolism , Humans , Mice , Cellular Senescence/genetics , CpG Islands , Embryonic Stem Cells/metabolism , Male , Female
2.
Nat Med ; 30(2): 360-372, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38355974

ABSTRACT

The search for biomarkers that quantify biological aging (particularly 'omic'-based biomarkers) has intensified in recent years. Such biomarkers could predict aging-related outcomes and could serve as surrogate endpoints for the evaluation of interventions promoting healthy aging and longevity. However, no consensus exists on how biomarkers of aging should be validated before their translation to the clinic. Here, we review current efforts to evaluate the predictive validity of omic biomarkers of aging in population studies, discuss challenges in comparability and generalizability and provide recommendations to facilitate future validation of biomarkers of aging. Finally, we discuss how systematic validation can accelerate clinical translation of biomarkers of aging and their use in gerotherapeutic clinical trials.


Subject(s)
Longevity , Research Design , Biomarkers , Consensus
4.
Nat Aging ; 4(1): 14-26, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38102454

ABSTRACT

Over the past decade, there has been a dramatic increase in efforts to ameliorate aging and the diseases it causes, with transient expression of nuclear reprogramming factors recently emerging as an intriguing approach. Expression of these factors, either systemically or in a tissue-specific manner, has been shown to combat age-related deterioration in mouse and human model systems at the cellular, tissue and organismal level. Here we discuss the current state of epigenetic rejuvenation strategies via partial reprogramming in both mouse and human models. For each classical reprogramming factor, we provide a brief description of its contribution to reprogramming and discuss additional factors or chemical strategies. We discuss what is known regarding chromatin remodeling and the molecular dynamics underlying rejuvenation, and, finally, we consider strategies to improve the practical uses of epigenetic reprogramming to treat aging and age-related diseases, focusing on the open questions and remaining challenges in this emerging field.


Subject(s)
Induced Pluripotent Stem Cells , Rejuvenation , Humans , Animals , Mice , Aging/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic
5.
Mol Cell ; 83(23): 4255-4271.e9, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37995687

ABSTRACT

Endogenous retroviruses (ERVs) are remnants of ancient parasitic infections and comprise sizable portions of most genomes. Although epigenetic mechanisms silence most ERVs by generating a repressive environment that prevents their expression (heterochromatin), little is known about mechanisms silencing ERVs residing in open regions of the genome (euchromatin). This is particularly important during embryonic development, where induction and repression of distinct classes of ERVs occur in short temporal windows. Here, we demonstrate that transcription-associated RNA degradation by the nuclear RNA exosome and Integrator is a regulatory mechanism that controls the productive transcription of most genes and many ERVs involved in preimplantation development. Disrupting nuclear RNA catabolism promotes dedifferentiation to a totipotent-like state characterized by defects in RNAPII elongation and decreased expression of long genes (gene-length asymmetry). Our results indicate that RNA catabolism is a core regulatory module of gene networks that safeguards RNAPII activity, ERV expression, cell identity, and developmental potency.


Subject(s)
Endogenous Retroviruses , Endogenous Retroviruses/genetics , RNA, Nuclear , Epigenesis, Genetic , Heterochromatin , Gene Expression
6.
Cell ; 186(18): 3758-3775, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37657418

ABSTRACT

With the rapid expansion of aging biology research, the identification and evaluation of longevity interventions in humans have become key goals of this field. Biomarkers of aging are critically important tools in achieving these objectives over realistic time frames. However, the current lack of standards and consensus on the properties of a reliable aging biomarker hinders their further development and validation for clinical applications. Here, we advance a framework for the terminology and characterization of biomarkers of aging, including classification and potential clinical use cases. We discuss validation steps and highlight ongoing challenges as potential areas in need of future research. This framework sets the stage for the development of valid biomarkers of aging and their ultimate utilization in clinical trials and practice.


Subject(s)
Aging , Longevity , Humans , Biomarkers
7.
Nat Commun ; 14(1): 5690, 2023 09 14.
Article in English | MEDLINE | ID: mdl-37709760

ABSTRACT

Generating primordial germ cell-like cells (PGCLCs) from human pluripotent stem cells (hPSCs) advances studies of human reproduction and development of infertility treatments, but often entails complex 3D aggregates. Here we develop a simplified, monolayer method to differentiate hPSCs into PGCs within 3.5 days. We use our simplified differentiation platform and single-cell RNA-sequencing to achieve further insights into PGCLC specification. Transient WNT activation for 12 h followed by WNT inhibition specified PGCLCs; by contrast, sustained WNT induced primitive streak. Thus, somatic cells (primitive streak) and PGCLCs are related-yet distinct-lineages segregated by temporally-dynamic signaling. Pluripotency factors including NANOG are continuously expressed during the transition from pluripotency to posterior epiblast to PGCs, thus bridging pluripotent and germline states. Finally, hPSC-derived PGCLCs can be easily purified by virtue of their CXCR4+PDGFRA-GARP- surface-marker profile and single-cell RNA-sequencing reveals that they harbor transcriptional similarities with fetal PGCs.


Subject(s)
Embryonic Development , Germ Cells , Humans , Cell Differentiation , Fetus , RNA
11.
Nat Commun ; 11(1): 1545, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32210226

ABSTRACT

Aging is characterized by a gradual loss of function occurring at the molecular, cellular, tissue and organismal levels. At the chromatin level, aging associates with progressive accumulation of epigenetic errors that eventually lead to aberrant gene regulation, stem cell exhaustion, senescence, and deregulated cell/tissue homeostasis. Nuclear reprogramming to pluripotency can revert both the age and the identity of any cell to that of an embryonic cell. Recent evidence shows that transient reprogramming can ameliorate age-associated hallmarks and extend lifespan in progeroid mice. However, it is unknown how this form of rejuvenation would apply to naturally aged human cells. Here we show that transient expression of nuclear reprogramming factors, mediated by expression of mRNAs, promotes a rapid and broad amelioration of cellular aging, including resetting of epigenetic clock, reduction of the inflammatory profile in chondrocytes, and restoration of youthful regenerative response to aged, human muscle stem cells, in each case without abolishing cellular identity.


Subject(s)
Cell Nucleus/metabolism , Cellular Reprogramming/physiology , Cellular Senescence/physiology , RNA, Messenger/metabolism , Rejuvenation/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Aging/physiology , Animals , Cells, Cultured , Chondrocytes , DNA Methylation/physiology , Endothelial Cells , Epigenesis, Genetic/physiology , Female , Fibroblasts , Gene Expression Profiling , Humans , Intravital Microscopy , Male , Mice , Middle Aged , Muscle Cells , Primary Cell Culture , Stem Cells , Young Adult
12.
Cell Stem Cell ; 24(5): 821-828.e5, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31051134

ABSTRACT

Genome editing of human pluripotent stem cells (hPSCs) provides powerful opportunities for in vitro disease modeling, drug discovery, and personalized stem cell-based therapeutics. Currently, only small edits can be engineered with high frequency, while larger modifications suffer from low efficiency and a resultant need for selection markers. Here, we describe marker-free genome editing in hPSCs using Cas9 ribonucleoproteins (RNPs) in combination with AAV6-mediated DNA repair template delivery. We report highly efficient and bi-allelic integration frequencies across multiple loci and hPSC lines, achieving mono-allelic editing frequencies of up to 94% at the HBB locus. Using this method, we show robust bi-allelic correction of homozygous sickle cell mutations in a patient-derived induced PSC (iPSC) line. Thus, this strategy shows significant utility for generating hPSCs with large gene integrations and/or single-nucleotide changes at high frequency and without the need for introducing selection genes, enhancing the applicability of hPSC editing for research and translational uses.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Genotype , Pluripotent Stem Cells/physiology , CRISPR-Associated Protein 9/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Repair , Gene Editing/methods , Gene Frequency , Genetic Engineering , Genetic Vectors/genetics , Homologous Recombination , Humans , Pathology, Molecular , Tissue Donors
13.
J Orthop Res ; 37(8): 1760-1770, 2019 08.
Article in English | MEDLINE | ID: mdl-31042308

ABSTRACT

There is intense clinical interest in the potential effects of platelet-rich plasma (PRP) for the treatment of osteoarthritis (OA). This study tested the hypotheses that (i) "lower" levels of the inflammatory mediators (IMs), interleukin-1ß, and tumor necrosis factor α (TNF-α) and (ii) "higher" levels of the growth factors (GFs), insulin-like growth factor 1, and transforming growth factor ß1 within leukocyte-poor PRP correlate with more favorable chondrocyte and macrophage responses in vitro. Samples were collected from 10 "healthy" young male (23-33 years old) human subjects (H-PRP) and nine older (62-85 years old) male patients with severe knee OA (OA-PRP). The samples were separated into groups of "high" or "low" levels of IM and GF based on multiplex cytokine and enzyme-linked immunosorbent assay data. Three-dimensional (3D) alginate bead chondrocyte cultures and monocyte-derived macrophage cultures were treated with 10% PRP from donors in different groups. Gene expression was analyzed by quantitative polymerase chain reaction. Contrary to our hypotheses, the effect of PRP on chondrocytes and macrophages was mainly influenced by the age and disease status of the PRP donor as opposed to the IM or GF groupings. While H-PRP showed similar effects on expression of chondrogenic markers (Col2a1 and Sox9) as the negative control group (p > 0.05), OA-PRP decreased chondrocyte expression of Col2a1 and Sox-9 messenger RNA by 40% and 30%, respectively (Col2a1, p = 0.015; Sox9, p = 0.037). OA-PRP also upregulated TNF-α and matrix metallopeptidase 9 (p < 0.001) gene expression in macrophages while H-PRP did not. This data suggests that PRP from older individuals with OA contain factors that may suppress chondrocyte matrix synthesis and promote macrophage inflammation in vitro. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1760-1770, 2019.


Subject(s)
Chondrocytes/metabolism , Cytokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Osteoarthritis, Knee/metabolism , Platelet-Rich Plasma/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Humans , Macrophages/metabolism , Male , Middle Aged , Young Adult
14.
Elife ; 72018 12 11.
Article in English | MEDLINE | ID: mdl-30526855

ABSTRACT

As part of the Reproducibility Project: Cancer Biology we published a Registered Report (Lesnik et al., 2016) that described how we intended to replicate selected experiments from the paper 'Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET' (Peinado et al., 2012). Here we report the results. We regenerated tumor cells stably expressing a short hairpin to reduce Met expression (shMet) using the same highly metastatic mouse melanoma cell line (B16-F10) as the original study, which efficiently downregulated Met in B16F10 cells similar to the original study (Supplementary Figure 5A; Peinado et al., 2012). Exosomes from control cells expressed Met, which was reduced in exosomes from shMet cells; however, we were unable to reliably detect phosphorylated Met in exosomes. We tested the effect of exosome-dependent Met signaling on primary tumor growth and metastasis. Similar to the results in the original study, we did not find a statistically significant change in primary tumor growth. Measuring lung and femur metastases, we found a small increase in metastatic burden with exosomes from control cells that was diminished when Met expression was reduced; however, while the effects were in the same direction as the original study (Figure 4E; Peinado et al., 2012), they were not statistically significant. Differences between the original study and this replication attempt, such as level of knockdown efficiency, cell line genetic drift, sample sizes, study endpoints, and variability of observed metastatic burden, are factors that might have influenced the outcomes. Finally, we report meta-analyses for each result.


Subject(s)
Bone Neoplasms/genetics , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Melanoma, Experimental/genetics , Proto-Oncogene Proteins c-met/genetics , Skin Neoplasms/genetics , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Communication , Cell Line, Tumor , Exosomes/pathology , Exosomes/transplantation , Female , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Lymphatic Metastasis , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Phenotype , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reproducibility of Results , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
15.
Nat Commun ; 9(1): 5078, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30510260

ABSTRACT

Royal jelly is the queen-maker for the honey bee Apis mellifera, and has cross-species effects on longevity, fertility, and regeneration in mammals. Despite this knowledge, how royal jelly or its components exert their myriad effects has remained poorly understood. Using mouse embryonic stem cells as a platform, here we report that through its major protein component Royalactin, royal jelly can maintain pluripotency by activating a ground-state pluripotency-like gene network. We further identify Regina, a mammalian structural analog of Royalactin that also induces a naive-like state in mouse embryonic stem cells. This reveals an important innate program for stem cell self-renewal with broad implications in understanding the molecular regulation of stem cell fate across species.


Subject(s)
Fatty Acids/pharmacology , Glycoproteins/pharmacology , Insect Proteins/pharmacology , Mammals/physiology , Mouse Embryonic Stem Cells/drug effects , Pluripotent Stem Cells/drug effects , Animals , Bees/metabolism , Chromatin , Fatty Acids/chemistry , Female , Fertility , Gene Expression Regulation, Developmental/drug effects , Glycoproteins/chemistry , Insect Proteins/chemistry , Lentivirus/genetics , Lentivirus/metabolism , Longevity , Mice , Models, Molecular , Recombinant Proteins , Teratoma/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
16.
Nat Methods ; 15(12): 1045-1047, 2018 12.
Article in English | MEDLINE | ID: mdl-30504872

ABSTRACT

Scarless genome editing in human pluripotent stem cells (hPSCs) represents a goal for both precise research applications and clinical translation of hPSC-derived therapies. Here we established a versatile and efficient method that combines CRISPR-Cas9-mediated homologous recombination with positive-negative selection of edited clones to generate scarless genetic changes in hPSCs.


Subject(s)
CRISPR-Cas Systems , Embryonic Stem Cells/metabolism , Gene Editing , Genome, Human , Homologous Recombination , Pluripotent Stem Cells/metabolism , RNA, Small Interfering/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation , Humans , Pluripotent Stem Cells/cytology
17.
Nat Commun ; 9(1): 4384, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30349051

ABSTRACT

Self-renewal and pluripotency in human embryonic stem cells (hESCs) depends upon the function of a remarkably small number of master transcription factors (TFs) that include OCT4, SOX2, and NANOG. Endogenous factors that regulate and maintain the expression of master TFs in hESCs remain largely unknown and/or uncharacterized. Here, we use a genome-wide, proteomics approach to identify proteins associated with the OCT4 enhancer. We identify known OCT4 regulators, plus a subset of potential regulators including a zinc finger protein, ZNF207, that plays diverse roles during development. In hESCs, ZNF207 partners with master pluripotency TFs to govern self-renewal and pluripotency while simultaneously controlling commitment of cells towards ectoderm through direct regulation of neuronal TFs, including OTX2. The distinct roles of ZNF207 during differentiation occur via isoform switching. Thus, a distinct isoform of ZNF207 functions in hESCs at the nexus that balances pluripotency and differentiation to ectoderm.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Microtubule-Associated Proteins/metabolism , Protein Isoforms/metabolism , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Chromatin Immunoprecipitation , Humans , Immunoprecipitation , Mass Spectrometry , Microtubule-Associated Proteins/genetics , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Protein Isoforms/genetics , RNA, Small Interfering/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
18.
Nat Struct Mol Biol ; 25(9): 885-893, 2018 09.
Article in English | MEDLINE | ID: mdl-30177761

ABSTRACT

Viral infection perturbs host cells and can be used to uncover regulatory mechanisms controlling cellular responses and susceptibility to infections. Using cell biological, biochemical, and genetic tools, we reveal that influenza A virus (IAV) infection induces global transcriptional defects at the 3' ends of active host genes and RNA polymerase II (RNAPII) run-through into extragenic regions. Deregulated RNAPII leads to expression of aberrant RNAs (3' extensions and host-gene fusions) that ultimately cause global transcriptional downregulation of physiological transcripts, an effect influencing antiviral response and virulence. This phenomenon occurs with multiple strains of IAV, is dependent on influenza NS1 protein, and can be modulated by SUMOylation of an intrinsically disordered region (IDR) of NS1 expressed by the 1918 pandemic IAV strain. Our data identify a strategy used by IAV to suppress host gene expression and indicate that polymorphisms in IDRs of viral proteins can affect the outcome of an infection.


Subject(s)
Influenza, Human/genetics , RNA Polymerase II/genetics , Terminator Regions, Genetic/genetics , Humans , Influenza A virus/pathogenicity , Influenza A virus/physiology , Virulence
19.
Stem Cells Dev ; 27(20): 1438-1448, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30153084

ABSTRACT

Human induced pluripotent stem cells (iPSCs) have the potential to repair/regenerate smooth muscle cells (SMCs) in different organs. However, there are many challenges in their translation to clinical therapies. In this study, we describe our observations of in vitro SMC differentiation in three iPSC lines derived from human fibroblasts using retroviral, episomal, and mRNA/miRNA reprogramming methods. We sought to elucidate correlations between differentiation characteristics and efficiencies that can facilitate large-scale production of differentiated cells for clinical applications, and to report differences in pluripotency marker expression in differentiated cells from different iPSC lines. A standardized SMC differentiation protocol was used to induce the CD31+/CD34+ vascular progenitor cell phenotype. These were sorted by magnetic-activated (MACS) and fluorescence-activated cell sorting (FACS), and then treated with PDGF-BB and smooth muscle growth medium for further differentiation into smooth muscle progenitor cells (pSMCs). The expression of SMC and pluripotency markers in early- and late-passage (P1 and P4) pSMCs was analyzed. A total of 36 differentiation runs was performed on the three patient iPSC lines. All pSMC populations expressed SMC markers and Ki67 consistent with the progenitor phenotype. Initial iPSC density correlated positively with the sorted cell FACS efficiency, and this correlation could be fit to a quadratic equation. We also observed that a specific "honeycomb" pattern of the starting cultured iPSCs cultured correlated with higher efficiency in all three iPSC lines. Pluripotency marker expression decreased significantly to nearly undetectable levels in all three lines. There was no significant change in SMC and pluripotent marker expression between passage 1 and 4. In summary, our observations suggest that the method of iPSC reprogramming does not affect iPSC differentiation into pSMCs. Protocol efficiency can be modeled mathematically and coupled with the initial "honeycomb" cell pattern to optimize production of large cell numbers for clinical therapies.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Myocytes, Smooth Muscle/cytology , Regeneration/genetics , Antigens, CD34/genetics , Becaplermin/pharmacology , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/metabolism , Ki-67 Antigen/genetics , Myoblasts/cytology , Myoblasts/metabolism , Myocytes, Smooth Muscle/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics
20.
Nat Cell Biol ; 20(8): 900-908, 2018 08.
Article in English | MEDLINE | ID: mdl-30013107

ABSTRACT

Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) is now routinely accomplished by overexpression of the four Yamanaka factors (OCT4, SOX2, KLF4, MYC (or OSKM))1. These iPSCs can be derived from patients' somatic cells and differentiated toward diverse fates, serving as a resource for basic and translational research. However, mechanistic insights into regulators and pathways that initiate the pluripotency network remain to be resolved. In particular, naturally occurring molecules that activate endogenous OCT4 and replace exogenous OCT4 in human iPSC reprogramming have yet to be found. Using a heterokaryon reprogramming system we identified NKX3-1 as an early and transiently expressed homeobox transcription factor. Following knockdown of NKX3-1, iPSC reprogramming is abrogated. NKX3-1 functions downstream of the IL-6-STAT3 regulatory network to activate endogenous OCT4. Importantly, NKX3-1 substitutes for exogenous OCT4 to reprogram both mouse and human fibroblasts at comparable efficiencies and generate fully pluripotent stem cells. Our findings establish an essential role for NKX3-1, a prostate-specific tumour suppressor, in iPSC reprogramming.


Subject(s)
Cellular Reprogramming Techniques , Cellular Reprogramming , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Time Factors , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL