Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Dev Cell ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38823395

ABSTRACT

Control of cellular identity requires coordination of developmental programs with environmental factors such as nutrient availability, suggesting that perturbing metabolism can alter cell state. Here, we find that nucleotide depletion and DNA replication stress drive differentiation in human and murine normal and transformed hematopoietic systems, including patient-derived acute myeloid leukemia (AML) xenografts. These cell state transitions begin during S phase and are independent of ATR/ATM checkpoint signaling, double-stranded DNA break formation, and changes in cell cycle length. In systems where differentiation is blocked by oncogenic transcription factor expression, replication stress activates primed regulatory loci and induces lineage-appropriate maturation genes despite the persistence of progenitor programs. Altering the baseline cell state by manipulating transcription factor expression causes replication stress to induce genes specific for alternative lineages. The ability of replication stress to selectively activate primed maturation programs across different contexts suggests a general mechanism by which changes in metabolism can promote lineage-appropriate cell state transitions.

2.
Nat Commun ; 15(1): 3159, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38605040

ABSTRACT

How RNA-binding proteins (RBPs) convey regulatory instructions to the core effectors of RNA processing is unclear. Here, we document the existence and functions of a multivalent RBP-effector interface. We show that the effector interface of a conserved RBP with an essential role in metazoan development, Unkempt, is mediated by a novel type of 'dual-purpose' peptide motifs that can contact two different surfaces of interacting proteins. Unexpectedly, we find that the multivalent contacts do not merely serve effector recruitment but are required for the accuracy of RNA recognition by Unkempt. Systems analyses reveal that multivalent RBP-effector contacts can repurpose the principal activity of an effector for a different function, as we demonstrate for the reuse of the central eukaryotic mRNA decay factor CCR4-NOT in translational control. Our study establishes the molecular assembly and functional principles of an RBP-effector interface.


Subject(s)
RNA-Binding Proteins , RNA , Animals , RNA-Binding Proteins/metabolism , RNA/metabolism , RNA Processing, Post-Transcriptional , Peptides/metabolism
3.
bioRxiv ; 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37790431

ABSTRACT

RNA-binding proteins (RBPs) are key regulators of gene expression, but how RBPs convey regulatory instructions to the core effectors of RNA processing is unclear. Here we document the existence and functions of a multivalent RBP-effector interface. We show that the effector interface of a deeply conserved RBP with an essential role in metazoan development, Unkempt, is mediated by a novel type of 'dual-purpose' peptide motifs that can contact two different surfaces of interacting proteins. Unexpectedly, we find that the multivalent contacts do not merely serve effector recruitment but are required for the accuracy of RNA recognition by the recruiting RBP. Systems analyses reveal that multivalent RBP-effector contacts can repurpose the principal activity of an effector for a different function, as we demonstrate for reuse of the central eukaryotic mRNA decay factor CCR4-NOT in translational control. Our study establishes the molecular assembly and functional principles of an RBP-effector interface, with implications for the evolution and function of RBP-operated regulatory networks.

4.
Nat Rev Genet ; 24(5): 276-294, 2023 05.
Article in English | MEDLINE | ID: mdl-36418462

ABSTRACT

RNA-binding proteins (RBPs) regulate essentially every event in the lifetime of an RNA molecule, from its production to its destruction. Whereas much has been learned about RNA sequence specificity and general functions of individual RBPs, the ways in which numerous RBPs instruct a much smaller number of effector molecules, that is, the core engines of RNA processing, as to where, when and how to act remain largely speculative. Here, we survey the known modes of communication between RBPs and their effectors with a particular focus on converging RBP-effector interactions and their roles in reducing the complexity of RNA networks. We discern the emerging unifying principles and discuss their utility in our understanding of RBP function, regulation of biological processes and contribution to human disease.


Subject(s)
RNA Processing, Post-Transcriptional , RNA , Humans , RNA/genetics , RNA/metabolism , RNA-Binding Proteins/genetics
5.
Nat Commun ; 13(1): 2350, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35487911

ABSTRACT

Cell fate commitment is driven by dynamic changes in chromatin architecture and activity of lineage-specific transcription factors (TFs). The chromatin assembly factor-1 (CAF-1) is a histone chaperone that regulates chromatin architecture by facilitating nucleosome assembly during DNA replication. Accumulating evidence supports a substantial role of CAF-1 in cell fate maintenance, but the mechanisms by which CAF-1 restricts lineage choice remain poorly understood. Here, we investigate how CAF-1 influences chromatin dynamics and TF activity during lineage differentiation. We show that CAF-1 suppression triggers rapid differentiation of myeloid stem and progenitor cells into a mixed lineage state. We find that CAF-1 sustains lineage fidelity by controlling chromatin accessibility at specific loci, and limiting the binding of ELF1 TF at newly-accessible diverging regulatory elements. Together, our findings decipher key traits of chromatin accessibility that sustain lineage integrity and point to a powerful strategy for dissecting transcriptional circuits central to cell fate commitment.


Subject(s)
Chromatin , Histone Chaperones , Chromatin Assembly Factor-1/genetics , Chromatin Assembly Factor-1/metabolism , Chromosomes/metabolism , Histone Chaperones/metabolism , Histones/metabolism
6.
Cell Rep ; 37(6): 109967, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758323

ABSTRACT

Stem and progenitor cells have the capacity to balance self-renewal and differentiation. Hematopoietic myeloid progenitors replenish more than 25 billion terminally differentiated neutrophils every day under homeostatic conditions and can increase this output in response to stress or infection. At what point along the spectrum of maturation do progenitors lose capacity for self-renewal and become irreversibly committed to differentiation? Using a system of conditional myeloid development that can be toggled between self-renewal and differentiation, we interrogate determinants of this "point of no return" in differentiation commitment. Irreversible commitment is due primarily to loss of open regulatory site access and disruption of a positive feedback transcription factor activation loop. Restoration of the transcription factor feedback loop extends the window of cell plasticity and alters the point of no return. These findings demonstrate how the chromatin state enforces and perpetuates cell fate and identify potential avenues for manipulating cell identity.


Subject(s)
Bone Marrow/physiology , Cell Lineage , Chromatin/genetics , Hematopoiesis , Hematopoietic Stem Cells/cytology , Myeloid Cells/cytology , Transcription Factors/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chromatin/metabolism , Female , Gene Expression Profiling , Mice , Transcription Factors/genetics
7.
Front Cell Dev Biol ; 9: 654915, 2021.
Article in English | MEDLINE | ID: mdl-33959610

ABSTRACT

An expanding repertoire of histone variants and specialized histone chaperone partners showcases the versatility of nucleosome assembly during different cellular processes. Recent research has suggested an integral role of nucleosome assembly pathways in both maintaining cell identity and influencing cell fate decisions during development and normal homeostasis. Mutations and altered expression profiles of histones and corresponding histone chaperone partners are associated with developmental defects and cancer. Here, we discuss the spatiotemporal deposition mechanisms of the Histone H3 variants and their influence on mammalian cell fate during development. We focus on H3 given its profound effect on nucleosome stability and its recently characterized deposition pathways. We propose that differences in deposition of H3 variants are largely dependent on the phase of the cell cycle and cellular potency but are also affected by cellular stress and changes in cell fate. We also discuss the utility of modern technologies in dissecting the spatiotemporal control of H3 variant deposition, and how this could shed light on the mechanisms of cell identity maintenance and lineage commitment. The current knowledge and future studies will help us better understand how organisms employ nucleosome dynamics in health, disease, and aging. Ultimately, these pathways can be manipulated to induce cell fate change in a therapeutic setting depending on the cellular context.

8.
Nat Cell Biol ; 23(4): 424-436, 2021 04.
Article in English | MEDLINE | ID: mdl-33820973

ABSTRACT

Although high-throughput RNA sequencing (RNA-seq) has greatly advanced small non-coding RNA (sncRNA) discovery, the currently widely used complementary DNA library construction protocol generates biased sequencing results. This is partially due to RNA modifications that interfere with adapter ligation and reverse transcription processes, which prevent the detection of sncRNAs bearing these modifications. Here, we present PANDORA-seq (panoramic RNA display by overcoming RNA modification aborted sequencing), employing a combinatorial enzymatic treatment to remove key RNA modifications that block adapter ligation and reverse transcription. PANDORA-seq identified abundant modified sncRNAs-mostly transfer RNA-derived small RNAs (tsRNAs) and ribosomal RNA-derived small RNAs (rsRNAs)-that were previously undetected, exhibiting tissue-specific expression across mouse brain, liver, spleen and sperm, as well as cell-specific expression across embryonic stem cells (ESCs) and HeLa cells. Using PANDORA-seq, we revealed unprecedented landscapes of microRNA, tsRNA and rsRNA dynamics during the generation of induced pluripotent stem cells. Importantly, tsRNAs and rsRNAs that are downregulated during somatic cell reprogramming impact cellular translation in ESCs, suggesting a role in lineage differentiation.


Subject(s)
RNA Processing, Post-Transcriptional/genetics , RNA, Small Untranslated/genetics , RNA-Seq , Transcriptome/genetics , DNA, Complementary/genetics , HeLa Cells , Humans , MicroRNAs/genetics , RNA, Ribosomal/genetics
10.
Cell Stem Cell ; 23(2): 289-305.e5, 2018 Aug 02.
Article in English | MEDLINE | ID: mdl-30017590

ABSTRACT

Cellular reprogramming converts differentiated cells into induced pluripotent stem cells (iPSCs). However, this process is typically very inefficient, complicating mechanistic studies. We identified and molecularly characterized rare, early intermediates poised to reprogram with up to 95% efficiency, without perturbing additional genes or pathways, during iPSC generation from mouse embryonic fibroblasts. Analysis of these cells uncovered transcription factors (e.g., Tfap2c and Bex2) that are important for reprogramming but dispensable for pluripotency maintenance. Additionally, we observed striking patterns of chromatin hyperaccessibility at pluripotency loci, which preceded gene expression in poised intermediates. Finally, inspection of these hyperaccessible regions revealed an early wave of DNA demethylation that is uncoupled from de novo methylation of somatic regions late in reprogramming. Our study underscores the importance of investigating rare intermediates poised to produce iPSCs, provides insights into reprogramming mechanisms, and offers a valuable resource for the dissection of transcriptional and epigenetic dynamics intrinsic to cell fate change.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , Cells, Cultured , Female , Male , Mice , Mice, Inbred BALB C
11.
Curr Opin Genet Dev ; 46: 83-94, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28692904

ABSTRACT

During embryonic development, cells become progressively restricted in their differentiation potential. This is thought to be regulated by dynamic changes in chromatin structure and associated modifications, which act together to stabilize distinct specialized cell lineages. Remarkably, differentiated cells can be experimentally reprogrammed to a stem cell-like state or to alternative lineages. Thus, cellular reprogramming provides a valuable platform to study the mechanisms that normally safeguard cell identity and uncover factors whose manipulation facilitates cell fate transitions. Recent work has identified the chromatin assembly factor complex CAF-1 as a potent barrier to cellular reprogramming. In addition, CAF-1 has been implicated in the reversion of pluripotent cells to a totipotent-like state and in various lineage conversion paradigms, suggesting that modulation of CAF-1 levels may endow cells with a developmentally more plastic state. Here, we review these exciting results, discuss potential mechanisms and speculate on the possibility of exploiting chromatin assembly pathways to manipulate cell identity.


Subject(s)
Cell Differentiation/genetics , Cell Plasticity/genetics , Cellular Reprogramming/genetics , Transcription Factors/genetics , Animals , Cell Lineage/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Humans
12.
Nat Microbiol ; 2: 16250, 2016 Dec 05.
Article in English | MEDLINE | ID: mdl-27918527

ABSTRACT

Influenza A virus (IAV) causes annual epidemics and occasional pandemics, and is one of the best-characterized human RNA viral pathogens1. However, a physiologically relevant role for the RNA interference (RNAi) suppressor activity of the IAV non-structural protein 1 (NS1), reported over a decade ago2, remains unknown3. Plant and insect viruses have evolved diverse virulence proteins to suppress RNAi as their hosts produce virus-derived small interfering RNAs (siRNAs) that direct specific antiviral defence4-7 by an RNAi mechanism dependent on the slicing activity of Argonaute proteins (AGOs)8,9. Recent studies have documented induction and suppression of antiviral RNAi in mouse embryonic stem cells and suckling mice10,11. However, it is still under debate whether infection by IAV or any other RNA virus that infects humans induces and/or suppresses antiviral RNAi in mature mammalian somatic cells12-21. Here, we demonstrate that mature human somatic cells produce abundant virus-derived siRNAs co-immunoprecipitated with AGOs in response to IAV infection. We show that the biogenesis of viral siRNAs from IAV double-stranded RNA (dsRNA) precursors in infected cells is mediated by wild-type human Dicer and potently suppressed by both NS1 of IAV as well as virion protein 35 (VP35) of Ebola and Marburg filoviruses. We further demonstrate that the slicing catalytic activity of AGO2 inhibits IAV and other RNA viruses in mature mammalian cells, in an interferon-independent fashion. Altogether, our work shows that IAV infection induces and suppresses antiviral RNAi in differentiated mammalian somatic cells.


Subject(s)
Host-Pathogen Interactions , Influenza A virus/immunology , RNA Interference , Animals , Argonaute Proteins/metabolism , Cell Line , Chromatin Immunoprecipitation , Humans , Protein Binding , RNA, Viral/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors
13.
Stem Cell Reports ; 6(5): 704-716, 2016 05 10.
Article in English | MEDLINE | ID: mdl-26947976

ABSTRACT

The generation of induced pluripotent stem cells (iPSCs) from differentiated cells following forced expression of OCT4, KLF4, SOX2, and C-MYC (OKSM) is slow and inefficient, suggesting that transcription factors have to overcome somatic barriers that resist cell fate change. Here, we performed an unbiased serial shRNA enrichment screen to identify potent repressors of somatic cell reprogramming into iPSCs. This effort uncovered the protein modifier SUMO2 as one of the strongest roadblocks to iPSC formation. Depletion of SUMO2 both enhances and accelerates reprogramming, yielding transgene-independent, chimera-competent iPSCs after as little as 38 hr of OKSM expression. We further show that the SUMO2 pathway acts independently of exogenous C-MYC expression and in parallel with small-molecule enhancers of reprogramming. Importantly, suppression of SUMO2 also promotes the generation of human iPSCs. Together, our results reveal sumoylation as a crucial post-transcriptional mechanism that resists the acquisition of pluripotency from fibroblasts using defined factors.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Cellular Reprogramming/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , RNA, Small Interfering/genetics
14.
Nature ; 528(7581): 218-24, 2015 Dec 10.
Article in English | MEDLINE | ID: mdl-26659182

ABSTRACT

Cellular differentiation involves profound remodelling of chromatic landscapes, yet the mechanisms by which somatic cell identity is subsequently maintained remain incompletely understood. To further elucidate regulatory pathways that safeguard the somatic state, we performed two comprehensive RNA interference (RNAi) screens targeting chromatin factors during transcription-factor-mediated reprogramming of mouse fibroblasts to induced pluripotent stem cells (iPS cells). Subunits of the chromatin assembly factor-1 (CAF-1) complex, including Chaf1a and Chaf1b, emerged as the most prominent hits from both screens, followed by modulators of lysine sumoylation and heterochromatin maintenance. Optimal modulation of both CAF-1 and transcription factor levels increased reprogramming efficiency by several orders of magnitude and facilitated iPS cell formation in as little as 4 days. Mechanistically, CAF-1 suppression led to a more accessible chromatin structure at enhancer elements early during reprogramming. These changes were accompanied by a decrease in somatic heterochromatin domains, increased binding of Sox2 to pluripotency-specific targets and activation of associated genes. Notably, suppression of CAF-1 also enhanced the direct conversion of B cells into macrophages and fibroblasts into neurons. Together, our findings reveal the histone chaperone CAF-1 to be a novel regulator of somatic cell identity during transcription-factor-induced cell-fate transitions and provide a potential strategy to modulate cellular plasticity in a regenerative setting.


Subject(s)
Cellular Reprogramming/genetics , Chromatin Assembly Factor-1/metabolism , Animals , Cells, Cultured , Chromatin/metabolism , Chromatin Assembly Factor-1/antagonists & inhibitors , Chromatin Assembly Factor-1/genetics , Gene Expression Regulation/genetics , Heterochromatin/metabolism , Mice , Nucleosomes/metabolism , RNA Interference , Transduction, Genetic
15.
Genes Dev ; 29(5): 501-12, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25737280

ABSTRACT

Cellular morphology is an essential determinant of cellular function in all kingdoms of life, yet little is known about how cell shape is controlled. Here we describe a molecular program that controls the early morphology of neurons through a metazoan-specific zinc finger protein, Unkempt. Depletion of Unkempt in mouse embryos disrupts the shape of migrating neurons, while ectopic expression confers neuronal-like morphology to cells of different nonneuronal lineages. We found that Unkempt is a sequence-specific RNA-binding protein and identified its precise binding sites within coding regions of mRNAs linked to protein metabolism and trafficking. RNA binding is required for Unkempt-induced remodeling of cellular shape and is directly coupled to a reduced production of the encoded proteins. These findings link post-transcriptional regulation of gene expression with cellular shape and have general implications for the development and disease of multicellular organisms.


Subject(s)
Cell Shape/genetics , Gene Expression Regulation, Developmental , Neurons/cytology , Animals , Brain/metabolism , Cell Line , Embryo, Mammalian , Gene Expression Profiling , HeLa Cells , Humans , Mice , Protein Binding , RNA, Messenger
16.
Cell Stem Cell ; 12(6): 699-712, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23665121

ABSTRACT

The chromatin state of pluripotency genes has been studied extensively in embryonic stem cells (ESCs) and differentiated cells, but their potential interactions with other parts of the genome remain largely unexplored. Here, we identified a genome-wide, pluripotency-specific interaction network around the Nanog promoter by adapting circular chromosome conformation capture sequencing. This network was rearranged during differentiation and restored in induced pluripotent stem cells. A large fraction of Nanog-interacting loci were bound by Mediator or cohesin in pluripotent cells. Depletion of these proteins from ESCs resulted in a disruption of contacts and the acquisition of a differentiation-specific interaction pattern prior to obvious transcriptional and phenotypic changes. Similarly, the establishment of Nanog interactions during reprogramming often preceded transcriptional upregulation of associated genes, suggesting a causative link. Our results document a complex, pluripotency-specific chromatin "interactome" for Nanog and suggest a functional role for long-range genomic interactions in the maintenance and induction of pluripotency.


Subject(s)
Cellular Reprogramming/genetics , Chromatin/genetics , Chromatin/metabolism , Genome/genetics , Homeodomain Proteins/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Homeodomain Proteins/metabolism , Humans , Mice , Nanog Homeobox Protein
17.
Cell ; 151(7): 1617-32, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23260147

ABSTRACT

Factor-induced reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) is inefficient, complicating mechanistic studies. Here, we examined defined intermediate cell populations poised to becoming iPSCs by genome-wide analyses. We show that induced pluripotency elicits two transcriptional waves, which are driven by c-Myc/Klf4 (first wave) and Oct4/Sox2/Klf4 (second wave). Cells that become refractory to reprogramming activate the first but fail to initiate the second transcriptional wave and can be rescued by elevated expression of all four factors. The establishment of bivalent domains occurs gradually after the first wave, whereas changes in DNA methylation take place after the second wave when cells acquire stable pluripotency. This integrative analysis allowed us to identify genes that act as roadblocks during reprogramming and surface markers that further enrich for cells prone to forming iPSCs. Collectively, our data offer new mechanistic insights into the nature and sequence of molecular events inherent to cellular reprogramming.


Subject(s)
Cellular Reprogramming , Cytological Techniques/methods , Induced Pluripotent Stem Cells/cytology , Animals , Genome-Wide Association Study , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Mice , Transcription Factors/metabolism
19.
J Virol ; 86(8): 4151-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22318144

ABSTRACT

RNA interference (RNAi) is a critical component of many cellular antiviral responses in plants, invertebrates, and mammals. However, its in vivo role in host protection from the negative-sense RNA virus influenza virus type A (flu) is unclear. Here we have examined the role of RNAi in host defense to flu by analyzing Argonaute 1 and 3 double-knockout mice deficient in components of the RNA-induced silencing complex. Compared to littermate controls, flu-infected double-knockout mice exhibited increased mortality, consistent with more severe alveolitis and pneumonitis. These data indicate that optimal resistance to flu requires Argonaute 1 and/or 3. Enhanced mortality of double-knockout mice was not associated either with increased viral replication or with differential pulmonary recruitment or function of innate and adaptive immune cells. Given the absence of detectable immune defects, our results support the notion that the enhanced flu susceptibility of double-knockout mice arises from an intrinsic impairment in the ability of lung cells to tolerate flu-elicited inflammation.


Subject(s)
Argonaute Proteins/genetics , Eukaryotic Initiation Factors/genetics , Genetic Predisposition to Disease , Influenza A virus/immunology , Orthomyxoviridae Infections/genetics , Animals , Humans , Lung/immunology , Lung/virology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , RNA Interference , Virus Replication
20.
Genes Dev ; 24(17): 1951-60, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20713509

ABSTRACT

The canonical microRNA (miRNA) biogenesis pathway requires two RNaseIII enzymes: Drosha and Dicer. To understand their functions in mammals in vivo, we engineered mice with germline or tissue-specific inactivation of the genes encoding these two proteins. Changes in proteomic and transcriptional profiles that were shared in Dicer- and Drosha-deficient mice confirmed the requirement for both enzymes in canonical miRNA biogenesis. However, deficiency in Drosha or Dicer did not always result in identical phenotypes, suggesting additional functions. We found that, in early-stage thymocytes, Drosha recognizes and directly cleaves many protein-coding messenger RNAs (mRNAs) with secondary stem-loop structures. In addition, we identified a subset of miRNAs generated by a Dicer-dependent but Drosha-independent mechanism. These were distinct from previously described mirtrons. Thus, in mammalian cells, Dicer is required for the biogenesis of multiple classes of miRNAs. Together, these findings extend the range of function of RNaseIII enzymes beyond canonical miRNA biogenesis, and help explain the nonoverlapping phenotypes caused by Drosha and Dicer deficiency.


Subject(s)
DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/metabolism , Endoribonucleases/deficiency , Endoribonucleases/metabolism , MicroRNAs/biosynthesis , Ribonuclease III/deficiency , Ribonuclease III/metabolism , Animals , Cell Differentiation , Cells, Cultured , DEAD-box RNA Helicases/genetics , Endoribonucleases/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Inverted Repeat Sequences , Mice , Phenotype , RNA, Messenger/metabolism , Ribonuclease III/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...