Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Exp Med ; 221(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38889332

ABSTRACT

ILC2s are key players in type 2 immunity and contribute to maintaining homeostasis. ILC2s are also implicated in the development of type 2 inflammation-mediated chronic disorders like asthma. While memory ILC2s have been identified in mouse, it is unknown whether human ILC2s can acquire immunological memory. Here, we demonstrate the persistence of CD45RO, a marker previously linked to inflammatory ILC2s, in resting ILC2s that have undergone prior activation. A high proportion of these cells concurrently reduce the expression of the canonical ILC marker CD127 in a tissue-specific manner. Upon isolation and in vitro stimulation of CD127-CD45RO+ ILC2s, we observed an augmented ability to proliferate and produce cytokines. CD127-CD45RO+ ILC2s are found in both healthy and inflamed tissues and display a gene signature of cell activation. Similarly, mouse memory ILC2s show reduced expression of CD127. Our findings suggest that human ILC2s can acquire innate immune memory and warrant a revision of the current strategies to identify human ILC2s.


Subject(s)
Immunity, Innate , Immunologic Memory , Interleukin-7 Receptor alpha Subunit , Lymphocytes , Humans , Immunologic Memory/immunology , Animals , Interleukin-7 Receptor alpha Subunit/metabolism , Lymphocytes/immunology , Mice , Immunity, Innate/immunology , Leukocyte Common Antigens/metabolism , Cytokines/metabolism , Inflammation/immunology , Female , Mice, Inbred C57BL
2.
Sci Immunol ; 8(87): eadf7702, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37774008

ABSTRACT

Allergic disorders are caused by a combination of hereditary and environmental factors. The hygiene hypothesis postulates that early-life microbial exposures impede the development of subsequent allergic disease. Recently developed "wildling" mice are genetically identical to standard laboratory specific pathogen-free (SPF) mice but are housed under seminatural conditions and have rich microbial exposures from birth. Thus, by comparing conventional SPF mice with wildlings, we can uncouple the impact of lifelong microbial exposures from genetic factors on the allergic immune response. We found that wildlings developed larger populations of antigen-experienced T cells than conventional SPF mice, which included interleukin-10-producing CD4 T cells specific for commensal Lactobacilli strains and allergy-promoting T helper 2 (TH2) cells. In models of airway exposure to house dust mite (HDM), recombinant interleukin-33, or Alternaria alternata, wildlings developed strong allergic inflammation, characterized by eosinophil recruitment, goblet cell metaplasia, and antigen-specific immunoglobulin G1 (IgG1) and IgE responses. Wildlings developed robust de novo TH2 cell responses to incoming allergens, whereas preexisting TH2 cells could also be recruited into the allergic immune response in a cytokine-driven and TCR-independent fashion. Thus, wildling mice, which experience diverse and lifelong microbial exposures, were not protected from developing pathological allergic immune responses. Instead, wildlings mounted robust allergic responses to incoming allergens, shedding new light on the hygiene hypothesis.


Subject(s)
Hypersensitivity , Th2 Cells , Mice , Animals , Cytokines , Allergens , Immunity
3.
Blood Adv ; 6(6): 1780-1796, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35016204

ABSTRACT

How genetic haploinsufficiency contributes to the clonal dominance of hematopoietic stem cells (HSCs) in del(5q) myelodysplastic syndrome (MDS) remains unresolved. Using a genetic barcoding strategy, we performed a systematic comparison on genes implicated in the pathogenesis of del(5q) MDS in direct competition with each other and wild-type (WT) cells with single-clone resolution. Csnk1a1 haploinsufficient HSCs expanded (oligo)clonally and outcompeted all other tested genes and combinations. Csnk1a1-/+ multipotent progenitors showed a proproliferative gene signature and HSCs showed a downregulation of inflammatory signaling/immune response. In validation experiments, Csnk1a1-/+ HSCs outperformed their WT counterparts under a chronic inflammation stimulus, also known to be caused by neighboring genes on chromosome 5. We therefore propose a crucial role for Csnk1a1 haploinsufficiency in the selective advantage of 5q-HSCs, implemented by creation of a unique competitive advantage through increased HSC self-renewal and proliferation capacity, as well as increased fitness under inflammatory stress.


Subject(s)
Chromosome Deletion , Myelodysplastic Syndromes , Haploinsufficiency , Hematopoietic Stem Cells/pathology , Humans , Myelodysplastic Syndromes/pathology
4.
Nat Commun ; 12(1): 2474, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33931648

ABSTRACT

As more clinically-relevant genomic features of myeloid malignancies are revealed, it has become clear that targeted clinical genetic testing is inadequate for risk stratification. Here, we develop and validate a clinical transcriptome-based assay for stratification of acute myeloid leukemia (AML). Comparison of ribonucleic acid sequencing (RNA-Seq) to whole genome and exome sequencing reveals that a standalone RNA-Seq assay offers the greatest diagnostic return, enabling identification of expressed gene fusions, single nucleotide and short insertion/deletion variants, and whole-transcriptome expression information. Expression data from 154 AML patients are used to develop a novel AML prognostic score, which is strongly associated with patient outcomes across 620 patients from three independent cohorts, and 42 patients from a prospective cohort. When combined with molecular risk guidelines, the risk score allows for the re-stratification of 22.1 to 25.3% of AML patients from three independent cohorts into correct risk groups. Within the adverse-risk subgroup, we identify a subset of patients characterized by dysregulated integrin signaling and RUNX1 or TP53 mutation. We show that these patients may benefit from therapy with inhibitors of focal adhesion kinase, encoded by PTK2, demonstrating additional utility of transcriptome-based testing for therapy selection in myeloid malignancy.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic/genetics , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/metabolism , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cohort Studies , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Female , Gene Fusion , Humans , INDEL Mutation , Integrins/genetics , Integrins/metabolism , Leukemia, Myeloid, Acute/genetics , Male , Polymorphism, Single Nucleotide , Prognosis , Prospective Studies , RNA-Seq , Risk Factors , Signal Transduction/genetics , Survival Analysis , Transcriptome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Exome Sequencing , Whole Genome Sequencing
5.
Exp Hematol ; 91: 22-31, 2020 11.
Article in English | MEDLINE | ID: mdl-32976949

ABSTRACT

The discovery that the immunomodulatory imide drugs (IMiDs) possess antitumor properties revolutionized the treatment of specific types of hematological cancers. Since then, much progress has been made in understanding why the IMiDs are so efficient in targeting the malignant clones in difficult-to-treat diseases. Despite their efficacy, IMiD resistance arises eventually. Herein we summarize the mechanisms of sensitivity and resistance to lenalidomide in del(5q) myelodysplastic syndrome and multiple myeloma, two diseases in which these drugs are at the therapeutic frontline. Understanding the molecular and cellular mechanisms underlying IMiD efficacy and resistance may allow development of specific strategies to eliminate the malignant clone in otherwise incurable diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Immunologic Factors/pharmacology , Lenalidomide/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Anemia, Macrocytic/drug therapy , Anemia, Macrocytic/physiopathology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Cell Differentiation/drug effects , Chromosome Deletion , Chromosomes, Human, Pair 5 , Cytokines/metabolism , Disease Progression , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ikaros Transcription Factor/antagonists & inhibitors , Immunologic Factors/therapeutic use , Lenalidomide/therapeutic use , Megakaryocytes/drug effects , Multiple Myeloma/drug therapy , Multiple Myeloma/physiopathology , Neoplasm Proteins/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology , Phosphoprotein Phosphatases/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/physiology
6.
Nat Cell Biol ; 22(5): 526-533, 2020 05.
Article in English | MEDLINE | ID: mdl-32251398

ABSTRACT

Interstitial deletion of the long arm of chromosome 5 (del(5q)) is the most common structural genomic variant in myelodysplastic syndromes (MDS)1. Lenalidomide (LEN) is the treatment of choice for patients with del(5q) MDS, but half of the responding patients become resistant2 within 2 years. TP53 mutations are detected in ~20% of LEN-resistant patients3. Here we show that patients who become resistant to LEN harbour recurrent variants of TP53 or RUNX1. LEN upregulated RUNX1 protein and function in a CRBN- and TP53-dependent manner in del(5q) cells, and mutation or downregulation of RUNX1 rendered cells resistant to LEN. LEN induced megakaryocytic differentiation of del(5q) cells followed by cell death that was dependent on calpain activation and CSNK1A1 degradation4,5. We also identified GATA2 as a LEN-responsive gene that is required for LEN-induced megakaryocyte differentiation. Megakaryocytic gene-promoter analyses suggested that LEN-induced degradation of IKZF1 enables a RUNX1-GATA2 complex to drive megakaryocytic differentiation. Overexpression of GATA2 restored LEN sensitivity in the context of RUNX1 or TP53 mutations by enhancing LEN-induced megakaryocytic differentiation. Screening for mutations that block LEN-induced megakaryocytic differentiation should identify patients who are resistant to LEN.


Subject(s)
Cell Differentiation/drug effects , Cell Differentiation/genetics , Chromosomes, Human, Pair 5/genetics , Lenalidomide/pharmacology , Megakaryocytes/drug effects , Myelodysplastic Syndromes/genetics , Cell Line , Chromosomes, Human, Pair 5/drug effects , Core Binding Factor Alpha 2 Subunit/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , GATA2 Transcription Factor/genetics , HEK293 Cells , Humans , Mutation/drug effects , Mutation/genetics , Tumor Suppressor Protein p53/genetics
7.
Nat Commun ; 9(1): 2418, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29925839

ABSTRACT

Expression of miR-143 and miR-145 is reduced in hematopoietic stem/progenitor cells (HSPCs) of myelodysplastic syndrome patients with a deletion in the long arm of chromosome 5. Here we show that mice lacking miR-143/145 have impaired HSPC activity with depletion of functional hematopoietic stem cells (HSCs), but activation of progenitor cells (HPCs). We identify components of the transforming growth factor ß (TGFß) pathway as key targets of miR-143/145. Enforced expression of the TGFß adaptor protein and miR-145 target, Disabled-2 (DAB2), recapitulates the HSC defect seen in miR-143/145-/- mice. Despite reduced HSC activity, older miR-143/145-/- and DAB2-expressing mice show elevated leukocyte counts associated with increased HPC activity. A subset of mice develop a serially transplantable myeloid malignancy, associated with expansion of HPC. Thus, miR-143/145 play a cell context-dependent role in HSPC function through regulation of TGFß/DAB2 activation, and loss of these miRNAs creates a preleukemic state.


Subject(s)
Hematopoietic Stem Cells/metabolism , MicroRNAs/metabolism , Myelodysplastic Syndromes/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Apoptosis Regulatory Proteins , Bone Marrow/pathology , Bone Marrow Transplantation , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Myelodysplastic Syndromes/pathology , Transplantation Chimera
8.
PLoS One ; 10(8): e0134569, 2015.
Article in English | MEDLINE | ID: mdl-26248042

ABSTRACT

A limited repertoire of PPP family of serine/threonine phosphatases with a highly conserved catalytic domain acts on thousands of protein targets to orchestrate myriad central biological roles. A major structural reorganization of human calcineurin, a ubiquitous Ser/Thr PPP regulated by calcium and calmodulin and targeted by immunosuppressant drugs cyclosporin A and FK506, is unveiled here. The new conformation involves trans- to cis-isomerization of proline in the SAPNY sequence, highly conserved across PPPs, and remodels the main regulatory site where NFATc transcription factors bind. Transitions between cis- and trans-conformations may involve peptidyl prolyl isomerases such as cyclophilin A and FKBP12, which are known to physically interact with and modulate calcineurin even in the absence of immunosuppressant drugs. Alternative conformations in PPPs provide a new perspective on interactions with substrates and other protein partners and may foster development of more specific inhibitors as drug candidates.


Subject(s)
Calcineurin/metabolism , Amino Acid Sequence , Binding Sites , Calcineurin/chemistry , Calcineurin/genetics , Catalytic Domain , Crystallography, X-Ray , Cyclophilin A/metabolism , Cyclosporine/chemistry , Cyclosporine/metabolism , HEK293 Cells , Humans , Isomerism , Molecular Dynamics Simulation , Molecular Sequence Data , NFATC Transcription Factors/chemistry , NFATC Transcription Factors/metabolism , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment , Tacrolimus Binding Protein 1A/metabolism
9.
Carcinogenesis ; 36(7): 792-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25916653

ABSTRACT

The members of the human regulators of calcineurin (RCAN) protein family are endogenous regulators of the calcineurin (CN)-cytosolic nuclear factor of activated T-cells (NFATc) pathway activation. This function is explained by the presence of a highly conserved calcipressin inhibitor of calcineurin (CIC) motif in RCAN proteins, which has been shown to compete with NFATc for the binding to CN and therefore are able to inhibit NFATc dephosphorylation and activation by CN. Very recently, emerging roles for NFATc proteins in transformation, tumor angiogenesis and metastasis have been described in different cancer cell types. In this work, we report that the overexpression of RCAN3 dramatically inhibits tumor growth and tumor angiogenesis in an orthotopic human breast cancer model. We suggest that RCAN3 exerts these effects in a CN-dependent manner, as mutation of the CIC motif in RCAN3 abolishes the tumor suppressor effect. Moreover, the expression of the EGFP-R3(178-210) peptide, spanning the CIC motif of RCAN3, is able to reproduce all the antitumor effects of RCAN3 full-length protein. Finally, we show that RCAN3 and the EGFP-R3(178-210) peptide inhibit the CN-NFATc signaling pathway and the induction of the NFATc-dependent gene cyclooxygenase-2. Our work suggests that the EGFP-R3(178-210) peptide possess potent tumor suppressor properties and therefore constitutes a novel lead for the development of potent and specific antitumoral agents. Moreover, we propose the targeting of the CN-NFATc pathway in the tumor cells constitutes an effective way to hamper tumor progression by impairing the paracrine network among tumor, endothelial and polymorphonucleated cells.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Peptide Fragments/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Breast Neoplasms/metabolism , Calcineurin/metabolism , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice, Inbred BALB C , Mice, Nude , NFATC Transcription Factors , Neovascularization, Pathologic/genetics , Peptide Fragments/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
10.
Biochem Biophys Res Commun ; 460(2): 295-301, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25783055

ABSTRACT

Cooperation between calcineurin (CN)-NFATc and RAF-MEK-ERK signaling pathways is essential in thymocyte positive selection. It is known that the Regulators of Calcineurin (RCAN) proteins can act either facilitating or suppressing CN-dependent signaling events. Here, we show that RCAN genes are expressed in lymphoid tissues, and address the role of RCAN proteins in T cell development. Overexpression of human RCAN3 and RCAN1 can modulate T cell development by increasing positive selection-related surface markers, as well as the "Erk(hi) competence state" in double positive thymocytes, a characteristic molecular signature of positive selection, without affecting CN activity. We also found that RCAN1/3 interact with RAF kinases and CN in a non-exclusive manner. Our data suggests that the balance of RCAN interactions with CN and/or RAF kinases may influence T cell positive selection.


Subject(s)
Carrier Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Muscle Proteins/physiology , Thymus Gland/immunology , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Carrier Proteins/immunology , Female , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins/immunology , Mice , Mice, Inbred C57BL , Muscle Proteins/immunology , Real-Time Polymerase Chain Reaction , Thymus Gland/embryology
11.
PLoS One ; 9(1): e85539, 2014.
Article in English | MEDLINE | ID: mdl-24465593

ABSTRACT

Recently there has been much interest in the Regulators of Calcineurin (RCAN) proteins which are important endogenous modulators of the calcineurin-NFATc signalling pathway. They have been shown to have a crucial role in cellular programmes such as the immune response, muscle fibre remodelling and memory, but also in pathological processes such as cardiac hypertrophy and neurodegenerative diseases. In vertebrates, the RCAN family form a functional subfamily of three members RCAN1, RCAN2 and RCAN3 whereas only one RCAN is present in the rest of Eukarya. In addition, RCAN genes have been shown to collocate with RUNX and CLIC genes in ACD clusters (ACD21, ACD6 and ACD1). How the RCAN genes and their clustering in ACDs evolved is still unknown. After analysing RCAN gene family evolution using bioinformatic tools, we propose that the three RCAN vertebrate genes within the ACD clusters, which evolved from single copy genes present in invertebrates and lower eukaryotes, are the result of two rounds of whole genome duplication, followed by a segmental duplication. This evolutionary scenario involves the loss or gain of some RCAN genes during evolution. In addition, we have analysed RCAN gene structure and identified the existence of several characteristic features that can be involved in RCAN evolution and gene expression regulation. These included: several transposable elements, CpG islands in the 5' region of the genes, the existence of antisense transcripts (NAT) associated with the three human genes, and considerable evidence for bidirectional promoters that regulate RCAN gene expression. Furthermore, we show that the CpG island associated with the RCAN3 gene promoter is unmethylated and transcriptionally active. All these results provide timely new insights into the molecular mechanisms underlying RCAN function and a more in depth knowledge of this gene family whose members are obvious candidates for the development of future therapies.


Subject(s)
Biological Evolution , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/genetics , Muscle Proteins/genetics , Animals , Chromosome Mapping , DNA-Binding Proteins , Genome , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , Vertebrates/genetics
12.
Biochim Biophys Acta ; 1833(10): 2311-21, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23732701

ABSTRACT

Cyclosporine A and FK506 produce immunosuppression by blocking calcineurin phosphatase activity and consequently activation of cytosolic Nuclear Factor of Activated T-cell (NFATc) transcription factor. Due to the chronic toxicity associated with their administration, the development of more specific immunosuppressants is currently an important unmet medical need. In this context, an immunosuppressant peptide derived from the CIC motif of the human Regulators of Calcineurin (RCAN) proteins has been shown to inhibit NFATc signaling without affecting general phosphatase activity of calcineurin. Here we show that protein kinase CK2 phosphorylates a conserved serine residue within the CIC motif of vertebrate RCANs, which increases its affinity for calcineurin and consequently its inhibition of NFATc-dependent gene expression in activated T-cells. Molecular modeling studies have led us to identify a positively charged interaction site on the surface of calcineurin where the phosphorylated serine residue of the CIC motif would normally locate. Finally, we have also identified RCAN3 as a new phosphoprotein with multiple phosphorylation sites. Therefore, our findings reveal for the first time a novel molecular mechanism underlying the regulation of calcineurin-NFATc signaling by means of phosphorylation of the CIC motif of RCAN proteins. The knowledge of how RCAN proteins modulate the calcineurin-NFATc pathway paves the way for the development of potent novel selective immunosuppressant drugs.


Subject(s)
Calcineurin/metabolism , Casein Kinase II/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Proteins/metabolism , NFATC Transcription Factors/metabolism , Amino Acid Sequence , Blotting, Western , Calcineurin/genetics , Casein Kinase II/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Circular Dichroism , DNA-Binding Proteins , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Luciferases/metabolism , Molecular Sequence Data , Muscle Proteins/genetics , NFATC Transcription Factors/genetics , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction
13.
J Biol Chem ; 284(14): 9394-401, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19189965

ABSTRACT

Calcineurin phosphatase plays a crucial role in T cell activation. Dephosphorylation of the nuclear factors of activated T cells (NFATs) by calcineurin is essential for activating cytokine gene expression and, consequently, the immune response. Current immunosuppressive protocols are based mainly on calcineurin inhibitors, cyclosporine A and FK506. Unfortunately, these drugs are associated with severe side effects. Therefore, immunosuppressive agents with higher selectivity and lower toxicity must be identified. The immunosuppressive role of the family of proteins regulators of calcineurin (RCAN, formerly known as DSCR1) which regulate the calcineurin-NFAT signaling pathway, has been described recently. Here, we identify and characterize the minimal RCAN sequence responsible for the inhibition of calcineurin-NFAT signaling in vivo. The RCAN-derived peptide spanning this sequence binds to calcineurin with high affinity. This interaction is competed by a peptide spanning the NFAT PXIXIT sequence, which binds to calcineurin and facilitates NFAT dephosphorylation and activation. Interestingly, the RCAN-derived peptide does not inhibit general calcineurin phosphatase activity, which suggests that it may have a specific immunosuppressive effect on the calcineurin-NFAT signaling pathway. As such, the RCAN-derived peptide could either be considered a highly selective immunosuppressive compound by itself or be used as a new tool for identifying innovative immunosuppressive agents. We developed a low throughput assay, based on the RCAN1-calcineurin interaction, which identifies dipyridamole as an efficient in vivo inhibitor of the calcineurin-NFAT pathway that does not affect calcineurin phosphatase activity.


Subject(s)
Calcineurin/metabolism , NFATC Transcription Factors/metabolism , Peptide Fragments/metabolism , Phosphoric Monoester Hydrolases/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Dipyridamole/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Protein Binding , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...