Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Stem Cells Dev ; 33(3-4): 89-103, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38164089

ABSTRACT

Mesenchymal stem cells (MSCs) directly differentiate into neurons and endothelial cells after transplantation, and their secretome has considerable potential for treating brain injuries. Previous studies have suggested that the effects of MSCs priming with exposure to hypoxia, cytokines, growth factors, or chemical agents could optimize the paracrine potency and therapeutic potential of MSCs. Studies have suggested that thrombin-primed Wharton's Jelly-derived mesenchymal stem cells (Th.WJ-MSCs) significantly enhance the neuroprotective beneficial effects of naive MSCs in brain injury such as hypoxic-ischemic brain injury (HIE) and intraventricular hemorrhage (IVH). This study aimed to characterize WJ-MSCs in terms of stem cell markers, differentiation, cell proliferation, and paracrine factors by comparing naive and Th.WJ-MSCs. We demonstrated that compared with naive MSCs, Th.MSCs significantly enhanced the neuroprotective effects in vitro. Moreover, we identified differentially expressed proteins in the conditioned media of naive and Th.WJ-MSCs by liquid chromatography-tandem mass spectrometry analysis. Secretome analysis of the conditioned medium of WJ-MSCs revealed that such neuroprotective effects were mediated by paracrine effects with secretomes of Th.WJ-MSCs, and hepatocyte growth factor was identified as a key paracrine mediator. These results can be applied further in the preclinical and clinical development of effective and safe cell therapeutics for brain injuries such as HIE and IVH.


Subject(s)
Brain Injuries , Mesenchymal Stem Cells , Neuroprotective Agents , STAT3 Transcription Factor , Wharton Jelly , Humans , Hepatocyte Growth Factor/metabolism , Neuroprotective Agents/pharmacology , Thrombin/pharmacology , Thrombin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Signal Transduction , Cell Differentiation , Immunologic Factors/metabolism , Brain Injuries/metabolism , Cell Proliferation
2.
Hemasphere ; 7(11): e977, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37908861

ABSTRACT

Aplastic anemia (AA) is a lethal hematological disorder; however, its pathogenesis is not fully understood. Although immunosuppressive therapy (IST) is a major treatment option for AA, one-third of patients do not respond to IST and its resistance mechanism remains elusive. To understand AA pathogenesis and IST resistance, we performed single-cell RNA sequencing (scRNA-seq) of bone marrow (BM) from healthy controls and patients with AA at diagnosis. We found that CD34+ early-stage erythroid precursor cells and PROM1+ hematopoietic stem cells were significantly depleted in AA, which suggests that the depletion of CD34+ early-stage erythroid precursor cells and PROM1+ hematopoietic stem cells might be one of the major mechanisms for AA pathogenesis related with BM-cell hypoplasia. More importantly, we observed the significant enrichment of CD8+ T cells and T cell-activating intercellular interactions in IST responders, indicating the association between the expansion and activation of T cells and the positive response of IST in AA. Taken together, our findings represent a valuable resource offering novel insights into the cellular heterogeneity in the BM of AA and reveal potential biomarkers for IST, building the foundation for future precision therapies in AA.

3.
Int J Mol Sci ; 21(24)2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33327533

ABSTRACT

The function of natural killer (NK) cell-derived interferon-γ (IFN-γ) expands to remove pathogens by increasing the ability of innate immune cells. Here, we identified the critical role of thioredoxin-interacting protein (TXNIP) in the production of IFN-γ in NK cells during bacterial infection. TXNIP inhibited the production of IFN-γ and the activation of transforming growth factor ß-activated kinase 1 (TAK1) activity in primary mouse and human NK cells. TXNIP directly interacted with TAK1 and inhibited TAK1 activity by interfering with the complex formation between TAK1 and TAK1 binding protein 1 (TAB1). Txnip-/- (KO) NK cells enhanced the activation of macrophages by inducing IFN-γ production during Pam3CSK4 stimulation or Staphylococcus aureus (S. aureus) infection and contributed to expedite the bacterial clearance. Our findings suggest that NK cell-derived IFN-γ is critical for host defense and that TXNIP plays an important role as an inhibitor of NK cell-mediated macrophage activation by inhibiting the production of IFN-γ during bacterial infection.


Subject(s)
Carrier Proteins/metabolism , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Thioredoxins/metabolism , Animals , Carrier Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Immunity, Innate/drug effects , Immunity, Innate/genetics , Killer Cells, Natural/immunology , Lipopeptides/pharmacology , Mice , Mice, Knockout , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcus aureus/pathogenicity , Thioredoxins/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
4.
Biochem Biophys Res Commun ; 507(1-4): 489-495, 2018 12 09.
Article in English | MEDLINE | ID: mdl-30448175

ABSTRACT

Inflammation comprises an innate immune response, and is mainly induced by macrophages to protect the host from pathogens and mechanical injuries. The p38 mitogen-activated protein kinase (MAPK) pathway is a key regulator of inflammatory responses in macrophages. Here, we investigated the anti-inflammatory effects of thioredoxin-interacting protein-derived peptide (TN13) in macrophages in vitro and in vivo. Human immunodeficiency virus (HIV) trans-activator protein (TAT)-conjugated TN13 (TAT-TN13) was found to penetrate RAW 264.7 cells and decrease p38 MAPK activation in a dose-dependent manner. We also showed that TAT-TN13 could significantly inhibit lipopolysaccharide (LPS)-induced expression of macrophage activation-related receptors including CD80, CD86, and MHC II, as well as the transcriptional activation of nuclear factor-kappa B (NF-κB) and activator protein-1 (AP-1) in RAW 264.7 cells and primary mouse splenic macrophages. Furthermore, TAT-TN13 decreased the LPS-induced production of proinflammatory cytokines and mediators such as tumor necrosis factor α (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), nitric oxide (NO), inducible NO synthase (iNOS), and cyclooxygenase 2 (COX-2) in RAW 264.7 cells and mice. These results indicate that TAT-TN13 can inhibit macrophage-derived inflammation by inhibiting p38 MAPK activity and might represent a potential novel drug for the treatment of inflammation-related diseases.


Subject(s)
Inflammation/enzymology , Inflammation/pathology , MAP Kinase Signaling System/drug effects , Peptides/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cytokines/blood , Inflammation/blood , Inflammation Mediators/metabolism , Lipopolysaccharides , Macrophage Activation/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RAW 264.7 Cells , Transcription Factor AP-1/metabolism
5.
Biochem Biophys Res Commun ; 506(1): 33-40, 2018 11 17.
Article in English | MEDLINE | ID: mdl-30336978

ABSTRACT

Overcoming drug resistance is one of key issues in treating refractory acute myeloid leukemia (AML). The Toll-like receptor 4 (TLR4) signaling pathway is involved in many aspects of biological functions of AML cells, including the regulation of pro-inflammatory cytokine products, myeloid differentiation, and survival of AML cells. Thus, targeting TLR4 of AML patients for therapeutic purposes should be carefully addressed. In this regard, we investigated the possible role of TLR4 as a regulatory factor against fludarabine (FA) cytotoxicity activity. Here, we identified the differential expression of TLR4 and CD14 receptors in AML cell lines and examined their relationship to FA sensitivity. We found that the stimulation of TLR4 with lipopolysaccharide (LPS) in a TLR4-expressing cell line, THP-1, increased cell viability under FA treatment condition and showed that TLR4 stimulation overcame FA sensitivity through the activation of NF-κB, which subsequently upregulated several anti-apoptotic genes. The inhibition of TLR4/NF-κB signaling could partially or completely reverse LPS-induced cell survival under FA treatment conditions. Interestingly, we found that the expression of thioredoxin-interacting protein (TXNIP), a well-known tumor suppressor, was induced by FA treatment; however, it was suppressed by LPS treatment. Furthermore, the expression level of TXNIP was critical for FA-induced cytotoxicity or LPS-induced FA resistance of THP-1 cells. Our data suggest that TXNIP plays an important role in FA-induced cytotoxicity and TLR4/NF-κB-mediated FA resistance of AML cells. Therefore, TXNIP may be a potential therapeutic target for AML treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/genetics , Gene Expression Regulation, Leukemic , NF-kappa B/genetics , Toll-Like Receptor 4/genetics , Vidarabine/analogs & derivatives , Apoptosis/drug effects , Carrier Proteins/immunology , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/genetics , HL-60 Cells , Humans , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/pharmacology , NF-kappa B/immunology , Signal Transduction , THP-1 Cells , Toll-Like Receptor 4/immunology , Vidarabine/pharmacology
6.
Aging Cell ; 17(6): e12836, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30168649

ABSTRACT

Aging is associated with an inevitable and universal loss of cell homeostasis and restricts an organism's lifespan by an increased susceptibility to diseases and tissue degeneration. The glucose uptake associated with producing energy for cell survival is one of the major causes of ROS production under physiological conditions. However, the overall mechanisms by which glucose uptake results in cellular senescence remain mysterious. In this study, we found that TXNIP deficiency accelerated the senescent phenotypes of MEF cells under high glucose condition. TXNIP-/- MEF cells showed greater induced glucose uptake and ROS levels than wild-type cells, and N-acetylcysteine (NAC) treatment rescued the cellular senescence of TXNIP-/- MEF cells. Interestingly, TXNIP-/- MEF cells showed continuous activation of AKT during long-term subculture, and AKT signaling inhibition completely blocked the cellular senescence of TXNIP-/- MEF cells. In addition, we found that TXNIP interacted with AKT via the PH domain of AKT, and their interaction was increased by high glucose or H2 O2 treatment. The inhibition of AKT activity by TXNIP was confirmed using western blotting and an in vitro kinase assay. TXNIP deficiency in type 1 diabetes mice (Akita) efficiently decreased the blood glucose levels and finally increased mouse survival. However, in normal mice, TXNIP deficiency induced metabolic aging of mice and cellular senescence of kidney cells by inducing AKT activity and aging-associated gene expression. Altogether, these results suggest that TXNIP regulates cellular senescence by inhibiting AKT pathways via a direct interaction under conditions of glucose-derived metabolic stress.


Subject(s)
Carrier Proteins/metabolism , Cellular Senescence , Glucose/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Stress, Physiological , Thioredoxins/metabolism , Animals , Cellular Senescence/drug effects , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Kidney/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Binding/drug effects , Stress, Physiological/drug effects
7.
Sci Rep ; 7: 46153, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28383049

ABSTRACT

Suppressor of cytokine signaling (SOCS) proteins are negative regulators of cytokine responses. Although recent reports have shown regulatory roles for SOCS proteins in innate and adaptive immunity, their roles in natural killer (NK) cell development are largely unknown. Here, we show that SOCS2 is involved in NK cell development. SOCS2-/- mice showed a high frequency of NK cells in the bone marrow and spleen. Knockdown of SOCS2 was associated with enhanced differentiation of NK cells in vitro, and the transplantation of hematopoietic stem cells (HSCs) into congenic mice resulted in enhanced differentiation in SOCS2-/- HSCs. We found that SOCS2 could inhibit Janus kinase 2 (JAK2) activity and JAK2-STAT5 signaling pathways via direct interaction with JAK2. Furthermore, SOCS2-/- mice showed a reduction in lung metastases and an increase in survival following melanoma challenge. Overall, our findings suggest that SOCS2 negatively regulates the development of NK cells by inhibiting JAK2 activity via direct interaction.


Subject(s)
Cell Differentiation , Janus Kinase 2/antagonists & inhibitors , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Biomarkers/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Hematopoietic Stem Cells/metabolism , Interleukin-15/metabolism , Janus Kinase 2/metabolism , Mice, Inbred C57BL , Protein Binding , STAT5 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/deficiency
8.
Cell Signal ; 34: 110-120, 2017 06.
Article in English | MEDLINE | ID: mdl-28323005

ABSTRACT

The nuclear factor kappa B (NF-κB) pathway is pivotal in controlling survival and apoptosis of cancer cells. Macrophage migration inhibitory factor (MIF), a cytokine that regulates the immune response and tumorigenesis under inflammatory conditions, is upregulated in various tumors. However, the intracellular functions of MIF are unclear. In this study, we found that MIF directly interacted with thioredoxin-interacting protein (TXNIP), a tumor suppressor and known inhibitor of NF-κB activity, and MIF significantly induced NF-κB activation. MIF competed with TXNIP for NF-κB activation, and the intracellular MIF induced NF-κB target genes, including c-IAP2, Bcl-xL, ICAM-1, MMP2 and uPA, by inhibiting the interactions between TXNIP and HDACs or p65. Furthermore, we identified the interaction motifs between MIF and TXNIP via site-directed mutagenesis of their cysteine (Cys) residues. Cys57 and Cys81 of MIF and Cys36 and Cys120 of TXNIP were responsible for the interaction. MIF reversed the TXNIP-induced suppression of cell proliferation and migration. Overall, we suggest that MIF induces NF-κB activity by counter acting the inhibitory effect of TXNIP on the NF-κB pathway via direct interaction with TXNIP. These findings reveal a novel intracellular function of MIF in the progression of cancer.


Subject(s)
Carrier Proteins/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , NF-kappa B/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Proliferation/drug effects , Gene Expression/drug effects , HEK293 Cells , HeLa Cells , Humans , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides/toxicity , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/genetics , Matrix Metalloproteinase 2/metabolism , Mutagenesis, Site-Directed , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology , bcl-X Protein/metabolism
9.
Nat Commun ; 7: 13674, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27929088

ABSTRACT

Ageing is a natural process in living organisms throughout their lifetime, and most elderly people suffer from ageing-associated diseases. One suggested way to tackle such diseases is to rejuvenate stem cells, which also undergo ageing. Here we report that the thioredoxin-interacting protein (TXNIP)-p38 mitogen-activated protein kinase (p38) axis regulates the ageing of haematopoietic stem cells (HSCs), by causing a higher frequency of long-term HSCs, lineage skewing, a decrease in engraftment, an increase in reactive oxygen species and loss of Cdc42 polarity. TXNIP inhibits p38 activity via direct interaction in HSCs. Furthermore, cell-penetrating peptide (CPP)-conjugated peptide derived from the TXNIP-p38 interaction motif inhibits p38 activity via this docking interaction. This peptide dramatically rejuvenates aged HSCs in vitro and in vivo. Our findings suggest that the TXNIP-p38 axis acts as a regulatory mechanism in HSC ageing and indicate the potent therapeutic potential of using CPP-conjugated peptide to rejuvenate aged HSCs.


Subject(s)
Carrier Proteins/physiology , Cellular Senescence , Hematopoietic Stem Cells/physiology , MAP Kinase Signaling System , Thioredoxins/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Female , Male , Mice
10.
PLoS Pathog ; 9(10): e1003646, 2013.
Article in English | MEDLINE | ID: mdl-24098117

ABSTRACT

Thioredoxin-interacting protein (TXNIP) has multiple functions, including tumor suppression and involvement in cell proliferation and apoptosis. However, its role in the inflammatory process remains unclear. In this report, we demonstrate that Txnip⁻/⁻ mice are significantly more susceptible to lipopolysaccharide (LPS)-induced endotoxic shock. In response to LPS, Txnip⁻/⁻ macrophages produced significantly higher levels of nitric oxide (NO) and inducible nitric oxide synthase (iNOS), and an iNOS inhibitor rescued Txnip⁻/⁻ mice from endotoxic shock-induced death, demonstrating that NO is a major factor in TXNIP-mediated endotoxic shock. This susceptibility phenotype of Txnip⁻/⁻ mice occurred despite reduced IL-1ß secretion due to increased S-nitrosylation of NLRP3 compared to wild-type controls. Taken together, these data demonstrate that TXNIP is a novel molecule that links NO synthesis and NLRP3 inflammasome activation during endotoxic shock.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Shock, Septic/metabolism , Thioredoxins/metabolism , Animals , Carrier Proteins/genetics , Inflammasomes/genetics , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nitric Oxide/genetics , Nitric Oxide Synthase Type II/genetics , Shock, Septic/chemically induced , Shock, Septic/genetics , Thioredoxins/genetics
11.
Cell Metab ; 18(1): 75-85, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23823478

ABSTRACT

Reactive oxygen species (ROS) are critical determinants of the fate of hematopoietic stem cells (HSCs) and hematopoiesis. Thioredoxin-interacting protein (TXNIP), which is induced by oxidative stress, is a known regulator of intracellular ROS. Txnip(-/-) old mice exhibited elevated ROS levels in hematopoietic cells and showed a reduction in hematopoietic cell population. Loss of TXNIP led to a dramatic reduction of mouse survival under oxidative stress. TXNIP directly regulated p53 protein by interfering with p53- mouse double minute 2 (MDM2) interactions and increasing p53 transcriptional activity. Txnip(-/-) mice showed downregulation of the antioxidant genes induced by p53. Introduction of TXNIP or p53 into Txnip(-/-) bone marrow cells rescued the HSC frequency and greatly increased survival in mice following oxidative stress. Overall, these data indicate that TXNIP is a regulator of p53 and plays a pivotal role in the maintenance of the hematopoietic cells by regulating intracellular ROS during oxidative stress.


Subject(s)
Carrier Proteins/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Oxidative Stress/physiology , Signal Transduction/physiology , Thioredoxins/physiology , Tumor Suppressor Protein p53/physiology , Animals , Antioxidants/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Bone Marrow Transplantation , Carrier Proteins/genetics , Cells, Cultured , Hematopoietic Stem Cells/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Proto-Oncogene Proteins c-mdm2/physiology , Reactive Oxygen Species/metabolism , Thioredoxins/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
12.
Biochem Biophys Res Commun ; 425(2): 340-7, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22842576

ABSTRACT

Natural killer (NK) cells are a subset of lymphocytes crucial for innate and adaptive immune responses. Here we show a stimulatory effect of cryptotanshinone (CTS) and tanshinone IIA (TS), isolated from Salvia miltiorrhiza Bunge, on the differentiation of NK cells. In the presence of IL-15, tanshinones increased NK cell maturation, NK cell differentiation and the expression of several transcription factors, including Id2, GATA3, T-bet, and Ets-1. Additionally, tanshinones increased p38 MAPK phosphorylation during NK cell differentiation. Furthermore, the p38 inhibitor SB203580 blocked the developmental effects of the tanshinones and suppressed Id2, T-bet, and Ets-1 expression during NK cell differentiation. These results suggest that tanshinones significantly increased IL-15-induced NK cell differentiation via enhancing the p38 phosphorylation and the expression of transcription factors.


Subject(s)
Abietanes/pharmacology , Cell Differentiation/drug effects , Drugs, Chinese Herbal/pharmacology , Interleukin-15/pharmacology , Killer Cells, Natural/drug effects , Lymphocyte Subsets/drug effects , Phenanthrenes/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Differentiation/immunology , Imidazoles/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Phosphorylation , Pyridines/pharmacology , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
13.
J Immunol ; 185(2): 917-28, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20543098

ABSTRACT

NK cells are capable of killing virus-infected or tumor cells and producing IFN-gamma. Resting NK cells, however, have only minimal cytolytic activity and secrete a low level of IFN-gamma. The cytokine IL-15 can promote the expression of effector functions by resting NK cells. In this study, we demonstrate that suppressor of cytokine signaling 2 (SOCS2) has a novel role in IL-15-primed human NK cell function. SOCS2 expression was upregulated in NK cells following stimulation with IL-15. During IL-15-mediated NK cell priming, SOCS2 interacted with phosphorylated proline-rich tyrosine kinase 2 (Pyk2) at tyrosine 402 (p-Pyk2(Tyr402)) and induced the proteasome-mediated degradation of p-Pyk2(Tyr402) via ubiquitination. Knockdown of SOCS2 resulted in the accumulation of p-Pyk2(Tyr402) and blocked NK cell effector functions. In addition, NK cell cytolytic activity and IFN-gamma production were inhibited by overexpression of the wild-type of Pyk2 but not by the overexpression of tyrosine 402 mutant of Pyk2. These results suggest that SOCS2 regulates human NK cell effector functions via control of phosphorylated Pyk2 depending on IL-15 existence.


Subject(s)
Focal Adhesion Kinase 2/metabolism , Interleukin-15/pharmacology , Killer Cells, Natural/drug effects , Suppressor of Cytokine Signaling Proteins/metabolism , Blotting, Western , Cell Differentiation/drug effects , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Focal Adhesion Kinase 2/genetics , Humans , Infant, Newborn , Interferon-gamma/metabolism , Jurkat Cells , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mutation , Phosphorylation/drug effects , Protein Binding , RNA Interference , Receptors, Interleukin-15/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/genetics , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...