Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Leukemia ; 38(7): 1570-1580, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38454120

ABSTRACT

Although Bruton's tyrosine kinase (BTK) inhibitors (BTKi) have significantly improved patient prognosis, mantle cell lymphoma (MCL) is still considered incurable due to primary and acquired resistance. We have recently shown that aberrant expression of the Src-family tyrosine kinase hematopoietic cell kinase (HCK) in MCL correlates with poor prognosis, and that genetic HCK perturbation impairs growth and integrin-mediated adhesion of MCL cells. Here, we show that KIN-8194, a dual inhibitor of BTK and HCK with in vivo activity against Myd88-L265P-driven diffuse large B-cell lymphoma and Waldenström Macroglobulinemia, has a potent growth inhibitory effect in MCL cell lines and primary MCL cells, irrespective of their sensitivity to BTKi (ibrutinib and acalabrutinib). In BTKi-resistant cells this is mediated by inhibition of HCK, which results in repression of AKT-S6 signaling. In addition, KIN-8194 inhibits integrin-mediated adhesion of BTKi-sensitive and insensitive MCL cells to fibronectin and stromal cells in an HCK-dependent manner. Finally, we show that MCL cells with acquired BTKi resistance retain their sensitivity to KIN-8194. Taken together, our data demonstrate that KIN-8194 inhibits growth and integrin-mediated adhesion of BTKi-sensitive MCL cells, as well as MCL cells with primary or acquired BTKi resistance. This renders KIN-8194 a promising novel treatment for MCL patients.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Cell Adhesion , Cell Proliferation , Drug Resistance, Neoplasm , Integrins , Lymphoma, Mantle-Cell , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-hck , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/pathology , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/genetics , Humans , Cell Adhesion/drug effects , Drug Resistance, Neoplasm/drug effects , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Proto-Oncogene Proteins c-hck/metabolism , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/antagonists & inhibitors , Integrins/metabolism , Cell Proliferation/drug effects , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Pyrimidines/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Signal Transduction/drug effects
2.
Pathol Res Pract ; 247: 154534, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37201466

ABSTRACT

Laryngeal squamous cell carcinoma (LSCC) is the most lethal cancer in head and neck tumors. Although hematopoietic cell kinase (HCK) has been proven to be an oncogene in several solid tumors, its roles in LSCC remain obscure. This is the first study to evaluate the clinical value of HCK in LSCC, with the aim of exploring its expression status and potential molecular mechanisms underlying LSCC. LSCC tissue-derived gene chips and RNA-seq data were collected for a quantitive integration of HCK mRNA expression level. To confirm the protein expression level of HCK, a total of 82 LSCC tissue specimens and 56 non-tumor laryngeal epithelial controls were collected for in-house tissue microarrays and immunohistochemical staining. Kaplan-Meier curves were generated to determine the ability of HCK in predicting overall survival, progress-free survival, and disease-free survival of LSCC patients. LSCC overexpressed genes and HCK co-expressed genes were intersected to preliminarily explore the enriched signaling pathways of HCK. It was noticed that HCK mRNA was markedly overexpressed in 323 LSCC tissues compared with 196 non-LSCC controls (standardized mean difference = 0.81, p < 0.0001). Upregulated HCK mRNA displayed a moderate discriminatory ability between LSCC tissues and non-tumor laryngeal epithelial controls (area under the curve = 0.78, sensitivity = 0.76, specificity = 0.68). The higher expression level of HCK mRNA could predict worse overall survival and disease-free survival for LSCC patients (p = 0.041 and p = 0.013). Lastly, upregulated co-expression genes of HCK were significantly enriched in leukocyte cell-cell adhesion, secretory granule membrane, and extracellular matrix structural constituent. Immune-related pathways were the predominantly activated signals, such as cytokine-cytokine receptor interaction, Th17 cell differentiation, and Toll-like receptor signaling pathway. In conclusion, HCK was upregulated in LSCC tissues and could be utilized as a risk predictor. HCK may promote the development of LSCC by disturbing immune signaling pathways.


Subject(s)
Laryngeal Neoplasms , Squamous Cell Carcinoma of Head and Neck , Humans , Gene Expression Regulation, Neoplastic/genetics , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Prognosis , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/metabolism , RNA, Messenger/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology
3.
Cell Rep ; 41(2): 111479, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36223746

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with a low 5-year survival rate and is associated with poor response to therapy. Elevated expression of the myeloid-specific hematopoietic cell kinase (HCK) is observed in PDAC and correlates with reduced patient survival. To determine whether aberrant HCK signaling in myeloid cells is involved in PDAC growth and metastasis, we established orthotopic and intrasplenic PDAC tumors in wild-type and HCK knockout mice. Genetic ablation of HCK impaired PDAC growth and metastasis by inducing an immune-stimulatory endotype in myeloid cells, which in turn reduced the desmoplastic microenvironment and enhanced cytotoxic effector cell infiltration. Consequently, genetic ablation or therapeutic inhibition of HCK minimized metastatic spread, enhanced the efficacy of chemotherapy, and overcame resistance to anti-PD1, anti-CTLA4, or stimulatory anti-CD40 immunotherapy. Our results provide strong rationale for HCK to be developed as a therapeutic target to improve the response of PDAC to chemo- and immunotherapy.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Proto-Oncogene Proteins c-hck , Animals , Carcinoma, Pancreatic Ductal/genetics , Mice , Myeloid Cells/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-hck/genetics , Tumor Microenvironment , Pancreatic Neoplasms
4.
Dis Markers ; 2022: 3199589, 2022.
Article in English | MEDLINE | ID: mdl-35280440

ABSTRACT

Background: The tumor microenvironment (TME) plays a significant role in the progression and prognosis of acute myeloid leukemia (AML). This study is aimed at exploring TME-associated biomarkers and identify their potential mechanism in the microenvironment of AML. Method: In this study, the stromal, immune, and ESTIMATE scores of AML patients were evaluated with the ESTIMATE and CIBERSORT algorithms; then, the AML samples were divided into high- and low-score groups. We evaluated the association between clinicopathological characteristics, survival rate, and the stromal/immune/ESTIMATE scores. Furthermore, we identified TME-associated differentially expressed genes (DEGs) then carried out pathway enrichment analysis, protein-protein interaction (PPI) network, Cox regression analysis, and Kaplan-Meier survival analysis to select the most crucial genes. In addition, we further explored the potential mechanism of HCK in the AML microenvironment. Results: We identified 624 TME-associated DEGs and found that HCK was the most promising biomarker associated with AML. The results of the gene set enrichment analysis (GSEA) indicated that HCK was mainly involved in immune and inflammation-related signaling pathways. In addition, CIBERSORT analysis showed that HCK was closely related to tumor immune infiltration, with HCK expression associated with various infiltrating immune cells, including B cells, T cells, tumor-associated macrophages (TAM), NK cells, plasma cells, eosinophils, and neutrophils. Furthermore, HCK expression was closely related with ELN risk stratification in patients with AML. Conclusion: HCK could regulate immune cell infiltration in the microenvironment of AML and may act as a potential biomarker for the treatment and prognosis of AML patients.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins c-hck/genetics , Aged , Algorithms , Correlation of Data , Female , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Prognosis , Survival Rate
5.
J Allergy Clin Immunol ; 149(4): 1464-1472.e3, 2022 04.
Article in English | MEDLINE | ID: mdl-34536415

ABSTRACT

BACKGROUND: Inborn errors of immunity are genetic disorders characterized by various degrees of immune dysregulation that can manifest as immune deficiency, autoimmunity, or autoinflammation. The routine use of next-generation sequencing in the clinic has facilitated the identification of an ever-increasing number of inborn errors of immunity, revealing the roles of immunologically important genes in human pathologies. However, despite this progress, treatment is still extremely challenging. OBJECTIVE: We sought to report a new monogenic autoinflammatory disorder caused by a de novo activating mutation, p.Tyr515∗, in hematopoietic cell kinase (HCK). The disease is characterized by cutaneous vasculitis and chronic pulmonary inflammation that progresses to fibrosis. METHODS: Whole-exome sequencing, Sanger sequencing, mass spectrometry, and western blotting were performed to identify and characterize the pathogenic HCK mutation. Dysregulation of mutant HCK was confirmed ex vivo in primary cells and in vitro in transduced cell lines. RESULTS: Mutant HCK lacking the C-terminal inhibitory tyrosine Tyr522 exhibited increased kinase activity and enhanced myeloid cell priming, migration and effector functions, such as production of the inflammatory cytokines IL-1ß, IL-6, IL-8, and TNF-α, and production of reactive oxygen species. These aberrant functions were reflected by inflammatory leukocyte infiltration of the lungs and skin. Moreover, an overview of the clinical course of the disease, including therapies, provides evidence for the therapeutic efficacy of the Janus kinase 1/2 inhibitor ruxolitinib in inflammatory lung disease. CONCLUSIONS: We propose HCK-driven pulmonary and cutaneous vasculitis as a novel autoinflammatory disorder of inborn errors of immunity.


Subject(s)
Vasculitis , src-Family Kinases , Humans , Lung , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/metabolism , Vasculitis/genetics , Vasculitis/pathology , src-Family Kinases/genetics
6.
PLoS Pathog ; 17(11): e1009728, 2021 11.
Article in English | MEDLINE | ID: mdl-34780577

ABSTRACT

The accessory protein Nef of human and simian immunodeficiency viruses (HIV and SIV) is an important pathogenicity factor known to interact with cellular protein kinases and other signaling proteins. A canonical SH3 domain binding motif in Nef is required for most of these interactions. For example, HIV-1 Nef activates the tyrosine kinase Hck by tightly binding to its SH3 domain. An archetypal contact between a negatively charged SH3 residue and a highly conserved arginine in Nef (Arg77) plays a key role here. Combining structural analyses with functional assays, we here show that Nef proteins have also developed a distinct structural strategy-termed the "R-clamp"-that favors the formation of this salt bridge via buttressing Arg77. Comparison of evolutionarily diverse Nef proteins revealed that several distinct R-clamps have evolved that are functionally equivalent but differ in the side chain compositions of Nef residues 83 and 120. Whereas a similar R-clamp design is shared by Nef proteins of HIV-1 groups M, O, and P, as well as SIVgor, the Nef proteins of SIV from the Eastern chimpanzee subspecies (SIVcpzP.t.s.) exclusively utilize another type of R-clamp. By contrast, SIV of Central chimpanzees (SIVcpzP.t.t.) and HIV-1 group N strains show more heterogenous R-clamp design principles, including a non-functional evolutionary intermediate of the aforementioned two classes. These data add to our understanding of the structural basis of SH3 binding and kinase deregulation by Nef, and provide an interesting example of primate lentiviral protein evolution.


Subject(s)
Evolution, Molecular , HIV Infections/metabolism , Lentivirus/genetics , Proto-Oncogene Proteins c-hck/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , src Homology Domains , Amino Acid Sequence , Animals , HIV Infections/genetics , HIV Infections/virology , HIV-1/physiology , Humans , Proto-Oncogene Proteins c-hck/genetics , Sequence Homology, Amino Acid , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , nef Gene Products, Human Immunodeficiency Virus/genetics
7.
J Virol ; 95(17): e0047121, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34106001

ABSTRACT

Human immunodeficiency virus (HIV) or simian immunodeficiency virus (SIV) infection causes myelodysplasia, anemia, and accumulation of inflammatory monocytes (CD14+ CD16+) through largely unknown cellular and molecular pathways. The mouse cells thought to be equivalent to human CD14+ CD16+ cells are CD11b+ Gr1+ myeloid-derived suppressor cells (MDSC). We used HIV transgenic (Tg) mouse models to study MDSC, namely, CD4C/Nef Tg mice expressing nef in dendritic cells (DC), pDC, CD4+ T, and other mature and immature myeloid cells and CD11c/Nef Tg mice with a more restricted expression, mainly in DC and pDC. Both Tg strains showed expansion of granulocytic and CD11b+ Gr1low/int cells with MDSC characteristics. Fetal liver cell transplantation revealed that this expansion was stroma-independent and abrogated in mixed Tg/non-Tg 50% chimera. Tg bone marrow (BM) erythroid progenitors were decreased and myeloid precursors increased, suggesting an aberrant differentiation likely driving CD11b+ Gr1+ cell expansion, apparently cell autonomously in CD4C/Nef Tg mice and likely through a bystander effect in CD11c/Nef Tg mice. Hck was activated in Tg spleen, and Nef-mediated CD11b+ Gr1+ cell expansion was abrogated in Hck/Lyn-deficient Nef Tg mice, indicating a requirement of Hck/Lyn for this Nef function. IL-17 and granulocyte colony-stimulating factor (G-CSF) were elevated in Nef Tg mice. Increased G-CSF levels were normalized in Tg mice treated with anti-IL-17 antibodies. Therefore, Nef expression in myeloid precursors causes severe BM failure, apparently cell autonomously. More cell-restricted expression of Nef in DC and pDC appears sufficient to induce BM differentiation impairment, granulopoiesis, and expansion of MDSC at the expense of erythroid maturation, with IL-17→G-CSF as one likely bystander contributor. IMPORTANCE HIV-1 and SIV infection often lead to myelodysplasia, anemia, and accumulation of inflammatory monocytes (CD14+ CD16+), with the latter likely involved in neuroAIDS. We found that some transgenic (Tg) mouse models of AIDS also develop accumulation of mature and immature cells of the granulocytic lineage, decreased erythroid precursors, and expansion of MDSC (equivalent to human CD14+ CD16+ cells). We identified Nef as being responsible for these phenotypes, and its expression in mouse DC appears sufficient for their development through a bystander mechanism. Nef expression in myeloid progenitors may also favor myeloid cell expansion, likely in a cell-autonomous way. Hck/Lyn is required for the Nef-mediated accumulation of myeloid cells. Finally, we identified G-CSF under the control of IL-17 as one bystander mediator of MDSC expansion. Our findings provide a framework to determine whether the Nef>Hck/Lyn>IL-17>G-CSF pathway is involved in human AIDS and whether it represents a valid therapeutic target.


Subject(s)
Granulocyte Colony-Stimulating Factor/metabolism , HIV Infections/immunology , Interleukin-17/metabolism , Myeloid-Derived Suppressor Cells/immunology , Proto-Oncogene Proteins c-hck/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , src-Family Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Female , Granulocyte Colony-Stimulating Factor/genetics , Granulocytes/immunology , Granulocytes/metabolism , Granulocytes/pathology , HIV Infections/metabolism , HIV Infections/virology , HIV-1/immunology , Humans , Interleukin-17/genetics , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/virology , Proto-Oncogene Proteins c-hck/genetics , nef Gene Products, Human Immunodeficiency Virus/genetics , src-Family Kinases/genetics
8.
J Exp Clin Cancer Res ; 40(1): 210, 2021 Jun 24.
Article in English | MEDLINE | ID: mdl-34167558

ABSTRACT

BACKGROUND: Leukaemia stem cells (LSCs) are responsible for the initiation, maintenance, and recurrence of acute myeloid leukaemia (AML), an aggressive haematological malignancy associated with drug resistance and relapse. Identifying therapeutic LSC targets is critical to curing AML. METHODS: Bioinformatics databases were used to identify therapeutic LSC targets. The conditional knockout mice were used to analyse the role of HCK in leukaemogenesis or normal haematopoiesis. Colony-forming assays, cell counting, and flow cytometry were used to detect the viability and function of leukaemia cells. RT-PCR, western blotting, and RNA sequencing were used to detect mRNA and protein expression. RESULT: HCK is expressed at higher levels in LSCs than in haematopoietic stem cells (HSCs), and high HCK levels are correlated with reduced survival time in AML patients. Knockdown of HCK leads to cell cycle arrest, which results in a dramatic decrease in the proliferation and colony formation in human AML cell lines. Moreover, HCK is required for leukemogenesis and leukaemia maintenance in vivo and in vitro. HCK is necessary for the self-renewal of LSCs during serial transplantation and limiting dilution assay. The phenotypes resulting from HCK deficiency can be rescued by CDK6 overexpression in the human cell line. RNA sequencing and gene expression have demonstrated that HCK may sustain cell cycle entry and maintain the self-renewal ability of LSCs through activating the ERK1/2-c-Myc-CDK6 signalling axis. In contrast, HCK deletion does not affect normal haematopoiesis or haematopoietic reconstruction in mice. CONCLUSIONS: HCK maintains the self-renewal of leukaemia stem cells via CDK6 in AML and may be an ideal therapeutic target for eradicating LSCs without influencing normal haematopoiesis.


Subject(s)
Cyclin-Dependent Kinase 6/metabolism , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Self Renewal , Databases, Genetic , Gene Deletion , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Humans , K562 Cells , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Knockout , Neoplasm Transplantation
9.
Microvasc Res ; 136: 104172, 2021 07.
Article in English | MEDLINE | ID: mdl-33894273

ABSTRACT

BACKGROUND: VSMC proliferation and migration pathways play important roles in plaque formation in the vessel stenosis and re-stenosis processes. The microRNAs affect the expression of many genes that regulate these cellular processes. The aim of this study was to investigate the effects of miR-181b, miR-204, and miR-599 on the gene and protein expression levels of hematopoietic cell kinase (HCK) in VSMCs. METHODS: miR-181b, miR-204 were predicted for the suppression of HCK in the chemokine signaling pathway using bioinformatics tools. Then, the VSMCs were transfected by PEI-containing microRNAs. The HCK gene and protein expression levels were evaluated using RT-qPCR and Western blotting techniques, respectively. Moreover, the cellular proliferation and migration were evaluated by MTT and scratch assay methods. RESULTS: The miR-181b and miR-204 decreased significantly the HCK gene and (total and phosphorylated) protein expression levels. Also, the miR-599 did not show any significant effects on the HCK gene and protein levels. The data also showed that miR-181b, miR-204, and miR-599 prevent significantly the proliferation and migration of VSMCs. CONCLUSION: The downregulation of HCK by miR-181b and miR-204 suppressed the VSMC proliferation and migration.


Subject(s)
Cell Movement , Cell Proliferation , MicroRNAs/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Proto-Oncogene Proteins c-hck/metabolism , Cells, Cultured , Down-Regulation , Humans , MicroRNAs/genetics , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/ultrastructure , Proto-Oncogene Proteins c-hck/genetics , Signal Transduction
10.
J Biol Chem ; 296: 100449, 2021.
Article in English | MEDLINE | ID: mdl-33617879

ABSTRACT

Hck, a Src family nonreceptor tyrosine kinase (SFK), has recently been established as an attractive pharmacological target to improve pulmonary function in COVID-19 patients. Hck inhibitors are also well known for their regulatory role in various malignancies and autoimmune diseases. Curcumin has been previously identified as an excellent DYRK-2 inhibitor, but curcumin's fate is tainted by its instability in the cellular environment. Besides, small molecules targeting the inactive states of a kinase are desirable to reduce promiscuity. Here, we show that functionalization of the 4-arylidene position of the fluorescent curcumin scaffold with an aryl nitrogen mustard provides a stable Hck inhibitor (Kd = 50 ± 10 nM). The mustard curcumin derivative preferentially interacts with the inactive conformation of Hck, similar to type-II kinase inhibitors that are less promiscuous. Moreover, the lead compound showed no inhibitory effect on three other kinases (DYRK2, Src, and Abl). We demonstrate that the cytotoxicity may be mediated via inhibition of the SFK signaling pathway in triple-negative breast cancer and murine macrophage cells. Our data suggest that curcumin is a modifiable fluorescent scaffold to develop selective kinase inhibitors by remodeling its target affinity and cellular stability.


Subject(s)
Curcumin/pharmacology , Drug Design , Epithelial Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-hck/antagonists & inhibitors , Animals , Cell Line, Tumor , Cloning, Molecular , Curcumin/analogs & derivatives , Curcumin/chemical synthesis , Drug Stability , Epithelial Cells/enzymology , Epithelial Cells/pathology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , HT29 Cells , Humans , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-hck/chemistry , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/metabolism , RAW 264.7 Cells , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , src-Family Kinases/genetics , src-Family Kinases/metabolism , Dyrk Kinases
11.
Leukemia ; 35(3): 881-886, 2021 03.
Article in English | MEDLINE | ID: mdl-32591642

ABSTRACT

Mantle cell lymphoma (MCL) is an aggressive non-Hodgkin lymphoma subtype arising from naïve B cells. Although novel therapeutics have improved patient prognosis, drug resistance remains a key problem. Here, we show that the SRC-family tyrosine kinase hematopoietic cell kinase (HCK), which is primarily expressed in the hematopoietic lineage but not in mature B cells, is aberrantly expressed in MCL, and that high expression of HCK is associated with inferior prognosis of MCL patients. HCK expression is controlled by the toll-like receptor (TLR) adaptor protein MYD88 and can be enhanced by TLR agonists in MCL cell lines and primary MCL. In line with this, primary MCL with high HCK expression are enriched for a TLR-signaling pathway gene set. Silencing of HCK expression results in cell cycle arrest and apoptosis. Furthermore, HCK controls integrin-mediated adhesion of MCL cells to extracellular matrix and stromal cells. Taken together, our data indicate that TLR/MYD88-controlled aberrant expression of HCK plays a critical role in MCL proliferation and survival as well as in retention of the malignant cells in the growth- and survival-supporting lymphoid organ microenvironment, thereby contributing to lymphomagenesis. These novel insights provide a strong rationale for therapeutic targeting of HCK in MCL.


Subject(s)
Biomarkers, Tumor/metabolism , Lymphoma, Mantle-Cell/pathology , Proto-Oncogene Proteins c-hck/metabolism , Tumor Microenvironment , Biomarkers, Tumor/genetics , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/metabolism , Prognosis , Proto-Oncogene Proteins c-hck/genetics , Signal Transduction
12.
Int J Med Sci ; 17(17): 2773-2789, 2020.
Article in English | MEDLINE | ID: mdl-33162805

ABSTRACT

The role of HCK expression in the prognosis of breast cancer patients is unclear. Thus, this study aimed to explore the clinical implications of HCK expression in breast cancer. We assessed HCK expression and genetic variations in breast cancer using Oncomine, GEPIA, UALCAN, and cBioPortal databases. Then, immunochemistry was used to analyze HCK expression in breast cancer specimens, non-cancer tissues and metastatic cancer tissues. Consequently, we evaluated the effect of HCK expression on survival outcomes set as disease-free survival (DFS) and overall survival (OS). Finally, STRING, Coexpedia, and TISIDB database were explored to identify the molecular functions and regulation pathways of HCK. We found that breast cancer tissues have more HCK mRNA transcripts than non-cancer tissues. Patients with HCK expression had significantly shorter DFS and OS. The ratio of HCK expression was higher in cancer tissues than in non-cancer tissues. These results from STRING database, FunRich software, and TISIDB database showed that HCK was involved in mediating multiple biological processes including immune response-regulating signaling pathway, cell growth and maintenance through multiple signaling pathways including epithelial to mesenchymal transition, PI3K/AKT signaling pathway, and focal adhesion. Overall, HCK may be an oncogene in the development of breast cancer and thus may as a novel biomarker and therapeutic target for breast cancer.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/mortality , Carcinoma, Ductal, Breast/mortality , Neoplasm Recurrence, Local/epidemiology , Proto-Oncogene Proteins c-hck/genetics , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/blood , Breast/pathology , Breast/surgery , Breast Neoplasms/blood , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/blood , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/therapy , Cell Line, Tumor , Chemotherapy, Adjuvant/methods , Disease-Free Survival , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Focal Adhesions/drug effects , Focal Adhesions/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunochemistry , Mastectomy , Middle Aged , Molecular Targeted Therapy/methods , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Phosphatidylinositol 3-Kinases , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-hck/antagonists & inhibitors , Proto-Oncogene Proteins c-hck/blood , Risk Assessment/methods , Signal Transduction/drug effects , Signal Transduction/genetics
13.
Int J Mol Sci ; 21(22)2020 Nov 21.
Article in English | MEDLINE | ID: mdl-33233470

ABSTRACT

Kinase drug discovery represents an active area of therapeutic research, with previous pharmaceutical success improving patient outcomes across a wide variety of human diseases. In pancreatic ductal adenocarcinoma (PDAC), innovative pharmaceutical strategies such as kinase targeting have been unable to appreciably increase patient survival. This may be due, in part, to unchecked desmoplastic reactions to pancreatic tumors. Desmoplastic stroma enhances tumor development and progression while simultaneously restricting drug delivery to the tumor cells it protects. Emerging evidence indicates that many of the pathologic fibrotic processes directly or indirectly supporting desmoplasia may be driven by targetable protein tyrosine kinases such as Fyn-related kinase (FRK); B lymphoid kinase (BLK); hemopoietic cell kinase (HCK); ABL proto-oncogene 2 kinase (ABL2); discoidin domain receptor 1 kinase (DDR1); Lck/Yes-related novel kinase (LYN); ephrin receptor A8 kinase (EPHA8); FYN proto-oncogene kinase (FYN); lymphocyte cell-specific kinase (LCK); tec protein kinase (TEC). Herein, we review literature related to these kinases and posit signaling networks, mechanisms, and biochemical relationships by which this group may contribute to PDAC tumor growth and desmoplasia.


Subject(s)
Adenocarcinoma/genetics , Desmoplastic Small Round Cell Tumor/genetics , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/pathology , Desmoplastic Small Round Cell Tumor/pathology , Discoidin Domain Receptor 1/genetics , Disease Progression , Humans , Pancreatic Neoplasms/pathology , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-hck/genetics , Signal Transduction , src-Family Kinases/genetics
14.
Biosci Rep ; 40(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32484210

ABSTRACT

The hematopoietic cell kinase (HCK), a member of the Src family protein-tyrosine kinases (SFKs), is primarily expressed in cells of the myeloid and B lymphocyte lineages. Nevertheless, the roles of HCK in glioblastoma (GBM) remain to be examined. Thus, we aimed to investigate the effects of HCK on GBM development both in vitro and in vivo, as well as the underlying mechanism. The present study found that HCK was highly expressed in both tumor tissues from patients with GBM and cancer cell lines. HCK enhanced cell viability, proliferation, and migration, and induced cell apoptosis in vitro. Tumor xenografts results also demonstrated that HCK knockdown significantly inhibited tumor growth. Interestingly, gene set enrichment analysis (GSEA) showed HCK was closed associated with epithelial mesenchymal transition (EMT) and TGFß signaling in GBM. In addition, we also found that HCK accentuates TGFß-induced EMT, suggesting silencing HCK inhibited EMT through the inactivation of Smad signaling pathway. In conclusion, our findings indicated that HCK is involved in GBM progression via mediating EMT process, and may be served as a promising therapeutic target for GBM.


Subject(s)
Brain Neoplasms/enzymology , Epithelial-Mesenchymal Transition , Glioblastoma/enzymology , Proto-Oncogene Proteins c-hck/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Proto-Oncogene Proteins c-hck/genetics , Signal Transduction , Tumor Burden
15.
Biochim Biophys Acta Gen Subj ; 1864(7): 129604, 2020 07.
Article in English | MEDLINE | ID: mdl-32224253

ABSTRACT

A combination of small angle X-ray scattering (SAXS) and molecular dynamics (MD) simulations based on a coarse grained model is used to examine the effect of glycine substitutions in the short connector between the SH3 and SH2 domains of Hck, a member of the Src-family kinases. It has been shown previously that the activity of cSrc kinase is upregulated by substitution of 3 residues by glycine in the SH3-SH2 connector. Here, analysis of SAXS data indicates that the population of Hck in the disassembled state increases from 25% in the wild type kinase to 76% in the glycine mutant. This is consistent with the results of free energy perturbation calculations showing that the mutation in the connector shifts the equilibrium from the assembled to the disassembled state. This study supports the notion that the SH3-SH2 connector helps to regulate the activity of tyrosine kinases by shifting the population of the active state of the multidomain protein independent of C-terminal phosphorylation.


Subject(s)
Glycine/genetics , Protein Conformation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-hck/ultrastructure , Binding Sites , Crystallography, X-Ray , Molecular Dynamics Simulation , Phosphorylation/genetics , Protein-Tyrosine Kinases/ultrastructure , Proto-Oncogene Proteins c-hck/genetics , Scattering, Small Angle , X-Ray Diffraction , src Homology Domains/genetics
16.
Int J Biol Sci ; 16(9): 1507-1525, 2020.
Article in English | MEDLINE | ID: mdl-32226298

ABSTRACT

Protein-protein interactions are key to define the function of nucleotide binding domain (NBD) and leucine-rich repeat (LRR) family, pyrin domain (PYD)-containing protein 12 (NLRP12). cDNA encoding the human PYD + NBD of NLRP12 was used as bait in a yeast two-hybrid screen with a human leukocyte cDNA library as prey. Hematopoiesis cell kinase (HCK), a member of the c-SRC family of non-receptor tyrosine kinases, was among the top hits. The C-terminal 40 amino acids of HCK selectively bound to NLRP12's PYD + NBD, but not to that of NLRP3 and NLRP8. Amino acids F503, I506, Q507, L510, and D511 of HCK are critical for the binding of HCK's C-terminal 40 amino acids to NLRP12's PYD + NBD. Additionally, the C-terminal 30 amino acids of HCK are sufficient to bind to NLRP12's PYD + NBD, but not to its PYD alone nor to its NBD alone. In cell lines that express HCK endogenously, it was co- immunoprecipitated with stably expressed exogenous NLRP12. Also, NLRP12 co-immunoprecipitated and co-localized with HCK when both were overexpressed in 293T cells. In addition, in this overexpression system, steady-state NLRP12 protein expression levels significantly decreased when HCK was co-expressed. Bioinformatic analysis showed that HCK mRNA co-occurred with NLRP12 mRNA, but not with other NLRP mRNAs, in blood and marrow samples from acute myeloid leukemia (AML) patients. The mRNA of NLRP12 is also co-expressed with HCK in AML patient samples, and the levels of mRNA expression of each are correlated. Together these data suggest that NLRP12, through its binding to HCK, may have an effect on the pathogenesis of AML.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins c-hck/metabolism , Bone Marrow/metabolism , Cell Line , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-hck/blood , Proto-Oncogene Proteins c-hck/chemistry , Proto-Oncogene Proteins c-hck/genetics , Two-Hybrid System Techniques
17.
Mol Neurobiol ; 57(5): 2447-2460, 2020 May.
Article in English | MEDLINE | ID: mdl-32146679

ABSTRACT

Microglial dysregulation, pertaining to impairment in phagocytosis, clearance and containment of amyloid-ß (Aß), and activation of neuroinflammation, has been posited to contribute to the pathogenesis of Alzheimer's disease (AD). Detailed cellular mechanisms that are disrupted during the disease course to display such impairment in microglia, however, remain largely undetermined. We hypothesize that loss of hematopoietic cell kinase (HCK), a phagocytosis-regulating member of the Src family tyrosine kinases that mediate signals from triggering receptor expressed on myeloid cells 2 and other immunoreceptors, impairs microglial homeostasis and Aß clearance, leading to the accelerated buildup of Aß pathology and cognitive decline during the early stage of neuropathological development. To elucidate the pivotal role of HCK in AD, we generated a constitutive knockout of HCK in the Tg2576 mouse model of AD. We found that HCK deficiency accelerated cognitive decline along with elevated Aß level and plaque burden, attenuated microglial Aß phagocytosis, induced iNOS expression in microglial clusters, and reduced pre-synaptic protein at the hippocampal regions. Our findings substantiate that HCK plays a prominent role in regulating microglial neuroprotective functions and attenuating early AD neuropathology.


Subject(s)
Alzheimer Disease/enzymology , Microglia/enzymology , Proto-Oncogene Proteins c-hck/deficiency , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Disease Progression , Exploratory Behavior , Female , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/pathology , Morris Water Maze Test , Myeloid Cells/enzymology , Neuroimmunomodulation , Phagocytosis , Plaque, Amyloid , Proto-Oncogene Proteins c-hck/genetics , Recognition, Psychology
18.
Food Chem Toxicol ; 138: 111187, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32061728

ABSTRACT

The objective of this study was to determine the immunotoxic effects of deoxynivalenol (DON) in weaning piglets, and potential efficacy of a modified hydrated sodium calcium aluminosilicate (HSCAS) adsorbent to reduce DON toxicity. Four groups of 21-day-old male piglets (n = 7/group) were fed a control diet or diet containing 1.0 or 3.0 mg DON/kg, or 3.0 mg DON/kg plus 0.05% modified HSCAS for 4 weeks. Compared to the control, the DON diets decreased serum porcine circovirus antibody titer and the dermal hypersensitivity response to OVA at day 21 or 28. DON also induced focal necrosis and proliferation of cortical lymphocytes and apoptosis and increased the total antioxidant capacity and reduced glutathione, protein carbonyl concentrations in thymus. DON increased thymus mRNA, protein and (or) enzyme levels, cytokines (IL-6, IL-10, and TNF-α) and apoptosis-related genes (Caspase-3), while hematopoietic cell kinase (HCK) decreased. Intriguingly, the modified HSCAS alleviated the DON-induced changes on serum antibody titer, and thymic histopathology, apoptosis, redox status, inflammation and apoptosis signaling. In conclusion, these findings help to explain the toxic effects and mechanisms of DON and demonstrated the modified HSCAS adsorbent could be used to reduce the toxicity of DON in weaning piglets.


Subject(s)
Adaptive Immunity , Aluminum Silicates/chemistry , Trichothecenes/toxicity , Animal Feed/analysis , Animals , Animals, Newborn , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antioxidants/pharmacology , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Catalase/metabolism , Circovirus/immunology , Cytokines/blood , Diet/veterinary , Food Contamination/analysis , Glutathione/metabolism , Male , Malondialdehyde/metabolism , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/metabolism , Spleen/drug effects , Spleen/metabolism , Superoxide Dismutase/metabolism , Swine , Thymus Gland/drug effects , Thymus Gland/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Viral Vaccines/immunology , Weaning
19.
Mod Rheumatol ; 30(1): 85-92, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30486712

ABSTRACT

Objectives: To investigate the role of non-receptor tyrosine kinases (NRTKs) in inflammation-induced osteoclastogenesis.Methods: Microarray analyses of global mRNA expression during receptor activator of NF-κB ligand (RANKL) and RANKL plus tumor necrosis factor (TNF)-α-induced osteoclast differentiation were performed. The inhibitory effect on TNF-α-induced osteoclast differentiation of A-419259, a potent inhibitor of hematopoietic cell kinase (Hck), was examined. The in vivo therapeutic effect of A-419259 treatment on lipopolysaccharide (LPS)-induced inflammatory bone destruction was evaluated.Results: We confirmed that Hck expression was selectively increased among the NRTKs during the osteoclast differentiation induced by RANKL and TNF-α, but not by RANKL alone. RANKL and TNF-α-induced osteoclast differentiation and they were dose-dependently inhibited by A-419259 treatment through inhibition of the expression of key regulators of osteoclastogenesis, including Prdm1 and Nfatc1. Notably, LPS-induced inflammatory bone loss in murine calvarial bones was ameliorated by the administration of A-419259.Conclusions: Our results demonstrate that the administration of A-419259 is effective for the inhibition of osteoclast differentiation induced by TNF-α in the presence of RANKL. Therefore, an inhibitor of Hck may be useful as a potent anti-osteoclastogenic agent for the treatment of inflammatory bone destruction.


Subject(s)
Bone Resorption/genetics , Gene Expression Regulation , Inflammation/genetics , Osteoclasts/metabolism , Osteogenesis/drug effects , Proto-Oncogene Proteins c-hck/genetics , Pyrimidines/pharmacology , Pyrroles/pharmacology , Animals , Blotting, Western , Bone Resorption/drug therapy , Bone Resorption/metabolism , Cell Differentiation , Cells, Cultured , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred BALB C , Osteoclasts/drug effects , Osteoclasts/pathology , Proto-Oncogene Proteins c-hck/biosynthesis , RNA/genetics , src-Family Kinases
20.
PLoS One ; 14(12): e0225887, 2019.
Article in English | MEDLINE | ID: mdl-31790499

ABSTRACT

Unregulated protein-tyrosine kinase signaling is a common feature of AML, often involving mutations in Flt3 and overexpression of myeloid Src-family kinases (Hck, Fgr, Lyn). Here we show that high-level expression of these Src kinases predicts poor survival in a large cohort of AML patients. To test the therapeutic benefit of Flt3 and Src-family kinase inhibition, we used the pyrrolopyrimidine kinase inhibitor A-419259. This compound potently inhibits Hck, Fgr, and Lyn as well as Flt3 bearing an activating internal tandem duplication (ITD). Flt3-ITD expression sensitized human TF-1 myeloid cells to growth arrest by A-419259, supporting direct action on the Flt3-ITD kinase domain. Cells transformed with the Flt3-ITD mutants D835Y and F691L were resistant to A-419259, while co-expression of Hck or Fgr restored inhibitor sensitivity to Flt3-ITD D835Y. Conversely, Hck and Fgr mutants with engineered A-419259 resistance mutations decreased sensitivity of TF-1/Flt3-ITD cells. To investigate de novo resistance mechanisms, A-419259-resistant Flt3-ITD+ AML cell populations were derived via long-term dose escalation. Whole exome sequencing identified a distinct Flt3-ITD kinase domain mutation (N676S/T) among all A-419259 target kinases in each of six independent resistant cell populations. These studies show that Hck and Fgr expression influences inhibitor sensitivity and the pathway to acquired resistance in Flt3-ITD+ AML.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute , Mutation, Missense , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-hck , Proto-Oncogene Proteins , Pyrimidines/pharmacology , Pyrroles/pharmacology , fms-Like Tyrosine Kinase 3 , src-Family Kinases , Amino Acid Substitution , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Prognosis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-hck/biosynthesis , Proto-Oncogene Proteins c-hck/genetics , Exome Sequencing , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , src-Family Kinases/biosynthesis , src-Family Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL