Erb‑B2 Receptor Tyrosine Kinase 2 Is Negatively Regulated by the P53‑Responsive Microrna‑3184‑5P in Cervical Cancer Cells

Total Page:16

File Type:pdf, Size:1020Kb

Erb‑B2 Receptor Tyrosine Kinase 2 Is Negatively Regulated by the P53‑Responsive Microrna‑3184‑5P in Cervical Cancer Cells ONCOLOGY REPORTS 45: 95-106, 2021 Erb‑B2 Receptor Tyrosine Kinase 2 is negatively regulated by the p53‑responsive microRNA‑3184‑5p in cervical cancer cells HONGLI LIU1, YUZHI LI1, JING ZHANG1, NAN WU2, FEI LIU2, LIHUA WANG1, YUAN ZHANG1, JING LIU1, XUAN ZHANG3, SUYANG GUO1 and HONGTAO WANG4 Departments of 1Gynecological Oncology and 2Respiration and Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, First Affiliated Hospital of Bengbu Medical College; Departments of3 Gynecological Oncology and 4Immunology and Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China Received November 30, 2019; Accepted October 2, 2020 DOI: 10.3892/or.2020.7862 Abstract. The oncogenic role of Erb-B2 Receptor Tyrosine Introduction Kinase 2 (ERBB2) has been identified in several types of cancer, but less is known on its function and mechanism of Among women, cervical cancer is ranked 4th in global action in cervical cancer cells. The present study employed cancer-associated deaths (1), with over half a million deaths a multipronged approach to investigate the role of ERBB2 in in 2012 (2). Cervical cancer can be broadly categorized into cervical cancer. ERBB2 and microRNA (miR)-3184-5p expres- squamous cell carcinoma, which constitutes the majority of sion was assessed in patient-derived cervical cancer biopsy cases (70-80%) or adenocarcinoma, which comprises 10-15% tissues, revealing that higher levels of ERBB2 and lower levels of cases (3). Cervical cancer is frequently caused by the of miR-3184-5p were associated with clinicopathological indi- oncovirus human papillomavirus (HPV), mainly by types cators of cervical cancer progression. Furthermore, ERBB2 HPV-16 and -18 in ~70% of patients with cervical cancer (3). stimulated proliferation, migration and sphere-formation of The molecular drivers of the formation and development of cervical cancer cells in vitro. This effect was mediated by cervical cancer are often due to recurrent mutations within enhanced phosphatidylinositol-4,5-bisphosphate 3-kinase the PI3K/AKT signaling pathway (4-7) or from deactivating, catalytic subunit α activity. Additionally, it was revealed that loss-of-function mutations in the tumor suppressor TP53 miR-3184-5p directly suppressed ERBB2 in cervical cancer gene (p53 protein), which produces a particularly aggres- cells. The p53 activator Mithramycin A stimulated p53 and sive phenotype (8). Additionally, altered expression levels miR-3184-5p expression, thereby lowering the levels of ERBB2 of microRNAs (miRNAs/miRs), which are short, regula- and attenuating proliferation, migration and sphere-formation tory, non-coding RNAs, have been implicated in metastatic of cervical cancer cells. In conclusion, the findings of the cervical cancer (9,10). Among miRNAs associated with present study suggested ERBB2 as an oncogenic protein that cervical cancer (11-13), miR-3184 has been identified as a may promote invasiveness in cervical cancer cells. Treatment p53-responsive miRNA (14). However, its mechanistic role in of cervical cancer cells with the p53 activator Mithramycin cervical oncogenesis remains unknown. A restored the levels of the endogenous ERBB2 inhibitor Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2), also known miR-3184-5p and may represent a novel treatment strategy for as human epidermal growth factor receptor 2, is an established cervical cancer. oncogenic protein associated with tumorigenesis, cancer progression and acquisition of therapy resistance in breast, esophagogastric and colorectal cancer (15-17). However, the involvement of ERBB2 in cervical cancer development is less well known (18). Mithramycin A is an antibacterial and anticancer agent that was first purified from Streptomyces bacteria (19-21). It binds to G/C-rich stretches of DNA within minor grooves (19), which is one of its possible mechanisms for its antitumor properties (22). Mithramycin A possesses a Correspondence to: Mr. Hongtao Wang, Department of Immunology and Anhui Key Laboratory of Infection and favorable safety profile and inhibits tumor proliferationin vivo Immunity, Bengbu Medical College, 2600 Donghai Road, Bengbu, in cervical cancer murine xenograft models (23). Another Anhui 233030, P.R. China possible mechanism for the antitumor effects of Mithramycin E-mail: [email protected] A is via p53 activation (24); however, in cervical cancer, the influence of Mithramycin A on p53 and ERBB2 has not been Key words: cervical cancer, Erb-B2 Receptor Tyrosine Kinase 2, examined. microRNA-3184-5p, p53 Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) is the catalytic subunit of PI3K (25). In humans, PIK3CA acts as a proto-oncogene that supports 96 LIU et al: ONCOGENIC ERBB2 REGULATED BY p53/miR-3184-5p AXIS proliferation and metastasis of tumor cells (25), and PIK3CA plasmid vector (cat. no. PCMV6XL5) were obtained from copy number gains are associated with higher-grade tumors OriGene Technologies, Inc. For plasmid transfections, cells and poor patient prognosis in several malignancies, including were seeded in 6-well plates (1.5x105 cells/well) and tran- gastric, thyroid and prostate cancer (25). In patients with siently transfected with 1 µg plasmid using Lipofectamine® cervical cancer, PIK3CA mutations are significantly associ- 3000 transfection reagent (Invitrogen; Thermo Fisher ated with shorter survival times (PIK3CA mutant median Scientific, Inc.) for 20 min at room temperature and then survival of 67.1 months vs. non-mutant median survival of incubated in fresh medium at 37˚C for an additional 48 h 90.3 months) (26). The aforementioned evidence suggests prior to subsequent experimentation. siRNAs targeting human that targeting PIK3CA activity may be a rational approach ERBB2 (ERBB2-siRNA; cat. no. sc-29405), human PIK3CA in combating cervical cancer. Therefore, the present study (PIK3CA-siRNA; cat. no. sc-39127) and scrambled control aimed to examine the association between the effects of the (scr-siRNA; cat. no. sc-37007) were obtained from Santa p53-responsive miR-3184-5p, ERBB2 and PIK3CA in cervical Cruz Biotechnology, Inc. For siRNA transfections, cells were cancer. seeded in 6-well plates (2x105 cells/well) and transiently transfected with 80 pmol siRNA using Lipofectamine 3000 Materials and methods transfection reagent for 7 h at 37˚C and then incubated in fresh medium at 37˚C for an additional 48 h prior to subse- Patient samples. The present study was approved by the quent experimentation. miR-3184-5p (miRBase accession no. Ethics Review Committee of the First Affiliated Hospital MIMAT0015064) mirVana® miRNA mimic (cat. no. 4464066) of Bengbu Medical College (approval no. 201858; Bengbu, and mirVana® miRNA inhibitor (cat. no. 4464084), as well as China). Patients who donated cervical cancer tissues gave the corresponding controls (cat. nos. 4464058 and 4464078, their informed consent in writing prior to their involvement respectively), were obtained from AmbionÒ (Thermo Fisher in the study. All specimens were harvested following biopsy Scientific, Inc.). For miRNA mimic/inhibitor transfec- or surgery (cone biopsy, radical trachelectomy or radical tions, HeLa and SiHa cells were seeded in 6-well plates hysterectomy) at the aforementioned hospital between (1x105 cells/well) and transiently transfected with 3 nM mimic March 2000 and March 2002 from female patients (median or 10 nM inhibitor using Lipofectamine 3000 transfection age, 53 years; age range, 35-65 years) that had never received reagent for 48 h at 37˚C prior to subsequent experimentation. chemo- or radiotherapy, yielding 65 pairs of primary cervical cancer samples and matched healthy cervical tissue samples RT‑qPCR. TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.) (>2 cm from the tumor edge). Tumor specimens were staged was used to extract total RNA from cells or tissue samples, by a licensed pathologist according to the International of which a 100-ng aliquot from every sample was used for Federation of Gynecologists and Obstetricians (FIGO) staging cDNA preparation using the PrimeScript™ RT Reagent Kit system (27). The specimens were immediately kept in liquid according to the manufacturer's protocol (Takara Bio, Inc.). nitrogen and frozen at ‑80˚C until further use. A SuperScript™ III Platinum™ SYBR™ Green One-Step qRT-PCR kit or NCode™ SYBR™ Green miRNA qRT-PCR Cell lines, culture conditions and general materials. Human kit (both from Invitrogen; Thermo Fisher Scientific, Inc.) was cervical cancer HeLa and SiHa cell lines were obtained from used to quantify mRNA or miR-3184-5p expression, respec- the American Type Culture Collection. The non-cancerous, tively, on an ABI PRISM™ 7000 Sequence Detection System HPV-immortalized H8 cell line of cervical epithelial squa- (Applied Biosystems; Thermo Fisher Scientific, Inc.). The mous cells was obtained from The Cell Bank of Type Culture mRNA qPCR thermocycling conditions were as follows: 95˚C Collection of the Chinese Academy of Sciences. Cells were for 3 min, followed by 45 cycles of 95˚C for 15 sec and 60˚C grown in DMEM/F12 with 10% FBS (both from Invitrogen; for 45 sec. The miRNA qPCR thermocycling conditions were Thermo Fisher Scientific, Inc.) at 37˚C in a 5% CO2 incubator. as follows: 50˚C for 2 min and 95˚C for 2 min, followed by Reverse transcription-quantitative PCR (RT-qPCR) was 40 cycles of 95˚C for 15 sec and 60˚C for 50 sec. The following regularly employed to check for contaminating mycoplasma primers were obtained from OriGene Technologies, Inc., and
Recommended publications
  • JAK Inhibitors for Treatment of Psoriasis: Focus on Selective TYK2 Inhibitors
    Drugs https://doi.org/10.1007/s40265-020-01261-8 CURRENT OPINION JAK Inhibitors for Treatment of Psoriasis: Focus on Selective TYK2 Inhibitors Miguel Nogueira1 · Luis Puig2 · Tiago Torres1,3 © Springer Nature Switzerland AG 2020 Abstract Despite advances in the treatment of psoriasis, there is an unmet need for efective and safe oral treatments. The Janus Kinase– Signal Transducer and Activator of Transcription (JAK–STAT) pathway plays a signifcant role in intracellular signalling of cytokines of numerous cellular processes, important in both normal and pathological states of immune-mediated infamma- tory diseases. Particularly in psoriasis, where the interleukin (IL)-23/IL-17 axis is currently considered the crucial pathogenic pathway, blocking the JAK–STAT pathway with small molecules would be expected to be clinically efective. However, relative non-specifcity and low therapeutic index of the available JAK inhibitors have delayed their integration into the therapeutic armamentarium of psoriasis. Current research appears to be focused on Tyrosine kinase 2 (TYK2), the frst described member of the JAK family. Data from the Phase II trial of BMS-986165—a selective TYK2 inhibitor—in psoriasis have been published and clinical results are encouraging, with a large Phase III programme ongoing. Further, the selective TYK2 inhibitor PF-06826647 is being tested in moderate-to-severe psoriasis in a Phase II clinical trial. Brepocitinib, a potent TYK2/JAK1 inhibitor, is also being evaluated, as both oral and topical treatment. Results of studies with TYK2 inhibitors will be important in assessing the clinical efcacy and safety of these drugs and their place in the therapeutic armamentarium of psoriasis.
    [Show full text]
  • Biomarker Testing in Non- Small Cell Lung Cancer (NSCLC)
    The biopharma business of Merck KGaA, Darmstadt, Germany operates as EMD Serono in the U.S. and Canada. Biomarker testing in non- small cell lung cancer (NSCLC) Copyright © 2020 EMD Serono, Inc. All rights reserved. US/TEP/1119/0018(1) Lung cancer in the US: Incidence, mortality, and survival Lung cancer is the second most common cancer diagnosed annually and the leading cause of mortality in the US.2 228,820 20.5% 57% Estimated newly 5-year Advanced or 1 survival rate1 metastatic at diagnosed cases in 2020 diagnosis1 5.8% 5-year relative 80-85% 2 135,720 survival with NSCLC distant disease1 Estimated deaths in 20201 2 NSCLC, non-small cell lung cancer; US, United States. 1. National Institutes of Health (NIH), National Cancer Institute. Cancer Stat Facts: Lung and Bronchus Cancer website. www.seer.cancer.gov/statfacts/html/lungb.html. Accessed May 20, 2020. 2. American Cancer Society. What is Lung Cancer? website. https://www.cancer.org/cancer/non-small-cell-lung-cancer/about/what-is-non-small-cell-lung-cancer.html. Accessed May 20, 2020. NSCLC is both histologically and genetically diverse 1-3 NSCLC distribution by histology Prevalence of genetic alterations in NSCLC4 PTEN 10% DDR2 3% OTHER 25% PIK3CA 12% LARGE CELL CARCINOMA 10% FGFR1 20% SQUAMOUS CELL CARCINOMA 25% Oncogenic drivers in adenocarcinoma Other or ADENOCARCINOMA HER2 1.9% 40% KRAS 25.5% wild type RET 0.7% 55% NTRK1 1.7% ROS1 1.7% Oncogenic drivers in 0% 20% 40% 60% RIT1 2.2% squamous cell carcinoma Adenocarcinoma DDR2 2.9% Squamous cell carcinoma NRG1 3.2% Large cell carcinoma
    [Show full text]
  • Targeting the Function of the HER2 Oncogene in Human Cancer Therapeutics
    Oncogene (2007) 26, 6577–6592 & 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00 www.nature.com/onc REVIEW Targeting the function of the HER2 oncogene in human cancer therapeutics MM Moasser Department of Medicine, Comprehensive Cancer Center, University of California, San Francisco, CA, USA The year 2007 marks exactly two decades since human HER3 (erbB3) and HER4 (erbB4). The importance of epidermal growth factor receptor-2 (HER2) was func- HER2 in cancer was realized in the early 1980s when a tionally implicated in the pathogenesis of human breast mutationally activated form of its rodent homolog neu cancer (Slamon et al., 1987). This finding established the was identified in a search for oncogenes in a carcinogen- HER2 oncogene hypothesis for the development of some induced rat tumorigenesis model(Shih et al., 1981). Its human cancers. An abundance of experimental evidence human homologue, HER2 was simultaneously cloned compiled over the past two decades now solidly supports and found to be amplified in a breast cancer cell line the HER2 oncogene hypothesis. A direct consequence (King et al., 1985). The relevance of HER2 to human of this hypothesis was the promise that inhibitors of cancer was established when it was discovered that oncogenic HER2 would be highly effective treatments for approximately 25–30% of breast cancers have amplifi- HER2-driven cancers. This treatment hypothesis has led cation and overexpression of HER2 and these cancers to the development and widespread use of anti-HER2 have worse biologic behavior and prognosis (Slamon antibodies (trastuzumab) in clinical management resulting et al., 1989).
    [Show full text]
  • Kinase-Targeted Cancer Therapies: Progress, Challenges and Future Directions Khushwant S
    Bhullar et al. Molecular Cancer (2018) 17:48 https://doi.org/10.1186/s12943-018-0804-2 REVIEW Open Access Kinase-targeted cancer therapies: progress, challenges and future directions Khushwant S. Bhullar1, Naiara Orrego Lagarón2, Eileen M. McGowan3, Indu Parmar4, Amitabh Jha5, Basil P. Hubbard1 and H. P. Vasantha Rupasinghe6,7* Abstract The human genome encodes 538 protein kinases that transfer a γ-phosphate group from ATP to serine, threonine, or tyrosine residues. Many of these kinases are associated with human cancer initiation and progression. The recent development of small-molecule kinase inhibitors for the treatment of diverse types of cancer has proven successful in clinical therapy. Significantly, protein kinases are the second most targeted group of drug targets, after the G-protein- coupled receptors. Since the development of the first protein kinase inhibitor, in the early 1980s, 37 kinase inhibitors have received FDA approval for treatment of malignancies such as breast and lung cancer. Furthermore, about 150 kinase-targeted drugs are in clinical phase trials, and many kinase-specific inhibitors are in the preclinical stage of drug development. Nevertheless, many factors confound the clinical efficacy of these molecules. Specific tumor genetics, tumor microenvironment, drug resistance, and pharmacogenomics determine how useful a compound will be in the treatment of a given cancer. This review provides an overview of kinase-targeted drug discovery and development in relation to oncology and highlights the challenges and future potential for kinase-targeted cancer therapies. Keywords: Kinases, Kinase inhibition, Small-molecule drugs, Cancer, Oncology Background Recent advances in our understanding of the fundamen- Kinases are enzymes that transfer a phosphate group to a tal molecular mechanisms underlying cancer cell signaling protein while phosphatases remove a phosphate group have elucidated a crucial role for kinases in the carcino- from protein.
    [Show full text]
  • Hepatocyte Growth Factor Receptor (C-Met)/Fc Chimera Human
    HGF RECEPTOR (C-Met)/FC CHIMERA Human, Recombinant Expressed in mouse NSO cells Product Number H 0536 Product Description c-Met can also be negatively modulated by phos- Recombinant Human HGF Receptor (c-Met)/Fc phorylation of Ser 985 by protein kinase C. Other Chimera is produced from the extracellular domain of ligand-receptor activities involve binding that leads to human HGF receptor protein fused to the C-terminal 6X enhanced integrin-mediated B cell and lymphoma cell histidine-tagged Fc region of human IgG1 by a adhesion.4, 5 polypeptide linker.1 It is a disulfide-linked tetramer containing two proteolytically cleaved α and β subunits. Normal HGF-Met signaling is needed for embryonic Each α and β subunit heterodimer contain 1152 amino development and abnormal signaling has been 6 acid residues with a calculated molecular mass of implicated in tumorigenesis. approximately 129 kDa. Human and mouse HGF receptors share 89% amino acid identity and human Reagent HGF can bind to the mouse receptor. Recombinant Human HGF Receptor (c-Met)/Fc Chimera is supplied as approximately 100 µg of protein Hepatocyte growth factor receptor (HGF R), a product lyophilized from a 0.2 µm filtered solution in phosphate of the proto-oncogene c-Met, is a heterodimeric buffered saline (PBS) containing 5 mg of bovine serum transmembrane glycoprotein that is a receptor-type albumin. tyrosine kinase.2 The c-Met heterodimer is composed of an α chain that is disulfide-linked to a β chain. The Preparation Instructions α chain is exposed to the cell surface and the β chain Reconstitute the contents of the vial using sterile spans the plasma membrane.
    [Show full text]
  • Protein Tyrosine Kinases: Their Roles and Their Targeting in Leukemia
    cancers Review Protein Tyrosine Kinases: Their Roles and Their Targeting in Leukemia Kalpana K. Bhanumathy 1,*, Amrutha Balagopal 1, Frederick S. Vizeacoumar 2 , Franco J. Vizeacoumar 1,3, Andrew Freywald 2 and Vincenzo Giambra 4,* 1 Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; [email protected] (A.B.); [email protected] (F.J.V.) 2 Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada; [email protected] (F.S.V.); [email protected] (A.F.) 3 Cancer Research Department, Saskatchewan Cancer Agency, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada 4 Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy * Correspondence: [email protected] (K.K.B.); [email protected] (V.G.); Tel.: +1-(306)-716-7456 (K.K.B.); +39-0882-416574 (V.G.) Simple Summary: Protein phosphorylation is a key regulatory mechanism that controls a wide variety of cellular responses. This process is catalysed by the members of the protein kinase su- perfamily that are classified into two main families based on their ability to phosphorylate either tyrosine or serine and threonine residues in their substrates. Massive research efforts have been invested in dissecting the functions of tyrosine kinases, revealing their importance in the initiation and progression of human malignancies. Based on these investigations, numerous tyrosine kinase inhibitors have been included in clinical protocols and proved to be effective in targeted therapies for various haematological malignancies.
    [Show full text]
  • Hirbe AC, Kaushal M, Sharma MK
    Original Article Clinical Genomic Profiling Identifies TYK2 Mutation and Overexpression in Patients With Neurofibromatosis Type 1-Associated Malignant Peripheral Nerve Sheath Tumors Angela C. Hirbe, MD, PhD1; Madhurima Kaushal, MS2; Mukesh Kumar Sharma, PhD2; Sonika Dahiya, MD2; Melike Pekmezci, MD3; Arie Perry, MD3,4; and David H. Gutmann, MD, PhD5 BACKGROUND: Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas that arise at an estimated frequency of 8% to 13% in individuals with neurofibromatosis type 1 (NF1). Compared with their sporadic counterparts, NF1-associated MPNSTs (NF1-MPNSTs) develop in young adults, frequently recur (approximately 50% of cases), and carry a dismal prognosis. As such, most individuals affected with NF1-MPNSTs die within 5 years of diagnosis, despite surgical resection combined with radiotherapy and che- motherapy. METHODS: Clinical genomic profiling was performed using 1000 ng of DNA from 7 cases of NF1-MPNST, and bioinformatic analyses were conducted to identify genes with actionable mutations. RESULTS: A total of 3 women and 4 men with NF1-MPNST were identified (median age, 38 years). Nonsynonymous mutations were discovered in 4 genes (neurofibromatosis type 1 [NF1], ROS proto-oncogene 1 [ROS1], tumor protein p53 [TP53], and tyrosine kinase 2 [TYK2]), which in addition were mutated in other MPNST cases in this sample set. Consistent with their occurrence in individuals with NF1, all tumors had at least 1 mutation in the NF1 gene. Whereas TP53 gene mutations are frequently observed in other cancers, ROS1 mutations are common in melanoma (15%-35%), anoth- er neural crest-derived malignancy. In contrast, TYK2 mutations are uncommon in other malignancies (<7%).
    [Show full text]
  • Trastuzumab Mechanism of Action; 20 Years of Research to Unravel a Dilemma
    cancers Review Trastuzumab Mechanism of Action; 20 Years of Research to Unravel a Dilemma Hamid Maadi 1, Mohammad Hasan Soheilifar 2, Won-Shik Choi 1, Abdolvahab Moshtaghian 3,4 and Zhixiang Wang 5,* 1 Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada; [email protected] (H.M.); [email protected] (W.-S.C.) 2 Department of Medical Laser, Medical Laser Research Center, Yara Institute, ACECR, Tehran 1315795613, Iran; [email protected] 3 Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar 4741695447, Iran; [email protected] 4 Deputy of Research and Technology, Semnan University of Medical Sciences, Semnan 3514799442, Iran 5 Department of Medical Genetics and Signal, Transduction Research Group, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada * Correspondence: [email protected] Simple Summary: Overexpression of HER2 receptors have been identified in various types of cancer including breast cancer and ovarian cancer. HER2 overexpression is generally associated with poor clinical outcomes in patients with HER2-positve tumors. Trastuzumab, an antibody specifically targeting HER2 receptors, showed promising clinical benefits for patients with HER2-positive tumors. Studies show that trastuzumab suppresses HER2 receptors’ oncogenic functions in HER2-postive tumors. Moreover, trastuzumab has been shown to provoke immune responses against the HER2- amplified tumors. Citation: Maadi, H.; Soheilifar, M.H.; Choi, W.-S.; Moshtaghian, A.; Wang, Z. Trastuzumab Mechanism of Abstract: Trastuzumab as a first HER2-targeted therapy for the treatment of HER2-positive breast Action; 20 Years of Research to cancer patients was introduced in 1998.
    [Show full text]
  • Anti-Hepatocyte Growth Factor Receptor (C-Met) Produced in Goat, Affinity Isolated Antibody
    Anti-Hepatocyte Growth Factor Receptor (c-Met) produced in goat, affinity isolated antibody Catalog Number H9786 Product Description Autophosphorylation at two tyrosines upregulates Anti-Hepatocyte Growth Factor Receptor (c-Met) is kinase activity while phosphorylation at two other produced in goat using purified recombinant human tyrosines generates SH2 docking sites for adapter hepatocyte growth factor receptor (HGF R) extracellular proteins such as Shc, Grb2, CrK/CRKL, and Gab1. domain expressed in mouse NS0 cells as immunogen. The antibody is purified using human HGF R affinity Receptor activation has been correlated to the chromatography. activation of the Ras pathway, which culminates in the activation and consequent nuclear translocation of MAP Anti-Hepatocyte Growth Factor Receptor (c-Met) will kinase. c-Met can also be negatively modulated by neutralize receptor-ligand interaction. The antibody may phosphorylation of Ser985 by protein kinase C. Other also be used in functional ELISA, immunoblotting, ligand-receptor activities involve binding that leads to immunohistochemistry, and flow cytometry. enhanced integrin-mediated B cell and lymphoma cell adhesion.4,5 Normal HGF-Met signaling is needed for Hepatocyte growth factor receptor (HGF R), a product embryonic development and abnormal signaling and of the proto-oncogene c-Met, is a heterodimeric has been implicated in tumorigenesis.6 transmembrane glycoprotein that is a receptor-type tyrosine kinase.2 The c-Met heterodimer is composed of Reagent an chain that is disulfide-linked to a chain. Each Lyophilized from 0.2 m-filtered solution in phosphate and subunit heterodimer contains 1,152 amino acid buffered saline containing carbohydrates. residues with a calculated molecular mass of 129 kDa.
    [Show full text]
  • TYK2 Variants in B-Acute Lymphoblastic Leukaemia
    G C A T T A C G G C A T genes Article TYK2 Variants in B-Acute Lymphoblastic Leukaemia 1, , 1, 1, Edgar Turrubiartes-Martínez y z , Irene Bodega-Mayor y, Pablo Delgado-Wicke y , Francisca Molina-Jiménez 1, Diana Casique-Aguirre 2 , Martín González-Andrade 3 , Inmaculada Rapado 4, Mireia Camós 5, Cristina Díaz-de-Heredia 6 , Eva Barragán 7, Manuel Ramírez-Orellana 8 , Beatriz Aguado 9 , Ángela Figuera 9, Joaquín Martínez-López 4,10,11 and Elena Fernández-Ruiz 1,12,* 1 Molecular Biology Unit, University Hospital La Princesa and Research Institute (IIS-IP), Diego de León 62, 28006 Madrid, Spain; [email protected] (E.T.-M.); [email protected] (I.B.-M.); [email protected] (P.D.-W.); [email protected] (F.M.-J.) 2 School of Medicine, Latin University of Mexico (ULM), Avenida San Jose 100, 38085 Celaya, Mexico; [email protected] 3 Biochemistry Department, Biosensors and Molecular Modelling Lab, Autonomous National University of México (UNAM), 04510 Mexico City, Mexico; [email protected] 4 Haematology Department, University Hospital 12 Octubre and Research Institute (i+12), Avenida de Córdoba s/n., 28041 Madrid, Spain; [email protected] (I.R.); [email protected] (J.M.-L.) 5 Haematology Department, University Hospital Sant Joan de Déu and Research Institute (IRSJD), Passeig Sant Joan de Déu 2, 08950 Esplugues de Llobregat, Spain; [email protected] 6 Paediatric Haematology and Oncology Department, University Hospital Vall d’Hebron and Research Institute (VHIR), Passeig de la Vall d’Hebron 119-129, 08035
    [Show full text]
  • Janus Kinases in Leukemia
    cancers Review Janus Kinases in Leukemia Juuli Raivola 1, Teemu Haikarainen 1, Bobin George Abraham 1 and Olli Silvennoinen 1,2,3,* 1 Faculty of Medicine and Health Technology, Tampere University, 33014 Tampere, Finland; juuli.raivola@tuni.fi (J.R.); teemu.haikarainen@tuni.fi (T.H.); bobin.george.abraham@tuni.fi (B.G.A.) 2 Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, 00014 Helsinki, Finland 3 Fimlab Laboratories, Fimlab, 33520 Tampere, Finland * Correspondence: olli.silvennoinen@tuni.fi Simple Summary: Janus kinase/signal transducers and activators of transcription (JAK/STAT) path- way is a crucial cell signaling pathway that drives the development, differentiation, and function of immune cells and has an important role in blood cell formation. Mutations targeting this path- way can lead to overproduction of these cell types, giving rise to various hematological diseases. This review summarizes pathogenic JAK/STAT activation mechanisms and links known mutations and translocations to different leukemia. In addition, the review discusses the current therapeutic approaches used to inhibit constitutive, cytokine-independent activation of the pathway and the prospects of developing more specific potent JAK inhibitors. Abstract: Janus kinases (JAKs) transduce signals from dozens of extracellular cytokines and function as critical regulators of cell growth, differentiation, gene expression, and immune responses. Deregu- lation of JAK/STAT signaling is a central component in several human diseases including various types of leukemia and other malignancies and autoimmune diseases. Different types of leukemia harbor genomic aberrations in all four JAKs (JAK1, JAK2, JAK3, and TYK2), most of which are Citation: Raivola, J.; Haikarainen, T.; activating somatic mutations and less frequently translocations resulting in constitutively active JAK Abraham, B.G.; Silvennoinen, O.
    [Show full text]
  • Tyrosine Kinase Inhibitors for Solid Tumors in the Past 20 Years (2001–2020) Liling Huang†, Shiyu Jiang† and Yuankai Shi*
    Huang et al. J Hematol Oncol (2020) 13:143 https://doi.org/10.1186/s13045-020-00977-0 REVIEW Open Access Tyrosine kinase inhibitors for solid tumors in the past 20 years (2001–2020) Liling Huang†, Shiyu Jiang† and Yuankai Shi* Abstract Tyrosine kinases are implicated in tumorigenesis and progression, and have emerged as major targets for drug discovery. Tyrosine kinase inhibitors (TKIs) inhibit corresponding kinases from phosphorylating tyrosine residues of their substrates and then block the activation of downstream signaling pathways. Over the past 20 years, multiple robust and well-tolerated TKIs with single or multiple targets including EGFR, ALK, ROS1, HER2, NTRK, VEGFR, RET, MET, MEK, FGFR, PDGFR, and KIT have been developed, contributing to the realization of precision cancer medicine based on individual patient’s genetic alteration features. TKIs have dramatically improved patients’ survival and quality of life, and shifted treatment paradigm of various solid tumors. In this article, we summarized the developing history of TKIs for treatment of solid tumors, aiming to provide up-to-date evidence for clinical decision-making and insight for future studies. Keywords: Tyrosine kinase inhibitors, Solid tumors, Targeted therapy Introduction activation of tyrosine kinases due to mutations, translo- According to GLOBOCAN 2018, an estimated 18.1 cations, or amplifcations is implicated in tumorigenesis, million new cancer cases and 9.6 million cancer deaths progression, invasion, and metastasis of malignancies. occurred in 2018 worldwide [1]. Targeted agents are In addition, wild-type tyrosine kinases can also func- superior to traditional chemotherapeutic ones in selec- tion as critical nodes for pathway activation in cancer.
    [Show full text]