1
|
Choi KH, Lee DK, Jeong J, Ahn Y, Go DM, Kim DY, Lee CK. Inhibition of BMP-mediated SMAD pathway supports the pluripotency of pig embryonic stem cells in the absence of feeder cells. Theriogenology 2024; 225:67-80. [PMID: 38795512 DOI: 10.1016/j.theriogenology.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/18/2024] [Accepted: 05/18/2024] [Indexed: 05/28/2024]
Abstract
Here, we examined the effects of the BMP signaling pathway inhibitor LDN-193189 on the pluripotency of porcine embryonic stem cells (ESCs) in the absence of feeder cells using molecular and transcriptomic techniques. Additionally, the effects of some extracellular matrix components on porcine ESC pluripotency were evaluated to develop an optimized and sustainable feeder-free culture system for porcine ESCs. Feeder cells were found to play an important role in supporting the pluripotency of porcine ESCs by blocking trophoblast and mesodermal differentiation through the inhibition of the BMP pathway. Additionally, treatment with LDN-193189, an inhibitor of the BMP pathway, maintained the pluripotency and homogeneity of porcine ESCs for an extended period in the absence of feeder cells by stimulating the secretion of chemokines and suppressing differentiation, based on transcriptome analysis. Conclusively, these results suggest that LDN-193189 could be a suitable replacement for feeder cells in the maintenance of porcine ESC pluripotency during culture. Additionally, these findings contribute to the understanding of pluripotency gene networks and comparative embryogenesis.
Collapse
Affiliation(s)
- Kwang-Hwan Choi
- Research and Development Center, Space F Corporation, Hwaseong, Gyeonggi-do, 18471, Republic of Korea; Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Dong-Kyung Lee
- Research and Development Center, Space F Corporation, Hwaseong, Gyeonggi-do, 18471, Republic of Korea; Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinsol Jeong
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yelim Ahn
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Du-Min Go
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea; Institute of Green Bio Science and Technology, Seoul National University, Pyeong Chang, 25354, Republic of Korea.
| |
Collapse
|
2
|
Kim JH, Kang KW, Park Y, Kim BS. CXCR2 inhibition overcomes ponatinib intolerance by eradicating chronic myeloid leukemic stem cells through PI3K/Akt/mTOR and dipeptidylpeptidase Ⅳ (CD26). Heliyon 2023; 9:e22091. [PMID: 38045173 PMCID: PMC10692791 DOI: 10.1016/j.heliyon.2023.e22091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 12/05/2023] Open
Abstract
This study explores the therapeutic potential of targeting CXCR2 in patients afflicted with ponatinib-resistant chronic myeloid leukemia (CML). Ponatinib, a third-generation tyrosine kinase inhibitor (TKI), was initially designed for treating patients with CML harboring the T315I mutation. However, resistance or intolerance issues may lead to treatment discontinuation. Additionally, TKIs have exhibited limitations in eradicating quiescent CML stem cells. Our investigation reveals the activation of CXC chemokine receptor 2 (CXCR2) signaling in response to chemotherapeutic stress. Treatment with the CXCR2 antagonist, SB225002, effectively curtails cell proliferation and triggers apoptosis in ponatinib-resistant CML cells. SB225002 intervention also results in the accumulation of reactive oxygen species and disruption of mitochondrial function, phenomena associated with TKI chemoresistance and apoptosis. Furthermore, we demonstrate that activated CXCR2 expression induces the activity of dipeptidylpeptidase Ⅳ (DPP4/CD26), a CML leukemic stem cell marker, and concomitantly inhibits the PI3K/Akt/mTOR pathway cascades. These findings underscore the novel role of CXCR2 in the regulation of not only ponatinib-resistant CML cells, but also CML leukemic stem cells. Consequently, our study proposes that targeting CXCR2 holds promise as a viable therapeutic strategy for addressing patients with CML grappling with ponatinib resistance.
Collapse
Affiliation(s)
- Ji-Hea Kim
- Institute of Stem Cell Research, Korea University College of Medicine, Seoul, South Korea
- Department of Biomedical Science, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Ka-Won Kang
- Department of Internal Medicine, Anam Hospital Korea University Medical Center, Seoul, South Korea
| | - Yong Park
- Department of Internal Medicine, Anam Hospital Korea University Medical Center, Seoul, South Korea
| | - Byung Soo Kim
- Department of Internal Medicine, Anam Hospital Korea University Medical Center, Seoul, South Korea
| |
Collapse
|
3
|
Yang J, Bergdorf K, Yan C, Luo W, Chen SC, Ayers GD, Liu Q, Liu X, Boothby M, Weiss VL, Groves SM, Oleskie AN, Zhang X, Maeda DY, Zebala JA, Quaranta V, Richmond A. CXCR2 expression during melanoma tumorigenesis controls transcriptional programs that facilitate tumor growth. Mol Cancer 2023; 22:92. [PMID: 37270599 PMCID: PMC10239119 DOI: 10.1186/s12943-023-01789-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/16/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND Though the CXCR2 chemokine receptor is known to play a key role in cancer growth and response to therapy, a direct link between expression of CXCR2 in tumor progenitor cells during induction of tumorigenesis has not been established. METHODS To characterize the role of CXCR2 during melanoma tumorigenesis, we generated tamoxifen-inducible tyrosinase-promoter driven BrafV600E/Pten-/-/Cxcr2-/- and NRasQ61R/INK4a-/-/Cxcr2-/- melanoma models. In addition, the effects of a CXCR1/CXCR2 antagonist, SX-682, on melanoma tumorigenesis were evaluated in BrafV600E/Pten-/- and NRasQ61R/INK4a-/- mice and in melanoma cell lines. Potential mechanisms by which Cxcr2 affects melanoma tumorigenesis in these murine models were explored using RNAseq, mMCP-counter, ChIPseq, and qRT-PCR; flow cytometry, and reverse phosphoprotein analysis (RPPA). RESULTS Genetic loss of Cxcr2 or pharmacological inhibition of CXCR1/CXCR2 during melanoma tumor induction resulted in key changes in gene expression that reduced tumor incidence/growth and increased anti-tumor immunity. Interestingly, after Cxcr2 ablation, Tfcp2l1, a key tumor suppressive transcription factor, was the only gene significantly induced with a log2 fold-change greater than 2 in these three different melanoma models. CONCLUSIONS Here, we provide novel mechanistic insight revealing how loss of Cxcr2 expression/activity in melanoma tumor progenitor cells results in reduced tumor burden and creation of an anti-tumor immune microenvironment. This mechanism entails an increase in expression of the tumor suppressive transcription factor, Tfcp2l1, along with alteration in the expression of genes involved in growth regulation, tumor suppression, stemness, differentiation, and immune modulation. These gene expression changes are coincident with reduction in the activation of key growth regulatory pathways, including AKT and mTOR.
Collapse
Affiliation(s)
- J Yang
- TVHS Department of Veterans Affairs, Nashville, TN, 37212, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
| | - K Bergdorf
- TVHS Department of Veterans Affairs, Nashville, TN, 37212, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
| | - C Yan
- TVHS Department of Veterans Affairs, Nashville, TN, 37212, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
| | - W Luo
- TVHS Department of Veterans Affairs, Nashville, TN, 37212, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
| | - S C Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37203-1742, USA
| | - G D Ayers
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37203-1742, USA
| | - Q Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37203-1742, USA
| | - X Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, 37203-1742, USA
| | - M Boothby
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - V L Weiss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - S M Groves
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - A N Oleskie
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
| | - X Zhang
- Department of Genomic Medicine, MD Anderson Cancer Center, University of Texas, Houston, TX, 77030, USA
| | - D Y Maeda
- Syntrix Pharmaceuticals, Auburn, WA, 98001, USA
| | - J A Zebala
- Syntrix Pharmaceuticals, Auburn, WA, 98001, USA
| | - V Quaranta
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA
- Department of Biochemistry, Vanderbilt University, TN, 37240, Nashville, USA
| | - A Richmond
- TVHS Department of Veterans Affairs, Nashville, TN, 37212, USA.
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, 37240, USA.
| |
Collapse
|
4
|
Yang J, Bergdorf K, Yan C, Luo W, Chen SC, Ayers D, Liu Q, Liu X, Boothby M, Groves SM, Oleskie AN, Zhang X, Maeda DY, Zebala JA, Quaranta V, Richmond A. CXCR2 expression during melanoma tumorigenesis controls transcriptional programs that facilitate tumor growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.22.529548. [PMID: 36865260 PMCID: PMC9980137 DOI: 10.1101/2023.02.22.529548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Background Though the CXCR2 chemokine receptor is known to play a key role in cancer growth and response to therapy, a direct link between expression of CXCR2 in tumor progenitor cells during induction of tumorigenesis has not been established. Methods To characterize the role of CXCR2 during melanoma tumorigenesis, we generated tamoxifen-inducible tyrosinase-promoter driven Braf V600E /Pten -/- /Cxcr2 -/- and NRas Q61R /INK4a -/- /Cxcr2 -/- melanoma models. In addition, the effects of a CXCR1/CXCR2 antagonist, SX-682, on melanoma tumorigenesis were evaluated in Braf V600E /Pten -/- and NRas Q61R /INK4a -/- mice and in melanoma cell lines. Potential mechanisms by which Cxcr2 affects melanoma tumorigenesis in these murine models were explored using RNAseq, mMCP-counter, ChIPseq, and qRT-PCR; flow cytometry, and reverse phosphoprotein analysis (RPPA). Results Genetic loss of Cxcr2 or pharmacological inhibition of CXCR1/CXCR2 during melanoma tumor induction resulted in key changes in gene expression that reduced tumor incidence/growth and increased anti-tumor immunity. Interestingly, after Cxcr2 ablation, Tfcp2l1 , a key tumor suppressive transcription factor, was the only gene significantly induced with a log 2 fold-change greater than 2 in these three different melanoma models. Conclusions Here, we provide novel mechanistic insight revealing how loss of Cxcr2 expression/activity in melanoma tumor progenitor cells results in reduced tumor burden and creation of an anti-tumor immune microenvironment. This mechanism entails an increase in expression of the tumor suppressive transcription factor, Tfcp2l1, along with alteration in the expression of genes involved in growth regulation, tumor suppression, stemness, differentiation, and immune modulation. These gene expression changes are coincident with reduction in the activation of key growth regulatory pathways, including AKT and mTOR.
Collapse
|
5
|
The Importance of CXCL1 in the Physiological State and in Noncancer Diseases of the Oral Cavity and Abdominal Organs. Int J Mol Sci 2022; 23:ijms23137151. [PMID: 35806156 PMCID: PMC9266754 DOI: 10.3390/ijms23137151] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/23/2022] [Accepted: 06/25/2022] [Indexed: 02/06/2023] Open
Abstract
CXCL1 is a CXC chemokine, CXCR2 ligand and chemotactic factor for neutrophils. In this paper, we present a review of the role of the chemokine CXCL1 in physiology and in selected major non-cancer diseases of the oral cavity and abdominal organs (gingiva, salivary glands, stomach, liver, pancreas, intestines, and kidneys). We focus on the importance of CXCL1 on implantation and placentation as well as on human pluripotent stem cells. We also show the significance of CXCL1 in selected diseases of the abdominal organs, including the gastrointestinal tract and oral cavity (periodontal diseases, periodontitis, Sjögren syndrome, Helicobacter pylori infection, diabetes, liver cirrhosis, alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), HBV and HCV infection, liver ischemia and reperfusion injury, inflammatory bowel disease (Crohn’s disease and ulcerative colitis), obesity and overweight, kidney transplantation and ischemic-reperfusion injury, endometriosis and adenomyosis).
Collapse
|
6
|
Lu V, Roy IJ, Torres A, Joly JH, Ahsan FM, Graham NA, Teitell MA. Glutamine-dependent signaling controls pluripotent stem cell fate. Dev Cell 2022; 57:610-623.e8. [PMID: 35216682 PMCID: PMC8930616 DOI: 10.1016/j.devcel.2022.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/01/2021] [Accepted: 01/31/2022] [Indexed: 12/30/2022]
Abstract
Human pluripotent stem cells (hPSCs) can self-renew indefinitely or can be induced to differentiate. We previously showed that exogenous glutamine (Gln) withdrawal biased hPSC differentiation toward ectoderm and away from mesoderm. We revealed that, although all three germ lineages are capable of de novo Gln synthesis, only ectoderm generates sufficient Gln to sustain cell viability and differentiation, and this finding clarifies lineage fate restrictions under Gln withdrawal. Furthermore, we found that Gln acts as a signaling molecule for ectoderm that supersedes lineage-specifying cytokine induction. In contrast, Gln in mesoderm and endoderm is the preferred precursor of α-ketoglutarate without a direct signaling role. Our work raises a question about whether the nutrient environment functions directly in cell differentiation during development. Interestingly, transcriptome analysis of a gastrulation-stage human embryo shows that unique Gln enzyme-encoding gene expression patterns may also distinguish germ lineages in vivo. Together, our study suggests that intracellular Gln may help coordinate differentiation of the three germ layers.
Collapse
Affiliation(s)
- Vivian Lu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Irena J Roy
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Alejandro Torres
- Molecular Biology Institute, University of California at Los Angeles, Los Angeles, CA, USA
| | - James H Joly
- Mork Family Department of Chemical Engineering and Materials Science, Los Angeles, CA 90089, USA
| | - Fasih M Ahsan
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Nicholas A Graham
- Mork Family Department of Chemical Engineering and Materials Science, Los Angeles, CA 90089, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Michael A Teitell
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California at Los Angeles, Los Angeles, CA, USA; Department of Bioengineering, Department of Pediatrics, California NanoSystems Institute, and Broad Center for Regenerative Medicine and Stem Cell Research, University of California at Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Wen YC, Liu YN, Yeh HL, Chen WH, Jiang KC, Lin SR, Huang J, Hsiao M, Chen WY. TCF7L1 regulates cytokine response and neuroendocrine differentiation of prostate cancer. Oncogenesis 2021; 10:81. [PMID: 34799554 PMCID: PMC8604986 DOI: 10.1038/s41389-021-00371-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/01/2021] [Accepted: 11/03/2021] [Indexed: 01/07/2023] Open
Abstract
Neuroendocrine differentiation (NED) is associated with WNT signaling activation and can be significantly observed after failure of androgen-deprivation therapy (ADT) for prostatic adenocarcinomas. Cytokine signaling is stimulated in NED prostate cancer; however, how ADT-upregulated WNT signaling promotes activation of cytokine signaling and contributes to NED of prostate cancer is poorly understood. In this study, we identified ADT-mediated upregulation of transcription factor 7 like 1 (TCF7L1), which increases the cytokine response and enhances NED of prostate cancer through interleukin (IL)-8/C-X-C motif chemokine receptor type 2 (CXCR2) signaling activation. ADT induced the secretion of WNT4 which upon engagement of TCF7L1 in prostate cancer cells, enhanced IL-8 and CXCR2 expressions. TCF7L1 directly binds to the regulatory sequence region of IL-8 and CXCR2 through WNT4 activation, thus upregulating IL-8/CXCR2 signaling-driven NED and cell motility. Analysis of prostate tissue samples collected from small-cell neuroendocrine prostate cancer (SCPC) and castration-resistant prostate cancer (CRPC) tumors showed an increased intensity of nuclear TCF7L1 associated with CXCR2. Our results suggest that induction of WNT4/TCF7L1 results in increased NED and malignancy in prostate cancer that is linked to dysregulation of androgen receptor signaling and activation of the IL-8/CXCR2 pathway.
Collapse
Affiliation(s)
- Yu-Ching Wen
- Department of Urology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hsiu-Lien Yeh
- General Education Development Center, Hsin Sheng Junior College of Medical Care and Management, Taoyuan, Taiwan
| | - Wei-Hao Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kuo-Ching Jiang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Shian-Ren Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jiaoti Huang
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Yu Chen
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan. .,Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
8
|
Lu V, Roy IJ, Teitell MA. Nutrients in the fate of pluripotent stem cells. Cell Metab 2021; 33:2108-2121. [PMID: 34644538 PMCID: PMC8568661 DOI: 10.1016/j.cmet.2021.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/07/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022]
Abstract
Pluripotent stem cells model certain features of early mammalian development ex vivo. Medium-supplied nutrients can influence self-renewal, lineage specification, and earliest differentiation of pluripotent stem cells. However, which specific nutrients support these distinct outcomes, and their mechanisms of action, remain under active investigation. Here, we evaluate the available data on nutrients and their metabolic conversion that influence pluripotent stem cell fates. We also discuss key questions open for investigation in this rapidly expanding area of increasing fundamental and practical importance.
Collapse
Affiliation(s)
- Vivian Lu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Irena J Roy
- Developmental and Stem Cell Biology, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael A Teitell
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
9
|
Kim JH, Lee SJ, Kang KW, Lee BH, Park Y, Kim BS. CXCR2, a novel target to overcome tyrosine kinase inhibitor resistance in chronic myelogenous leukemia cells. Biochem Pharmacol 2021; 190:114658. [PMID: 34146540 DOI: 10.1016/j.bcp.2021.114658] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 11/18/2022]
Abstract
Chronic myeloid leukemia (CML) is a reciprocal translocation disorder driven by a breakpoint cluster region (BCR)-Abelson leukemia virus (ABL) fusion gene that stimulates abnormal tyrosine kinase activity. Tyrosine kinase inhibitors (TKIs) are effective in treating Philadelphia chromosome (Ph) + CML patients. However, the appearance of TKI-resistant CML cells is a hurdle in CML treatment. Therefore, it is necessary to identify novel alternative treatments targeting tyrosine kinases. This study was designed to determine whether C-X-C chemokine receptor 2 (CXCR2) could be a novel target for TKI-resistant CML treatment. Interleukin 8 (IL-8), a CXCR2 ligand, was significantly increased in the bone marrow serum of initially diagnosed CML patients and TKI-resistant CML cell conditioned media. CXCR2 antagonists suppressed the proliferation of CML cells via cell cycle arrest in the G2/M phase. CXCR2 inhibition also attenuated mTOR, c-Myc, and BCR-ABL expression, leading to CML cell apoptosis, irrespective of TKI responsiveness. Moreover, SB225002, a CXCR2 antagonist, caused higher cell death in TKI-resistant CML cells than TKIs. Using a mouse xenograft model, we confirmed that SB225002 suppresses tumor growth, with a prominent effect on TKI-resistant CML cells. Our findings demonstrate that IL-8 is a prognostic factor for the progression of CML. Inhibiting the CXCR2-mTOR-c-Myc cascade is a promising therapeutic strategy to overcome TKI-sensitive and TKI-insensitive CML. Thus, CXCR2 blockade is a novel therapeutic strategy to treat CML, and SB225002, a commercially available CXCR2 antagonist, might be a candidate drug that could be used to treat TKI-resistant CML.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Agents/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Drug Delivery Systems
- Drug Resistance, Neoplasm
- Female
- Humans
- Imatinib Mesylate/pharmacology
- Interleukin-8/genetics
- Interleukin-8/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Male
- Middle Aged
- Phenylurea Compounds/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Pyrimidines/pharmacology
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Triazoles/pharmacology
- Young Adult
Collapse
Affiliation(s)
- Ji-Hea Kim
- Institute of Stem Cell Research, Korea University College of Medicine, Seoul, South Korea; Department of Biomedical Science, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Seung-Jin Lee
- Institute of Stem Cell Research, Korea University College of Medicine, Seoul, South Korea; Department of Biomedical Science, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Ka-Won Kang
- Department of Internal Medicine, Anam Hospital Korea University Medical Center, Seoul, South Korea
| | - Byung-Hyun Lee
- Department of Internal Medicine, Anam Hospital Korea University Medical Center, Seoul, South Korea
| | - Yong Park
- Department of Internal Medicine, Anam Hospital Korea University Medical Center, Seoul, South Korea
| | - Byung-Soo Kim
- Department of Internal Medicine, Anam Hospital Korea University Medical Center, Seoul, South Korea.
| |
Collapse
|
10
|
Silencing hTERT attenuates cancer stem cell-like characteristics and radioresistance in the radioresistant nasopharyngeal carcinoma cell line CNE-2R. Aging (Albany NY) 2020; 12:25599-25613. [PMID: 33234740 PMCID: PMC7803545 DOI: 10.18632/aging.104167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 09/18/2020] [Indexed: 12/21/2022]
Abstract
Objective: This study aimed to explore the effect of silencing hTERT on the CSC-like characteristics and radioresistance of CNE-2R cells. Results: Silencing hTERT suppressed CNE-2R cell proliferation and increased the cell apoptosis rate and radiosensitivity in vitro. Moreover, it could also inhibit the growth of xenografts and increase the apoptosis index and radiosensitivity in vivo. Further study discovered that after silencing hTERT, telomerase activity in CNE-2R cells was markedly suppressed, along with remarkably down-regulated stem cell-related protein levels both in vitro and in vivo. Conclusion: Silencing hTERT can suppress the CSC-like characteristics of CNE-2R cells to enhance their radiosensitivity, revealing that hTERT may become a potential target for treating radioresistant NPC. Methods: An RNAi lentiviral vector specific to the hTERT gene was constructed to infect CNE-2R cells, the hTERT silencing effect was verified through qPCR and Western blot assays, and telomerase activity was detected by PCR-ELISA. Moreover, radiosensitivity in vitro was detected through colony formation assays, CCK-8 assays and flow cytometry. Tumor growth and radioresistance were also evaluated using xenograft models, while the apoptosis index in xenografts was measured through TUNEL assay. Levels of stem cell-related proteins were determined in vitro and in vivo.
Collapse
|
11
|
Cheng CL, Yang SC, Lai CY, Wang CK, Chang CF, Lin CY, Chen WJ, Lin PY, Wu HC, Ma N, Lu FL, Lu J. CXCL14 Maintains hESC Self-Renewal through Binding to IGF-1R and Activation of the IGF-1R Pathway. Cells 2020; 9:cells9071706. [PMID: 32708730 PMCID: PMC7407311 DOI: 10.3390/cells9071706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 01/04/2023] Open
Abstract
Human embryonic stem cells (hESCs) have important roles in regenerative medicine, but only a few studies have investigated the cytokines secreted by hESCs. We screened and identified chemokine (C-X-C motif) ligand 14 (CXCL14), which plays crucial roles in hESC renewal. CXCL14, a C-X-C motif chemokine, is also named as breast and kidney-expressed chemokine (BRAK), B cell and monocyte-activated chemokine (BMAC), and macrophage inflammatory protein-2γ (MIP-2γ). Knockdown of CXCL14 disrupted the hESC self-renewal, changed cell cycle distribution, and further increased the expression levels of mesoderm and endoderm differentiated markers. Interestingly, we demonstrated that CXCL14 is the ligand for the insulin-like growth factor 1 receptor (IGF-1R), and it can activate IGF-1R signal transduction to support hESC renewal. Currently published literature indicates that all receptors in the CXCL family are G protein-coupled receptors (GPCRs). This report is the first to demonstrate that a CXCL protein can bind to and activate a receptor tyrosine kinase (RTK), and also the first to show that IGF-1R has another ligand in addition to IGFs. These findings broaden our understanding of stem cell biology and signal transduction.
Collapse
Affiliation(s)
- Chih-Lun Cheng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; (C.-L.C.); (H.-C.W.)
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (S.-C.Y.); (C.-Y.L.); (C.-K.W.); (C.-F.C.); (C.-Y.L.); (W.-J.C.); (P.-Y.L.)
| | - Shang-Chih Yang
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (S.-C.Y.); (C.-Y.L.); (C.-K.W.); (C.-F.C.); (C.-Y.L.); (W.-J.C.); (P.-Y.L.)
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
| | - Chien-Ying Lai
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (S.-C.Y.); (C.-Y.L.); (C.-K.W.); (C.-F.C.); (C.-Y.L.); (W.-J.C.); (P.-Y.L.)
| | - Cheng-Kai Wang
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (S.-C.Y.); (C.-Y.L.); (C.-K.W.); (C.-F.C.); (C.-Y.L.); (W.-J.C.); (P.-Y.L.)
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
| | - Ching-Fang Chang
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (S.-C.Y.); (C.-Y.L.); (C.-K.W.); (C.-F.C.); (C.-Y.L.); (W.-J.C.); (P.-Y.L.)
| | - Chun-Yu Lin
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (S.-C.Y.); (C.-Y.L.); (C.-K.W.); (C.-F.C.); (C.-Y.L.); (W.-J.C.); (P.-Y.L.)
| | - Wei-Ju Chen
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (S.-C.Y.); (C.-Y.L.); (C.-K.W.); (C.-F.C.); (C.-Y.L.); (W.-J.C.); (P.-Y.L.)
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Po-Yu Lin
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (S.-C.Y.); (C.-Y.L.); (C.-K.W.); (C.-F.C.); (C.-Y.L.); (W.-J.C.); (P.-Y.L.)
| | - Han-Chung Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; (C.-L.C.); (H.-C.W.)
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Nianhan Ma
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City 320, Taiwan;
| | - Frank Leigh Lu
- Department of Pediatrics, National Taiwan University Children’s Hospital, National Taiwan University Hospital, and National Taiwan University Medical College, Taipei 100, Taiwan
- Correspondence: (F.L.L.); (J.L.)
| | - Jean Lu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan; (C.-L.C.); (H.-C.W.)
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (S.-C.Y.); (C.-Y.L.); (C.-K.W.); (C.-F.C.); (C.-Y.L.); (W.-J.C.); (P.-Y.L.)
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei 106, Taiwan
- National Core Facility Program for Biotechnology, National RNAi Platform, Taipei 112, Taiwan
- Department of Life Science, Tzu Chi University, Hualien 970, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: (F.L.L.); (J.L.)
| |
Collapse
|
12
|
Kang KW, Lee SJ, Kim JH, Lee BH, Kim SJ, Park Y, Kim BS. Etoposide-mediated interleukin-8 secretion from bone marrow stromal cells induces hematopoietic stem cell mobilization. BMC Cancer 2020; 20:619. [PMID: 32615949 PMCID: PMC7330970 DOI: 10.1186/s12885-020-07102-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/23/2020] [Indexed: 12/25/2022] Open
Abstract
Background We assessed the mechanism of hematopoietic stem cell (HSC) mobilization using etoposide with granulocyte-colony stimulating factor (G-CSF), and determined how this mechanism differs from that induced by cyclophosphamide with G-CSF or G-CSF alone. Methods We compared the clinical features of 173 non-Hodgkin’s lymphoma patients who underwent autologous peripheral blood stem cell transplantation (auto-PBSCT). Additionally, we performed in vitro experiments to assess the changes in human bone marrow stromal cells (hBMSCs), which support the HSCs in the bone marrow (BM) niche, following cyclophosphamide or etoposide exposure. We also performed animal studies under standardized conditions to ensure the following: exclude confounding factors, mimic the conditions in clinical practice, and identify the changes in the BM niche caused by etoposide-induced chemo-mobilization or other mobilization methods. Results Retrospective analysis of the clinical data revealed that the etoposide with G-CSF mobilization group showed the highest yield of CD34+ cells and the lowest change in white blood cell counts during mobilization. In in vitro experiments, etoposide triggered interleukin (IL)-8 secretion from the BMSCs and caused long-term BMSC toxicity. To investigate the manner in which the hBMSC-released IL-8 affects hHSCs in the BM niche, we cultured hHSCs with or without IL-8, and found that the number of total, CD34+, and CD34+/CD45- cells in IL-8-treated cells was significantly higher than the respective number in hHSCs cultured without IL-8 (p = 0.014, 0.020, and 0.039, respectively). Additionally, the relative expression of CXCR2 (an IL-8 receptor), and mTOR and c-MYC (components of IL-8-related signaling pathways) increased 1 h after IL-8 treatment. In animal studies, the etoposide with G-CSF mobilization group presented higher IL-8-related cytokine and MMP9 expression and lower SDF-1 expression in the BM, compared to the groups not treated with etoposide. Conclusion Collectively, the unique mechanism of etoposide with G-CSF-induced mobilization is associated with IL-8 secretion from the BMSCs, which is responsible for the enhanced proliferation and mobilization of HSCs in the bone marrow; this was not observed with mobilization using cyclophosphamide with G-CSF or G-CSF alone. However, the long-term toxicity of etoposide toward BMSCs emphasizes the need for the development of more efficient and safe chemo-mobilization strategies.
Collapse
Affiliation(s)
- Ka-Won Kang
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University School of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Seung-Jin Lee
- Institute of Stem Cell Research, Korea University, Seoul, South Korea.,Department of Biomedical and Science, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Ji Hye Kim
- Institute of Stem Cell Research, Korea University, Seoul, South Korea.,Department of Biomedical and Science, Graduate School of Medicine, Korea University, Seoul, South Korea
| | - Byung-Hyun Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University School of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Seok Jin Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yong Park
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University School of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Byung Soo Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University School of Medicine, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841, South Korea. .,Institute of Stem Cell Research, Korea University, Seoul, South Korea. .,Department of Biomedical and Science, Graduate School of Medicine, Korea University, Seoul, South Korea.
| |
Collapse
|
13
|
Wang L, Wang H, Wang Y, Shen M, Li S. Photocatalyzed synthesis of unsymmetrical ureas via the oxidative decarboxylation of oxamic acids with PANI-g-C3N4-TiO2 composite under visible light. Tetrahedron Lett 2020. [DOI: 10.1016/j.tetlet.2020.151962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
14
|
Chen K, Chen L, Li L, Qu S, Yu B, Sun Y, Wan F, Chen X, Liang R, Zhu X. A positive feedback loop between Wnt/β-catenin signaling and hTERT regulates the cancer stem cell-like traits in radioresistant nasopharyngeal carcinoma cells. J Cell Biochem 2020; 121:4612-4622. [PMID: 32065421 DOI: 10.1002/jcb.29681] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Radioresistance may be induced by cancer stem cells (CSCs), while the biological traits of CSCs need to be retained by telomerase. The telomerase activity mainly depends on the transcriptional regulation of human telomerase reverse transcriptase (hTERT). Moreover, Wnt/β-catenin signaling is also considered essential for maintaining the CSC phenotypes. In the previous study, we discovered that the radioresistant nasopharyngeal carcinoma cells CNE-2R displayed CSC-like traits, as well as high expression of hTERT and β-catenin, but whether hTERT and β-catenin were involved in regulating the CSC-like traits and radiosensitivity of CNE-2R cells remained unclear. In this study, our results suggested that hTERT could positively regulate the expression of CSC-related proteins, as well as the cytoplasm- and nucleus-β-catenin, but it could not markedly regulate the expression of total β-catenin in CNE-2R cells. Meanwhile, Wnt/β-catenin signaling had a positive regulatory effect on the expression of hTERT and CSC-related proteins. Moreover, there was a β-catenin/hTERT protein complex in CNE-2R cells, indicating that β-catenin could directly interact with hTERT protein. Our results also revealed that silencing hTERT or suppressing Wnt/β-catenin signaling could attenuate telomerase activity and radioresistance of CNE-2R cells; while suppressing Wnt/β-catenin signaling, the telomerase activity and radioresistance could be reversed through overexpressing hTERT. Taken together, we have outlined a positive feedback loop between Wnt/β-catenin signaling and hTERT in CNE-2R cells, which can regulate the telomerase activity and CSC-like traits, thus regulating the radiosensitivity. Therefore, blocking Wnt/β-catenin signaling transduction and interfering with hTERT expression may be a promising approach for targeting radioresistant nasopharyngeal carcinoma cells with CSC-like traits.
Collapse
Affiliation(s)
- Kaihua Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Li Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Ling Li
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.,Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China.,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Nanning, Guangxi, China
| | - Song Qu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.,Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China.,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Nanning, Guangxi, China
| | - Binbin Yu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yongchu Sun
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Fangzhu Wan
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xishan Chen
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Renba Liang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Xiaodong Zhu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.,Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Ministry of Education, Nanning, Guangxi, China.,Guangxi Key Laboratory of Early Prevention and Treatment for Regional High-Incidence-Tumor, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
15
|
Chu H, Li W, Li H. C-X-C motif chemokine receptor type 2 correlates with higher disease stages and predicts worse prognosis, and its downregulation enhances chemotherapy sensitivity in triple-negative breast cancer. Transl Cancer Res 2020; 9:840-848. [PMID: 35117429 PMCID: PMC8798293 DOI: 10.21037/tcr.2019.12.38] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 10/17/2019] [Indexed: 11/06/2022]
Abstract
BACKGROUND This study aimed to explore the correlation of C-X-C motif chemokine receptor type 2 (CXCR2) expression with tumor stage and overall survival (OS) in triple-negative breast cancer (TNBC) patients, furthermore, to investigate the influence of CXCR2 downregulation on chemotherapy sensitivity in TNBC cells. METHODS One hundred fifty-eight TNBC patients underwent surgical excision were retrospectively reviewed, and CXCR2 expression in tumor tissue was determined by immunohistochemistry (IHC). In vitro, CXCR2 shRNA and control shRNA were transfected into HCC1937 cells respectively. Doxorubicin and docetaxel with different concentrations were used to treat HCC1937 cells respectively, followed by relative cell viability (%) and IC50 measurements. RESULTS There were 87 (55.1%) patients presented with CXCR2 high expression, and 71 (44.9%) patients presented with CXCR2 low expression. CXCR2 high expression was positively associated with pathological grade (P=0.007), N stage (P<0.001) and TNM stage (P<0.001), and it predicted unfavorable OS (P<0.001). Further analysis disclosed that CXCR2 high expression independently predicted decreased OS (P=0.028). In vitro, CXCR2 shRNA increased chemosensitivity of HCC1937 cells to doxorubicin and docetaxel, with reduced IC50 concentration of doxorubicin (P<0.05) and docetaxel (P<0.01) compared the control shRNA. CONCLUSIONS CXCR2 has the potential to serve as a biomarker for assisting TNBC management and prognosis, and targeting CXCR2 provides a novel strategy to circumvent the chemotherapy resistance.
Collapse
Affiliation(s)
- Huimin Chu
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Wuhan 430014, China
| | - Wenhuan Li
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Wuhan 430014, China
| | - Hai Li
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Wuhan 430014, China
| |
Collapse
|
16
|
Lee SJ, Kang KW, Kim JH, Lee BH, Jung JH, Park Y, Hong SC, Kim BS. CXCR2 Ligands and mTOR Activation Enhance Reprogramming of Human Somatic Cells to Pluripotent Stem Cells. Stem Cells Dev 2020; 29:119-132. [DOI: 10.1089/scd.2019.0188] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Seung-Jin Lee
- Institute of Stem Cell Research, Korea University College of Medicine, Seoul, Korea
- Department of Biomedical and Science, Graduate School of Medicine, Korea University, Seoul, Korea
| | - Ka-Won Kang
- Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Ji-Hea Kim
- Institute of Stem Cell Research, Korea University College of Medicine, Seoul, Korea
- Department of Biomedical and Science, Graduate School of Medicine, Korea University, Seoul, Korea
| | - Byung-Hyun Lee
- Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Ji-Hye Jung
- Institute of Stem Cell Research, Korea University College of Medicine, Seoul, Korea
| | - Yong Park
- Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| | - Soon-Cheol Hong
- Department of Obstetrics and Gynecology, Korea University Medical Center, Seoul, Korea
| | - Byung-Soo Kim
- Institute of Stem Cell Research, Korea University College of Medicine, Seoul, Korea
- Department of Biomedical and Science, Graduate School of Medicine, Korea University, Seoul, Korea
- Department of Internal Medicine, Korea University Medical Center, Seoul, Korea
| |
Collapse
|
17
|
Song C, Xu F, Ren Z, Zhang Y, Meng Y, Yang Y, Lingadahalli S, Cheung E, Li G, Liu W, Wan J, Zhao Y, Chen G. Elevated Exogenous Pyruvate Potentiates Mesodermal Differentiation through Metabolic Modulation and AMPK/mTOR Pathway in Human Embryonic Stem Cells. Stem Cell Reports 2019; 13:338-351. [PMID: 31353224 PMCID: PMC6700476 DOI: 10.1016/j.stemcr.2019.06.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 06/21/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023] Open
Abstract
Pyruvate is a key metabolite in glycolysis and the tricarboxylic acid (TCA) cycle. Exogenous pyruvate modulates metabolism, provides cellular protection, and is essential for the maintenance of human preimplantation embryos and human embryonic stem cells (hESCs). However, little is known about how pyruvate contributes to cell-fate determination during epiblast stage. In this study, we used hESCs as a model to demonstrate that elevated exogenous pyruvate shifts metabolic balance toward oxidative phosphorylation in both maintenance and differentiation conditions. During differentiation, pyruvate potentiates mesoderm and endoderm lineage specification. Pyruvate production and its mitochondrial metabolism are required in BMP4-induced mesoderm differentiation. However, the TCA-cycle metabolites do not have the same effect as pyruvate on differentiation. Further study shows that pyruvate increases AMP/ATP ratio, activates AMPK, and modulates the mTOR pathway to enhance mesoderm differentiation. This study reveals that exogenous pyruvate not only controls metabolism but also modulates signaling pathways in hESC differentiation.
Collapse
Affiliation(s)
- Chengcheng Song
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China
| | - Faxiang Xu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China
| | - Zhili Ren
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China
| | - Yumeng Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China
| | - Ya Meng
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China; Center of Interventional Radiology, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Jinan University, Zhuhai, Guangdong 519000, China
| | - Yiqi Yang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, China
| | | | - Edwin Cheung
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, China
| | - Gang Li
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, China
| | - Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China; Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Macau, China
| | - Jianbo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yang Zhao
- The MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Macau, China; Bioimaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Macau, China.
| |
Collapse
|
18
|
Lagunas AM, Francis M, Maniar NB, Nikolova G, Wu J, Crowe DL. Paracrine Interaction of Cancer Stem Cell Populations Is Regulated by the Senescence-Associated Secretory Phenotype (SASP). Mol Cancer Res 2019; 17:1480-1492. [PMID: 31043491 DOI: 10.1158/1541-7786.mcr-18-1356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/25/2019] [Accepted: 04/25/2019] [Indexed: 11/16/2022]
Abstract
Dyskeratosis congenita is a telomere DNA damage syndrome characterized by defective telomere maintenance, bone marrow failure, and increased head and neck cancer risk. The Pot1b-/-;Terc+/- mouse exhibits some features of dyskeratosis congenita, but head and neck cancer was not reported in this model. To model the head and neck cancer phenotype, we created unique Pot1b- and p53-null-mutant models which allow genetic lineage tracing of two distinct stem cell populations. Loss of Pot1b expression depleted stem cells via ATR/Chk1/p53 signaling. Tumorigenesis was inhibited in Pot1b-/-;p53+/+ mice due to cellular senescence. Pot1b-/-;p53-/- tumors also exhibited senescence, but proliferated and metastasized with expansion of Lgr6+ stem cells indicative of senescence-associated secretory phenotype. Selective depletion of the small K15+ stem cell fraction resulted in reduction of Lgr6+ cells and inhibition of tumorigenesis via senescence. Gene expression studies revealed that K15+ cancer stem cells regulate Lgr6+ cancer stem cell expansion via chemokine signaling. Genetic ablation of the chemokine receptor Cxcr2 inhibited cancer stem cell expansion and tumorigenesis via senescence. The effects of chemokines were primarily mediated by PI3K signaling, which is a therapeutic target in head and neck cancer. IMPLICATIONS: Paracrine interactions of cancer stem cell populations impact therapeutic options and patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Jianchun Wu
- University of Illinois Cancer Center, Chicago, Illinois
| | - David L Crowe
- University of Illinois Cancer Center, Chicago, Illinois.
| |
Collapse
|
19
|
Abstract
Mammalian target of rapamycin (mTOR) is a conserved serine/threonine kinase of the phosphatidylinositol kinase-related kinase family that regulates cell growth, metabolism, and autophagy. Extensive research has linked mTOR to several human diseases including cancer, neurodegenerative disorders, and aging. In this review, recent publications regarding the mechanisms underlying the role of mTOR in female reproduction under physiological and pathological conditions are summarized. Moreover, we assess whether strategies to improve or suppress mTOR expression could have therapeutic potential for reproductive diseases like premature ovarian failure, polycystic ovarian syndrome, and endometriosis.
Collapse
|
20
|
Wang L, Wang H, Li G, Min S, Xiang F, Liu S, Zheng W. Pd/C-Catalyzed Domino Synthesis of Urea Derivatives Using Chloroform as the Carbon Monoxide Source in Water. Adv Synth Catal 2018. [DOI: 10.1002/adsc.201800954] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Liang Wang
- School of Petrochemical Engineering; Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology; Changzhou University, Gehu Raod 1, Wujin, Changzhou; 213164 People's Republic of China
| | - Hao Wang
- School of Petrochemical Engineering; Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology; Changzhou University, Gehu Raod 1, Wujin, Changzhou; 213164 People's Republic of China
| | - Guiqing Li
- School of Petrochemical Engineering; Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology; Changzhou University, Gehu Raod 1, Wujin, Changzhou; 213164 People's Republic of China
| | - Shuliang Min
- School of Petrochemical Engineering; Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology; Changzhou University, Gehu Raod 1, Wujin, Changzhou; 213164 People's Republic of China
| | - Fangyuan Xiang
- School of Petrochemical Engineering; Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology; Changzhou University, Gehu Raod 1, Wujin, Changzhou; 213164 People's Republic of China
| | - Shiqi Liu
- School of Petrochemical Engineering; Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology; Changzhou University, Gehu Raod 1, Wujin, Changzhou; 213164 People's Republic of China
| | - Waigang Zheng
- School of Petrochemical Engineering; Jiangsu Key Laboratory of Advanced Catalytic Materials & Technology; Changzhou University, Gehu Raod 1, Wujin, Changzhou; 213164 People's Republic of China
| |
Collapse
|
21
|
Abstract
The mammalian target of rapamycin (mTOR) senses nutrients and growth factors to coordinate cell growth, metabolism and autophagy. Extensive research has mapped the signaling pathways regulated by mTOR that are involved in human diseases, such as cancer, and in diabetes and ageing. Recently, however, new studies have demonstrated important roles for mTOR in promoting the differentiation of adult stem cells, driving the growth and proliferation of stem and progenitor cells, and dictating the differentiation program of multipotent stem cell populations. Here, we review these advances, providing an overview of mTOR signaling and its role in murine and human stem and progenitor cells.
Collapse
Affiliation(s)
- Delong Meng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anderson R Frank
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jenna L Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA .,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|