1
|
Kobayashi G, Sekino Y, Nakahara H, Kobatake K, Goto K, Hayashi T, Sentani K, Hinata N. Distribution and clinicopathological characteristics of G-CSF expression in tumor cells and stromal cells in upper tract urothelial carcinoma. J Cancer Res Clin Oncol 2024; 151:18. [PMID: 39739128 DOI: 10.1007/s00432-024-06045-1] [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: 10/28/2024] [Accepted: 11/21/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Urothelial carcinoma (UC) is a common type of malignant disease; however, the diagnostic and prognostic markers of upper urinary tract urothelial cancer (UTUC) remain poorly understood because of its rarity. METHODS To clarify the clinicopathological significance of granulocyte-colony stimulating factor (G-CSF) in UTUC, we analyzed the expression and distribution of G-CSF in 112 upper tract urothelial carcinoma (UTUC) samples with immunohistochemistry. RESULTS In normal urothelium, G-CSF expression was weak or absent, whereas high expression of G-CSF was observed in UTUC tissues, both in tumor cells (TCs) and stromal cells (SCs). G-CSF expression in the TCs and SCs was associated with nodular/flat morphology, high grade, advanced T stage, and lymphovascular invasion in UTUC. G-CSF expression in SCs was associated with poor prognosis and was an independent prognostic factor. Public data showed that G-CSF expression was also associated with decreased progression-free survival and disease-specific survival. A prognostic model was constructed by incorporating the presence or absence of G-CSF expression along with clinicopathologic factors, which allowed for a more accurate prediction of poor prognosis. We further showed that G-CSF expression was associated with a high Ki-67 labeling index and with PD-L1, HER2, and p53 expression in UTUC. CONCLUSION G-CSF expression in TCs and SCs may play a crucial role in UTUC tumor progression. Notably, stromal G-CSF expression showed significant prognostic value, even when compared to major clinicopathological factors, suggesting that the evaluation of G-CSF expression may contribute to clinical decision-making in patients with UTUC.
Collapse
Affiliation(s)
- Go Kobayashi
- Laboratory of Molecular Pathology, Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yohei Sekino
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Hikaru Nakahara
- Department of Clinical and Molecular Genetics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kohei Kobatake
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Keisuke Goto
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Tetsutaro Hayashi
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kazuhiro Sentani
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuyuki Hinata
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
2
|
Mahdizadeh M, Karimi MA, Tajabadi Z, Kaveh V, Zamani S. The impact of delayed versus early administration of granulocyte colony-stimulating factor following autologous hematopoietic stem cell transplantation on transplantation outcome. Front Oncol 2024; 14:1468948. [PMID: 39411138 PMCID: PMC11473490 DOI: 10.3389/fonc.2024.1468948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Objectives Granulocyte colony-stimulating factor (G-CSF) is routinely administered after autologous hematopoietic stem cell transplantation (auto-HSCT) to decrease the duration of neutropenia and diminish the incidence of febrile neutropenia. Nevertheless, the most advantageous timeframe for administering G-CSF in the transplantation setting remains elusive. Material and Methods We conducted a cross-sectional study of 200 patients diagnosed with hematological malignancies who underwent auto-HSCT between July 2017 and January 2022. Patients were divided into two groups of 100 individuals based on the timing of G-CSF administration after auto-HSCT. In the first group, G-CSF was administered on post-transplantation day +1, while in the second group, G-CSF was administered on post-transplantation day +5. Patient demographics and clinical outcomes, including time to neutrophil engraftment, time to platelet engraftment, length of hospital stay, duration of fever, and incidence of bacterial and fungal bloodstream infections, were compared between the two groups. Results We identified a significantly shorter platelet engraftment time in the day +5 group than in the day +1 group (P<0.001), though the groups were similar regarding neutrophil engraftment time. The total number of G-CSF injections differed significantly according to the administration schedule. The number of red blood cells and length of hospital stay was greater in the day +1 group (all P<0.001). The incidence of bacterial and fungal bloodstream infections and duration of fever did not differ between the groups. Conclusion Delayed administration of G-CSF on day +5 is as effective as early administration and can positively influence platelet engraftment, transfusion support, and hospitalization time.
Collapse
Affiliation(s)
- Mahshid Mahdizadeh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Taleghani Hospital Clinical Research Development Unit, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Karimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohreh Tajabadi
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Kaveh
- Hematology and Oncology Department, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Zamani
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Deng J, Tan Y, Xu Z, Wang H. Advances in hematopoietic stem cells ex vivo expansion associated with bone marrow niche. Ann Hematol 2024:10.1007/s00277-024-05773-1. [PMID: 38684510 DOI: 10.1007/s00277-024-05773-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
Hematopoietic stem cells (HSCs) are an ideal source for the treatment of many hematological diseases and malignancies, as well as diseases of other systems, because of their two important features, self-renewal and multipotential differentiation, which have the ability to rebuild the blood system and immune system of the body. However, so far, the insufficient number of available HSCs, whether from bone marrow (BM), mobilized peripheral blood or umbilical cord blood, is still the main restricting factor for the clinical application. Therefore, strategies to expand HSCs numbers and maintain HSCs functions through ex vivo culture are urgently required. In this review, we outline the basic biology characteristics of HSCs, and focus on the regulatory factors in BM niche affecting the functions of HSCs. Then, we introduce several representative strategies used for HSCs from these three sources ex vivo expansion associated with BM niche. These findings have deepened our understanding of the mechanisms by which HSCs balance self-renewal and differentiation and provided a theoretical basis for the efficient clinical HSCs expansion.
Collapse
Affiliation(s)
- Ju Deng
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Tan
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhifang Xu
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongwei Wang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
4
|
Higazy D, Pham AD, van Hasselt C, Høiby N, Jelsbak L, Moser C, Ciofu O. In vivo evolution of antimicrobial resistance in a biofilm model of Pseudomonas aeruginosa lung infection. THE ISME JOURNAL 2024; 18:wrae036. [PMID: 38478426 PMCID: PMC10980832 DOI: 10.1093/ismejo/wrae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/10/2024] [Accepted: 02/29/2024] [Indexed: 04/01/2024]
Abstract
The evolution of antimicrobial resistance (AMR) in biofilms has been repeatedly studied by experimental evolution in vitro, but rarely in vivo. The complex microenvironment at the infection site imposes selective pressures on the bacterial biofilms, potentially influencing the development of AMR. We report here the development of AMR in an in vivo mouse model of Pseudomonas aeruginosa biofilm lung infection. The P. aeruginosa embedded in seaweed alginate beads underwent four successive lung infection passages with or without ciprofloxacin (CIP) exposure. The development of CIP resistance was assessed at each passage by population analysis of the bacterial populations recovered from the lungs of CIP-treated and control mice, with subsequent whole-genome sequencing of selected isolates. As inflammation plays a crucial role in shaping the microenvironment at the infection site, its impact was explored through the measurement of cytokine levels in the lung homogenate. A rapid development of AMR was observed starting from the second passage in the CIP-treated mice. Genetic analysis revealed mutations in nfxB, efflux pumps (mexZ), and two-component systems (parS) contribution to CIP resistance. The control group isolates exhibited mutations in the dipA gene, likely associated with biofilm dispersion. In the initial two passages, the CIP-treated group exhibited an elevated inflammatory response compared to the control group. This increase may potentially contribute to the release of mutagenic reactive oxygen species and the development of AMR. In conclusion, this study illustrates the complex relationship between infection, antibiotic treatment, and immune response.
Collapse
Affiliation(s)
- Doaa Higazy
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 N Copenhagen, Denmark
- Department of Microbiology, Faculty of Agriculture, Cairo University, 12613 Giza, Egypt
- Department of Clinical Microbiology, Rigshospitalet, University of Copenhagen, 2100 Ø Copenhagen, Denmark
| | - Anh Duc Pham
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, The Netherlands
| | - Coen van Hasselt
- Division of Systems Pharmacology & Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, The Netherlands
| | - Niels Høiby
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 N Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, University of Copenhagen, 2100 Ø Copenhagen, Denmark
| | - Lars Jelsbak
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Claus Moser
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 N Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, University of Copenhagen, 2100 Ø Copenhagen, Denmark
| | - Oana Ciofu
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, 2200 N Copenhagen, Denmark
| |
Collapse
|
5
|
Vimali J, Yong YK, Murugesan A, Vishnupriya K, Ashwin R, Daniel EA, Balakrishnan P, Raju S, Rosmawati M, Velu V, Larsson M, Shankar EM. Plasma interleukin-7 correlation with human immunodeficiency virus RNA and CD4+ T cell counts, and interleukin-5 with circulating hepatitis B virus DNA may have implications in viral control. Front Med (Lausanne) 2022; 9:1019230. [PMID: 36405584 PMCID: PMC9668853 DOI: 10.3389/fmed.2022.1019230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/12/2022] [Indexed: 08/30/2023] Open
Abstract
Chronic viral infections represent a leading cause of global morbidity and mortality. Chronic HBV, HCV, and HIV infections result in cytokine perturbations that may hold key implications in understanding the complex disease mechanisms driving virus persistence and/or resolution. Here, we determined the levels of various plasma cytokines using a commercial Bio-Plex Luminex cytokine array in chronic HBV (n = 30), HCV (n = 15), and HIV (n = 40) infections and correlated with corresponding plasma viral loads (PVLs) and liver parameters. We observed differential perturbations in cytokine profiles among the study groups. The cytokines levels positively correlated with PVL and liver transaminases. The monocyte-derived cytokines viz., MIP-1β, IL-8, and TNF-α, and Th2 cytokines like IL-4, IL-5, and IL-13 showed a better correlation with liver enzymes as compared to their corresponding PVLs. Our investigation also identified two cytokines viz., IL-5 and IL-7 that inversely correlated with HBV DNA and HIV PVLs, respectively. Regression analysis adjusted for age showed that every increase of IL-5 by one unit was associated with a reduction in HBV PVL by log10 0.4, whereas, every elevation by a unit of IL-7 was associated with decreased HIV PVL by log10 2.5. We also found that IL-7 levels correlated positively with absolute CD4+ T cell counts in HIV-infected patients. We concluded that plasma IL-5 and IL-7 may likely have a key role on viral control in HBV and HIV infections, respectively. A noteworthy increase in cytokines appears to bear protective and pathological significance, and indeed is reflective of the host's versatile immune armory against viral persistence.
Collapse
Affiliation(s)
- Jaisheela Vimali
- Infection Biology, Department of Biotechnology, Central University of Tamil Nadu, Thiruvarur, India
| | - Yean Kong Yong
- Laboratory Centre, Xiamen University Malaysia, Sepang, Selangor, Malaysia
| | - Amudhan Murugesan
- Department of Microbiology, Government Theni Medical College and Hospital, Theni, India
| | | | - Rajeev Ashwin
- Infection Biology, Department of Biotechnology, Central University of Tamil Nadu, Thiruvarur, India
| | - Evangeline Ann Daniel
- National Institute for Research in Tuberculosis, Indian Council of Medical Research, Chennai, India
| | - Pachamuthu Balakrishnan
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Sivadoss Raju
- State Public Health Laboratory, Directorate of Public Health and Preventive Medicine, Chennai, India
| | - Mohamed Rosmawati
- Division of Gastroenterology and Hepatology, Department of Medicine, Faculty of Medicine, Universiti Malaya Medical Center, Kuala Lumpur, Malaysia
| | - Vijayakumar Velu
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Biomedicine and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Esaki M. Shankar
- Infection Biology, Department of Biotechnology, Central University of Tamil Nadu, Thiruvarur, India
| |
Collapse
|
6
|
Lee JU, Choi JS, Kim MK, Min SA, Park JS, Park CS. Granulocyte colony-stimulating factor in bronchoalveolar lavage fluid is a potential biomarker for prognostic prediction of idiopathic pulmonary fibrosis. Korean J Intern Med 2022; 37:979-988. [PMID: 35730133 PMCID: PMC9449205 DOI: 10.3904/kjim.2021.442] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/25/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS Neutrophilia is frequently observed in bronchoalveolar lavage fluid (BALF) of idiopathic pulmonary fibrosis (IPF) patients. Granulocyte colony-stimulating factor (G-CSF) is a potent neutrophil-activating glycoprotein. However, the clinical implications of G-CSF remain poorly understood.in patients with IPF. Therefore, we evaluated the relationship between the G-CSF concentration in BALF and the progression of fibrosis, including in terms of the decline in lung function and long-term survival rate. METHODS G-CSF concentrations were measured in BALF using enzyme-linked immunosorbent assay (ELISA). The survival rate was estimated using Kaplan-Meier survival analyses. RESULTS G-CSF protein levels were significantly higher in IPF (n = 87; 1.88 [0 to 5.68 pg/mL]), nonspecific interstitial pneumonia (n = 22; 0.58 [0 to 11.64 pg/mL]), and hypersensitivity pneumonitis (n = 19; 2.48 [0.46 to 5.71 pg/mL]) patients than in normal controls (n = 33; 0 [0 to 0.68 pg/mL]) (all p < 0.01). A receiver operating characteristic curve showed a difference in G-CSF levels between IPF and NC (area under the curve, 0.769): The G-CSF cut-off of 0.96 pg/mL indicated 84.9% specificity and 63.2% sensitivity for IPF. The survival rate was significantly lower in the group with G-CSF > 2.872 pg/mL than in the group with ≤ 2.872 pg/mL (hazard ratio, 2.69; p = 0.041). The annual decline in diffusing capacity of the lung for carbon monoxide was positively correlated with the G-CSF level (p = 0.018). CONCLUSION G-CSF may participate in the development of IPF and be useful for predicting the prognosis of IPF. Therefore, G-CSF should be analyzed in BALF, in addition to differential cell counts.
Collapse
Affiliation(s)
- Jong-Uk Lee
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang University, Bucheon,
Korea
| | - Jae Sung Choi
- Division of Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan,
Korea
| | - Min Kyung Kim
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang University, Bucheon,
Korea
| | - Sun A Min
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang University, Bucheon,
Korea
| | - Jong-Sook Park
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon,
Korea
- PulmoBioPark Co. Ltd., Soonchunhyang University Bucheon Hospital, Bucheon,
Korea
| | - Choon-Sik Park
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon,
Korea
- PulmoBioPark Co. Ltd., Soonchunhyang University Bucheon Hospital, Bucheon,
Korea
| |
Collapse
|
7
|
Mousavi Motlagh SS, Seyedhamzeh M, Ahangari Cohan R, Shafiee Ardestani M, Vaziri B, Azadmanesh K, Saberi S, Masoumi V. Novel G-CSF conjugated anionic globular dendrimer: Preparation and biological activity assessment. Pharmacol Res Perspect 2021; 9:e00826. [PMID: 34269522 PMCID: PMC8283867 DOI: 10.1002/prp2.826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 11/18/2022] Open
Abstract
The most crucial role of granulocyte colony-stimulating factor (G-CSF) in the body is to increase the strength of immune system. In recent years, research on the use of nanoparticles in pharmaceuticals has been considered, most of which have been for drug-loading purposes. In this study, a novel G-CSF conjugated dendrimer was synthesized and characterized using different techniques. In vitro cytotoxicity was assessed on A549 and L929 cells, while abnormal toxicity was studied in mice. In vitro and in vivo biological activities were assessed in NFS60 cells and rats, respectively. In addition, in vivo distribution, plasma half-life, and histopathological effect were studied in rat. The characterization tests confirmed the successful conjugation. There was no difference between G-CSF cytotoxicity before and after conjugation, and no difference with the control group. No mice showed abnormal toxicity. Although in vitro biological activity revealed both conjugated and free G-CSF promote proliferation cells, biological activity decreased significantly after conjugation about one-third of the unconjugated form. Nonetheless, in vivo biological activity of conjugated G-CSF increased by more than 2.5-fold relative to the unconjugated form, totally. Fortunately, no histopathologic adverse effect was observed in vital rat tissues. Also, in vivo distribution of the conjugate was similar to the native protein with an enhanced terminal half-life. Our data revealed that G-CSF conjugated dendrimer could be considered as a candidate to improve the in vivo biological activity of G-CSF. Moreover, multivalent capability of the dendrimer may be used for other new potentials of G-CSF in future perspectives.
Collapse
Affiliation(s)
| | | | - Reza Ahangari Cohan
- Department of NanobiotechnologyNew Technologies Research GroupPasteur Institute of IranTehranIran
| | | | - Behrouz Vaziri
- Biotechnology Research CenterPasteur Institute of IranTehranIran
| | | | - Sahar Saberi
- Department of Biotechnology, Food and Drug Control LaboratoriesNational Food and Drug OrganizationTehranIran
| | - Vahideh Masoumi
- Department of Biotechnology, Food and Drug Control LaboratoriesNational Food and Drug OrganizationTehranIran
| |
Collapse
|
8
|
Theron AJ, Steel HC, Rapoport BL, Anderson R. Contrasting Immunopathogenic and Therapeutic Roles of Granulocyte Colony-Stimulating Factor in Cancer. Pharmaceuticals (Basel) 2020; 13:ph13110406. [PMID: 33233675 PMCID: PMC7699711 DOI: 10.3390/ph13110406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
Tumor cells are particularly adept at exploiting the immunosuppressive potential of neutrophils as a strategy to achieve uncontrolled proliferation and spread. Recruitment of neutrophils, particularly those of an immature phenotype, known as granulocytic myeloid-derived suppressor cells, is achieved via the production of tumor-derived granulocyte colony-stimulating factor (G-CSF) and neutrophil-selective chemokines. This is not the only mechanism by which G-CSF contributes to tumor-mediated immunosuppression. In this context, the G-CSF receptor is expressed on various cells of the adaptive and innate immune systems and is associated with induction of T cell polarization towards the Th2 and regulatory T cell (Treg) phenotypes. In contrast to the potentially adverse effects of sustained, endogenous production of G-CSF by tumor cells, stringently controlled prophylactic administration of recombinant (r) G-CSF is now a widely practiced strategy in medical oncology to prevent, and in some cases treat, chemotherapy-induced severe neutropenia. Following an overview of the synthesis, structure and function of G-CSF and its receptor, the remainder of this review is focused on: (i) effects of G-CSF on the cells of the adaptive and innate immune systems; (ii) mechanisms by which this cytokine promotes tumor progression and invasion; and (iii) current clinical applications and potential risks of the use of rG-CSF in medical oncology.
Collapse
Affiliation(s)
- Annette J. Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (H.C.S.); (B.L.R.); (R.A.)
- Correspondence: ; Tel.: +27-12-319-2355
| | - Helen C. Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (H.C.S.); (B.L.R.); (R.A.)
| | - Bernardo L. Rapoport
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (H.C.S.); (B.L.R.); (R.A.)
- The Medical Oncology Centre of Rosebank, Johannesburg 2196, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (H.C.S.); (B.L.R.); (R.A.)
| |
Collapse
|
9
|
Dewi KS, Fuad AM. Improving the Expression of Human Granulocyte Colony Stimulating Factor in Escherichia coli by Reducing the GC-content and Increasing mRNA Folding Free Energy at 5'-Terminal End. Adv Pharm Bull 2020; 10:610-616. [PMID: 33072539 PMCID: PMC7539321 DOI: 10.34172/apb.2020.073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/24/2019] [Accepted: 01/27/2020] [Indexed: 01/16/2023] Open
Abstract
Purpose: Strategy for improving the production of biopharmaceutical protein continues to develop due to increasing market demand. Human granulocyte colony stimulating factor (hG-CSF) is one of biopharmaceutical proteins that has many applications, and easily produced in Escherichia coli expression system. Previous studies reported that codon usage, rare codon, mRNA folding and GC-content at 5’-terminal end were crucial for protein production in E. coli. In the present study, the effect of reducing the GC-content and increasing the mRNA folding free energy at the 5’-terminal end on the expression level of hG-CSF proteins was investigated. Methods: Synonymous codon substitutions were performed to generate mutant variants of open reading frame (ORF) with lower GC-content at 5’-terminal ends. Oligoanalyzer tool was used to calculate the GC content of eight codons sequence after ATG. Whereas, mRNA folding free energy was predicted using KineFold and RNAfold tools. The template DNA was amplified using three variant forward primers and one same reverse primer. Those DNA fragments were individually cloned into pJexpress414 expression vector and were confirmed using restriction and DNA sequencing analyses. The confirmed constructs were transformed into E. coli NiCo21(DE3) host cells and the recombinant protein was expressed using IPTG-induction. Total protein obtained were characterized using SDS-PAGE, Western blot and ImageJ software analyses. Results: The result showed that the mutant variant with lower GC-content and higher mRNA folding free energy near the translation initiation region (TIR) could produce a higher amount of hG-CSF proteins compared to the original gene sequence. Conclusion: This study emphasized the important role of the nucleotide composition immediately downstream the start codon to achieve high-yield protein product on heterologous expression in E. coli.
Collapse
Affiliation(s)
- Kartika Sari Dewi
- Research Center for Biotechnology, Indonesian Institute of Sciences, Cibinong, Bogor, Indonesia, 16911
| | - Asrul Muhamad Fuad
- Research Center for Biotechnology, Indonesian Institute of Sciences, Cibinong, Bogor, Indonesia, 16911
| |
Collapse
|
10
|
Xu X, Gao J, Dai W, Wang D, Wu J, Wang J. Gene activation by a CRISPR-assisted trans enhancer. eLife 2019; 8:45973. [PMID: 30973327 PMCID: PMC6478495 DOI: 10.7554/elife.45973] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/10/2019] [Indexed: 12/15/2022] Open
Abstract
The deactivated CRISPR/Cas9 (dCas9) is now the most widely used gene activator. However, current dCas9-based gene activators are still limited by their unsatisfactory activity. In this study, we developed a new strategy, the CRISPR-assisted trans enhancer, for activating gene expression at high efficiency by combining dCas9-VP64/sgRNA with the widely used strong CMV enhancer. In this strategy, CMV enhancer DNA was recruited to target genes in trans by two systems: dCas9-VP64/csgRNA-sCMV and dCas9-VP64-GAL4/sgRNA-UAS-CMV. The former recruited trans enhancer by annealing between two short complementary oligonucleotides at the ends of the sgRNA and trans enhancer. The latter recruited trans enhancer by binding between GAL4 fused to dCas9 and UAS sequence of trans enhancer. The trans enhancer activated gene transcription as the natural looped cis enhancer. The trans enhancer could activate both exogenous reporter genes and variant endogenous genes in various cells, with much higher activation efficiency than that of current dCas9 activators.
Collapse
Affiliation(s)
- Xinhui Xu
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China
| | - Jinliang Gao
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China
| | - Wei Dai
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China
| | - Danyang Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China
| | - Jian Wu
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China
| | - Jinke Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China
| |
Collapse
|
11
|
Nold C, Stone J, O'Hara K, Davis P, Kiveliyk V, Blanchard V, Yellon SM, Vella AT. Block of Granulocyte-Macrophage Colony-Stimulating Factor Prevents Inflammation-Induced Preterm Birth in a Mouse Model for Parturition. Reprod Sci 2018; 26:551-559. [PMID: 30296925 DOI: 10.1177/1933719118804420] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE A multitude of factors promotes inflammation in the reproductive tract leading to preterm birth. Macrophages peak in the cervix prior to birth and their numbers are increased by the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF). We hypothesize GM-CSF is produced from multiple sites in the genital tract and is a key mediator in preterm birth. STUDY DESIGN Ectocervical, endocervical, and amniotic fluid mesenchymal stem cells were treated with lipopolysaccharide (LPS), and the concentration and expression of GM-CSF was measured. Pregnant CD-1 mice on gestational day 17 received LPS and an intravenous injection of either anti-mouse GM-CSF or control antibody. After 6 hours, the preterm birth rate was recorded. RESULTS Treatment with LPS increased the GM-CSF concentration and messenger RNA expression after 24 hours in all 3 cell lines ( P < .01). Mice treated with LPS and the GM-CSF antibody had a preterm birth rate of 25%, compared to a 66.7% preterm birth rate in controls, within 6 hours ( P < .05, χ2). Treatment with the anti-mouse GM-CSF antibody decreased the concentration of GM-CSF in the mouse serum ( P < .01) but did not alter the number of macrophages or collagen content in the cervix. CONCLUSION These studies demonstrate that GM-CSF is produced from multiple sites in the genital tract and that treatment with an antibody to GM-CSF prevents preterm birth. Curiously, the anti-mouse GM-CSF antibody did not decrease the number of macrophages in the cervix. Further research is needed to determine whether antibodies to GM-CSF can be utilized as a therapeutic agent to prevent preterm birth.
Collapse
Affiliation(s)
- Christopher Nold
- 1 Department of Women's Health, Hartford Hospital, Hartford, CT, USA.,2 Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Julie Stone
- 2 Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Kathleen O'Hara
- 2 Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Patricia Davis
- 2 Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Vladislav Kiveliyk
- 2 Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Vanessa Blanchard
- 3 Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Steven M Yellon
- 3 Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Anthony T Vella
- 2 Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
12
|
Dubnika A, Manoukian MA, Mohammadi MR, Parekh MB, Gurjarpadhye AA, Inayathullah M, Dubniks V, Lakey JR, Rajadas J. Cytokines as therapeutic agents and targets in heart disease. Cytokine Growth Factor Rev 2018; 43:54-68. [DOI: 10.1016/j.cytogfr.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/01/2018] [Accepted: 08/13/2018] [Indexed: 02/02/2023]
|
13
|
Weston BR, Li L, Tyson JJ. Mathematical Analysis of Cytokine-Induced Differentiation of Granulocyte-Monocyte Progenitor Cells. Front Immunol 2018; 9:2048. [PMID: 30279691 PMCID: PMC6153365 DOI: 10.3389/fimmu.2018.02048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 08/20/2018] [Indexed: 01/01/2023] Open
Abstract
Granulocyte-monocyte progenitor (GMP) cells play a vital role in the immune system by maturing into a variety of white blood cells, including neutrophils and macrophages, depending on exposure to cytokines such as various types of colony stimulating factors (CSF). Granulocyte-CSF (G-CSF) induces granulopoiesis and macrophage-CSF (M-CSF) induces monopoiesis, while granulocyte/macrophage-CSF (GM-CSF) favors monocytic and granulocytic differentiation at low and high concentrations, respectively. Although these differentiation pathways are well documented, the mechanisms behind the diverse behavioral responses of GMP cells to CSFs are not well understood. In this paper, we propose a mechanism of interacting CSF-receptors and transcription factors that control GMP differentiation, convert the mechanism into a set of differential equations, and explore the properties of this mathematical model using dynamical systems theory. Our model reproduces numerous experimental observations of GMP cell differentiation in response to varying dosages of G-CSF, M-CSF, and GM-CSF. In particular, we are able to reproduce the concentration-dependent behavior of GM-CSF induced differentiation, and propose a mechanism driving this behavior. In addition, we explore the differentiation of a fourth phenotype, monocytic myeloid-derived suppressor cells (M-MDSC), showing how they might fit into the classical pathways of GMP differentiation and how progenitor cells can be primed for M-MDSC differentiation. Finally, we use the model to make novel predictions that can be explored by future experimental studies.
Collapse
Affiliation(s)
- Bronson R Weston
- Program in Genetics, Bioinformatics, and Computational Biology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Liwu Li
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - John J Tyson
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
14
|
IL-35 induces N2 phenotype of neutrophils to promote tumor growth. Oncotarget 2018; 8:33501-33514. [PMID: 28432279 PMCID: PMC5464885 DOI: 10.18632/oncotarget.16819] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/24/2017] [Indexed: 12/18/2022] Open
Abstract
IL-35 is an immunosuppressive cytokine and exerts regulatory effects on T cells, B cells, macrophages and dendritic cells. Neutrophils are important innate immune cells that play key roles in tumor development. The effect of IL-35 on neutrophils remains unknown. Here, we report that IL-35 can induce N2 neutrophil polarization (protumor phenotype) by increasing G-CSF and IL-6 production, and promote neutrophil infiltration into tumor microenvironment. The sustained expression of IL-35 could promote chronic inflammation to augment the proangiogenic and immunosuppressive function of neutrophils. IL-35 stimulated macrophages to secrete proinflammatory cytokines IL-1β and IL-6. IL-1β stimulated γδ T cells to produce IL-17, which in turn increased the production of G-CSF. By increasing the expression of G-CSF and IL-6, IL-35 could up-regulate the expression of MMP-9 and Bv8, and down-regulate TRAIL expression in neutrophils, thus augmenting the proangiogenic function of neutrophils. Moreover, G-CSF/IL-6 induced the enhanced activation of STAT3 and ERK pathways in neutrophils, thus increasing the expression of iNOS to suppress T cell activation. Our findings suggest that IL-35 can promote tumor progression by functioning as an up-stream cytokine to promote cancer-associated inflammation and control neutrophil polarization. Targeting IL-35 might be an important approach for designing new strategy of tumor therapy.
Collapse
|
15
|
Calf Spleen Extractive Injection protects mice against cyclophosphamide-induced hematopoietic injury through G-CSF-mediated JAK2/STAT3 signaling. Sci Rep 2017; 7:8402. [PMID: 28827748 PMCID: PMC5566473 DOI: 10.1038/s41598-017-08970-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/20/2017] [Indexed: 12/22/2022] Open
Abstract
Calf Spleen Extractive Injection (CSEI), extracted from the spleen of healthy cows (within 24 hours of birth), is a small-peptide-enriched extraction and often used as an ancillary agent in cancer therapy. This study evaluated the hematopoietic function of CSEI and its underlying mechanisms, principally in CHRF, K562 cells, BMNCs and a mouse model of cyclophosphamide (CTX)-induced hematopoietic suppression. CSEI promoted the proliferation and differentiation of CHRF and K562 cells, activated hematopoietic- and proliferation-related factors RSK1p90, ELK1 and c-Myc, and facilitated the expression of differentiation- and maturation-related transcription factors GATA-1, GATA-2. In the mice with hematopoietic suppression, 3 weeks of CSEI administration enhanced the bodyweights and thymus indices, suppressed the spleen indices and strongly elevated the production of HSPCs, neutrophils and B cells in bone marrow, ameliorated bone marrow cellularity, and regulated the ratio of peripheral blood cells. Proteome profiling combined with ELISA revealed that CSEI regulated the levels of cytokines, especially G-CSF and its related factors, in the spleen and plasma. Additional data revealed that CSEI promoted phosphorylation of STAT3, which was stimulated by G-CSF in both mice spleen and cultured BMNCs. Taken together, CSEI has the potential to improve hematopoietic function via the G-CSF-mediated JAK2/STAT3 signaling pathway.
Collapse
|
16
|
López C, Zamorano P, Teuber S, Salas M, Otth C, Hidalgo MA, Concha I, Zambrano A. Interleukin-3 Prevents Cellular Death Induced by Oxidative Stress in HEK293 Cells. J Cell Biochem 2017; 118:1330-1340. [PMID: 27862234 DOI: 10.1002/jcb.25790] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/08/2016] [Indexed: 11/07/2022]
Abstract
Interleukin-3 (IL-3) is a well-characterized growth factor in hematopoietic cells, but it is also expressed in other cell types with poorly described functions. Many studies have provided evidence that IL-3 plays an important role in cell survival. We have previously shown that IL-3 is able to increase glucose uptake in HEK293 cells, suggesting that this factor requires sustained glucose metabolism to promote cell survival. In this study, we demonstrate that IL-3 contributes to cell survival under oxidative stress, a prominent feature in the pathophysiology of cancer, diabetes, and neurodegenerative diseases, as well as in the aging process. Our results suggest a molecular mechanism that involves signaling pathways mediated by PI-3k/Akt and Erk. Altogether, these findings show an important role for IL-3 in supporting the viability of non-hematopoietic systems. J. Cell. Biochem. 118: 1330-1340, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Camila López
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Patricia Zamorano
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Stefanie Teuber
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Mónica Salas
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Carola Otth
- Facultad de Medicina, Instituto de Microbiología Clínica, Valdivia, Los Ríos, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - María A Hidalgo
- Facultad de Ciencias Veterinarias, Instituto de Farmacología, Universidad Austral de Chile, Valdivia, Los Ríos, Chile
| | - Ilona Concha
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile
| | - Angara Zambrano
- Facultad de Ciencias, Instituto de Bioquímica y Microbiología, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
17
|
Ge F, Zhang Z, Hou J, Cao F, Zhang Y, Wang P, Wei H, Zhou J. Granulocyte colony-stimulating factor decreases the Th1/Th2 ratio in peripheral blood mononuclear cells from patients with chronic immune thrombocytopenic purpura in vitro. Thromb Res 2016; 148:76-84. [DOI: 10.1016/j.thromres.2016.10.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 10/06/2016] [Accepted: 10/26/2016] [Indexed: 01/06/2023]
|
18
|
Luo J, Feng XX, Luo C, Wang Y, Li D, Shu Y, Wang SS, Qin J, Li YC, Zou JM, Tian DA, Zhang GM, Feng ZH. 14,15-EET induces the infiltration and tumor-promoting function of neutrophils to trigger the growth of minimal dormant metastases. Oncotarget 2016; 7:43324-43336. [PMID: 27270316 PMCID: PMC5190026 DOI: 10.18632/oncotarget.9709] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/13/2016] [Indexed: 12/16/2022] Open
Abstract
Infiltrating neutrophils are known to promote in the development of tumor. However, it is unclear whether and how neutrophils are involved in triggering the growth of dormant metastases. Here we show that 14,15-epoxyeicosatrienoic acid (14,15-EET) can trigger the growth of dormant micrometastases by inducing neutrophilic infiltration and converting neutrophil function. 14,15-EET triggered neutrophil infiltration in metastatic lesions by activating STAT3 and JNK pathways to induce the expression of human IL-8 and murine CXCL15 in corresponding tumor cells. The continuous expression of hIL-8/mCXCL15 was maintained by the sustained and enhanced activation of JNK pathway. 14,15-EET up-regulated miR-155 expression by activating STAT3 and JNK pathways. miR-155 in turn down-regulated the expression of SHIP1 and DET1, thus augmenting the activation of JNK and c-Jun. Moreover, the function of neutrophils was converted from tumor-suppressing to tumor-promoting by 14,15-EET in vivo. By inducing the production of G-CSF/IL-6 in vivo, 14,15-EET induced the enhancement of STAT3 activation in neutrophils to increase MMP-9 expression and decrease TRAIL expression. Neutrophil-derived MMP-9 was required for 14,15-EET to induce angiogenesis during the growth of dormant micrometastases. Depleting neutrophils or inhibiting hIL-8/mCXCL15 up-regulation resulted in the failure of 14,15-EET to promote the development of micrometastases. These findings reveal a mechanism through which the infiltration and tumor-promoting function of neutrophils could be induced to trigger the growth of dormant metastases, which might be a driving force for the tumor recurrence based on dormant metastases.
Collapse
MESH Headings
- 8,11,14-Eicosatrienoic Acid/analogs & derivatives
- 8,11,14-Eicosatrienoic Acid/pharmacology
- Animals
- Chemokines, CXC/genetics
- Chemokines, CXC/metabolism
- Down-Regulation
- Granulocyte Colony-Stimulating Factor/metabolism
- Hep G2 Cells
- Humans
- Interleukin-6/metabolism
- Interleukin-8/genetics
- Interleukin-8/metabolism
- MCF-7 Cells
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- MicroRNAs/metabolism
- Neoplasm Invasiveness/pathology
- Neoplasm Micrometastasis/pathology
- Neoplasm Recurrence, Local/pathology
- Neovascularization, Pathologic/pathology
- Neutrophil Infiltration/drug effects
- Neutrophils/metabolism
- Neutrophils/pathology
- RNA Interference
- RNA, Small Interfering/metabolism
- STAT3 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Up-Regulation
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jing Luo
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - Xin-Xia Feng
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hongshan, Wuhan 430030, People's Republic of China
| | - Chao Luo
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - Yu Wang
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - Dong Li
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - Yu Shu
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - Shan-Shan Wang
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - Jian Qin
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - Yong-Chao Li
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - Jiu-Ming Zou
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - De-An Tian
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hongshan, Wuhan 430030, People's Republic of China
| | - Gui-Mei Zhang
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| | - Zuo-Hua Feng
- Department of Biochemistry and Molecular Biology, Wuhan University, School of Basic Medicine, Hubei, Wuhan 430030, People's Republic of China
| |
Collapse
|