1
|
Saadh MJ, Rasulova I, Khalil M, Farahim F, Sârbu I, Ciongradi CI, Omar TM, Alhili A, Jawad MJ, Hani T, Ali T, Lakshmaiya N. Natural killer cell-mediated immune surveillance in cancer: Role of tumor microenvironment. Pathol Res Pract 2024; 254:155120. [PMID: 38280274 DOI: 10.1016/j.prp.2024.155120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/29/2024]
Abstract
In the immunological surveillance against cancer, natural killer (NK) cells are essential effectors that help eradicate altered cells. The complex interactions that occur between NK cells and the tumor microenvironment (TME) are thoroughly examined in this review. The review examines how cytokine stimulation affects NK cell activation, focusing on the dynamic modulation of NK cell function within the TME. It looks at NK cell-related biomarkers such as PD-1/PD-L1, methylation HOXA9 (Homeobox A9), Stroma AReactive Invasion Front Areas (SARIFA), and NKG2A/HLA-E, providing critical information about prognosis and treatment outcomes. The changing landscape of immunotherapies-including checkpoint inhibitors, CAR-NK cells, and cytokine-based interventions-is examined in the context of enhancing NK cell activity. The review highlights the potential pathways for precision medicine going forward, focusing on customized immunotherapies based on unique biomarker profiles and investigating combination medicines to produce more robust anti-tumor responses.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan
| | - Irodakhon Rasulova
- MD, PhD, Senior Researcher, School of Humanities, Natural & Social Sciences, New Uzbekistan University, 54 Mustaqillik Ave., Tashkent, 100007, Uzbekistan; Department of Public Health, Samarkand State Medical University, Amir Temur street 18, Samarkand, Uzbekistan
| | | | - Farha Farahim
- Department of Nursing, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Ioan Sârbu
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| | - Carmen Iulia Ciongradi
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| | - Thabit Moath Omar
- Department of Medical Laboratory Technics, Al-Noor University College, Nineveh, Iraq
| | - Ahmed Alhili
- Medical Technical College, Al-Farahidi University, Iraq
| | | | - Thamer Hani
- Dentistry Department, Al-Turath University College, Baghdad, Iraq
| | - Talat Ali
- Department of Basic Medical Sciences, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SIMATS, Chennai, Tamil Nadu, India
| |
Collapse
|
2
|
Lujan DA, Ochoa JL, Beswick EJ, Howard TA, Hathaway HJ, Perrone-Bizzozero NI, Hartley RS. Cold-Inducible RNA Binding Protein Impedes Breast Tumor Growth in the PyMT Murine Model for Breast Cancer. Biomedicines 2024; 12:340. [PMID: 38397942 PMCID: PMC10886683 DOI: 10.3390/biomedicines12020340] [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: 12/15/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
RNA binding proteins (RBPs) post-transcriptionally regulate gene expression by associating with regulatory sequences in the untranslated regions of mRNAs. Cold-inducible RBP (CIRP) is a stress-induced RBP that was recently shown to modulate inflammation in response to cellular stress, where it increases or decreases pro-tumorigenic (proinflammatory) cytokines in different contexts. CIRP expression is altered in several cancers, including breast cancer, but the effects of CIRP on inflammation in breast cancer is not known. Here, we investigate if CIRP alters growth and the inflammatory profile of breast tumors. Transgenic mice overexpressing CIRP in the mammary epithelium were crossed with the PyMT mouse model of breast cancer, and the effects on both early and late tumorigenesis and inflammation were assessed. The effects of CIRP knockdown were also assessed in Py2T cell grafts. Overexpression of CIRP led to decreased tumorigenesis in the PyMT mouse model. Conversely, the knockdown of CIRP in Py2T cell grafts led to increased tumor growth. Luminex cytokine assays assessed the effects on the inflammatory environment. CIRP/PyMT mammary glands/mammary tumors and serum had decreased cytokines that promote inflammation, angiogenesis, and metastasis compared to PyMT mammary glands and serum, documenting a shift towards an environment less supportive of tumorigenesis. CIRP overexpression also decreased CD4+ helper T cells and increased CD8+ cytotoxic T cells in mammary tumors. Overall, these data support a role for CIRP as a potent antitumor molecule that suppresses both local and systemic pro-tumorigenic inflammation.
Collapse
Affiliation(s)
- Daniel A. Lujan
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| | - Joey L. Ochoa
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| | - Ellen J. Beswick
- Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, KY 40506, USA;
| | - Tamara A. Howard
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| | - Helen J. Hathaway
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| | - Nora I. Perrone-Bizzozero
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA;
| | - Rebecca S. Hartley
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; (D.A.L.); (J.L.O.); (T.A.H.); (H.J.H.)
| |
Collapse
|
3
|
Cheng X, Wang H, Wang Z, Zhu B, Long H. Tumor-associated myeloid cells in cancer immunotherapy. J Hematol Oncol 2023; 16:71. [PMID: 37415162 PMCID: PMC10324139 DOI: 10.1186/s13045-023-01473-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
Tumor-associated myeloid cells (TAMCs) are among the most important immune cell populations in the tumor microenvironment, and play a significant role on the efficacy of immune checkpoint blockade. Understanding the origin of TAMCs was found to be the essential to determining their functional heterogeneity and, developing cancer immunotherapy strategies. While myeloid-biased differentiation in the bone marrow has been traditionally considered as the primary source of TAMCs, the abnormal differentiation of splenic hematopoietic stem and progenitor cells, erythroid progenitor cells, and B precursor cells in the spleen, as well as embryo-derived TAMCs, have been depicted as important origins of TAMCs. This review article provides an overview of the literature with a focus on the recent research progress evaluating the heterogeneity of TAMCs origins. Moreover, this review summarizes the major therapeutic strategies targeting TAMCs with heterogeneous sources, shedding light on their implications for cancer antitumor immunotherapies.
Collapse
Affiliation(s)
- Xinyu Cheng
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China
| | - Huilan Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China
| | - Zhongyu Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China.
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China.
| |
Collapse
|
4
|
Ray AL, Saunders AS, Nofchissey RA, Reidy MA, Kamal M, Lerner MR, Fung KM, Lang ML, Hanson JA, Guo S, Urdaneta-Perez MG, Lewis SE, Cloyde M, Morris KT. G-CSF Is a Novel Mediator of T-Cell Suppression and an Immunotherapeutic Target for Women with Colon Cancer. Clin Cancer Res 2023; 29:2158-2169. [PMID: 36951682 PMCID: PMC10239359 DOI: 10.1158/1078-0432.ccr-22-3918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/16/2023] [Accepted: 03/21/2023] [Indexed: 03/24/2023]
Abstract
PURPOSE G-CSF enhances colon cancer development. This study defines the prevalence and effects of increased G-CSF signaling in human colon cancers and investigates G-CSF inhibition as an immunotherapeutic strategy against metastatic colon cancer. EXPERIMENTAL DESIGN Patient samples were used to evaluate G-CSF and G-CSF receptor (G-CSFR) levels by IHC with sera used to measure G-CSF levels. Peripheral blood mononuclear cells were used to assess the rate of G-CSFR+ T cells and IFNγ responses to chronic ex vivo G-CSF. An immunocompetent mouse model of peritoneal metastasis (MC38 cells in C57Bl/6J) was used to determine the effects of G-CSF inhibition (αG-CSF) on survival and the tumor microenvironment (TME) with flow and mass cytometry. RESULTS In human colon cancer samples, the levels of G-CSF and G-CSFR are higher compared to normal colon tissues from the same patient. High patient serum G-CSF is associated with increases in markers of poor prognosis, (e.g., VEGF, IL6). Circulating T cells from patients express G-CSFR at double the rate of T cells from controls. Prolonged G-CSF exposure decreases T cell IFNγ production. Treatment with αG-CSF shifts both the adaptive and innate compartments of the TME and increases survival (HR, 0.46; P = 0.0237) and tumor T-cell infiltration, activity, and IFNγ response with greater effects in female mice. There is a negative correlation between serum G-CSF levels and tumor-infiltrating T cells in patient samples from women. CONCLUSIONS These findings support G-CSF as an immunotherapeutic target against colon cancer with greater potential benefit in women.
Collapse
Affiliation(s)
- Anita L Ray
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Apryl S Saunders
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Robert A Nofchissey
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Megan A Reidy
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Maria Kamal
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Megan R Lerner
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Mark L Lang
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Joshua A Hanson
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Shaoxuan Guo
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Maria G Urdaneta-Perez
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Samara E Lewis
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Michael Cloyde
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Katherine T Morris
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
5
|
Jiang W, Ouyang X, Jiang C, Yin L, Yao Q, Pei X, Ji Z, Li M, Song S, Yang W, Huang S, Yang H, Shan B. A NOTCH1 Mutation Found in a Newly Established Ovarian Cancer Cell Line (FDOVL) Promotes Lymph Node Metastasis in Ovarian Cancer. Int J Mol Sci 2023; 24:ijms24065091. [PMID: 36982170 PMCID: PMC10049685 DOI: 10.3390/ijms24065091] [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: 01/16/2023] [Revised: 02/11/2023] [Accepted: 02/23/2023] [Indexed: 03/30/2023] Open
Abstract
Peritoneal implantation and lymph node metastasis have different driving mechanisms in ovarian cancer. Elucidating the underlying mechanism of lymph node metastasis is important for treatment outcomes. A new cell line, FDOVL, was established from a metastatic lymph node of a patient with primary platinum-resistant ovarian cancer and was then characterized. The effect of NOTCH1-p.C702fs mutation and NOTCH1 inhibitor on migration was evaluated in vitro and in vivo. Ten paired primary sites and metastatic lymph nodes were analyzed by RNA sequencing. The FDOVL cell line with serious karyotype abnormalities could be stably passaged and could be used to generated xenografts. NOTCH1-p.C702fs mutation was found exclusively in the FDOVL cell line and the metastatic lymph node. The mutation promoted migration and invasion in cell and animal models, and these effects were markedly repressed by the NOTCH inhibitor LY3039478. RNA sequencing confirmed CSF3 as the downstream effector of NOTCH1 mutation. Furthermore, the mutation was significantly more common in metastatic lymph nodes than in other peritoneal metastases in 10 paired samples (60% vs. 20%). The study revealed that NOTCH1 mutation is probably a driver of lymph node metastasis in ovarian cancer, which offers new ideas for the treatment of ovarian cancer lymph node metastasis with NOTCH inhibitors.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xueyan Ouyang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chunjuan Jiang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Lina Yin
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qianlan Yao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Xuan Pei
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhaodong Ji
- Department of Clinical Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ming Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Shaoli Song
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Wentao Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Shenglin Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Huijuan Yang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Boer Shan
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
6
|
Matos I, Barvalia M, Chehal MK, Robertson AG, Kulic I, Silva JAFD, Ranganathan A, Short A, Huang YH, Long E, Priatel JJ, Dhanji S, Nelson BH, Krebs DL, Harder KW. Tumor-derived GCSF Alters Tumor and Systemic Immune System Cell Subset Composition and Signaling. CANCER RESEARCH COMMUNICATIONS 2023; 3:404-419. [PMID: 36911097 PMCID: PMC9997410 DOI: 10.1158/2767-9764.crc-22-0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/01/2022] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
While immunotherapies such as immune checkpoint blockade and adoptive T-cell therapy improve survival for a subset of human malignancies, many patients fail to respond. Phagocytes including dendritic cells (DC), monocytes, and macrophages (MF) orchestrate innate and adaptive immune responses against tumors. However, tumor-derived factors may limit immunotherapy effectiveness by altering phagocyte signal transduction, development, and activity. Using Cytometry by Time-of-Flight, we found that tumor-derived GCSF altered myeloid cell distribution both locally and systemically. We distinguished a large number of GCSF-induced immune cell subset and signal transduction pathway perturbations in tumor-bearing mice, including a prominent increase in immature neutrophil/myeloid-derived suppressor cell (Neut/MDSC) subsets and tumor-resident PD-L1+ Neut/MDSCs. GCSF expression was also linked to distinct tumor-associated MF populations, decreased conventional DCs, and splenomegaly characterized by increased splenic progenitors with diminished DC differentiation potential. GCSF-dependent dysregulation of DC development was recapitulated in bone marrow cultures in vitro, using medium derived from GCSF-expressing tumor cell cultures. Importantly, tumor-derived GCSF impaired T-cell adoptive cell therapy effectiveness and was associated with increased tumor volume and diminished survival of mice with mammary cancer. Treatment with neutralizing anti-GCSF antibodies reduced colonic and circulatory Neut/MDSCs, normalized colonic immune cell composition and diminished tumor burden in a spontaneous model of mouse colon cancer. Analysis of human colorectal cancer patient gene expression data revealed a significant correlation between survival and low GCSF and Neut/MDSC gene expression. Our data suggest that normalizing GCSF bioactivity may improve immunotherapy in cancers associated with GCSF overexpression. Significance Tumor-derived GCSF leads to systemic immune population changes. GCSF blockade restores immune populations, improves immunotherapy, and reduces tumor size, paralleling human colorectal cancer data. GCSF inhibition may synergize with current immunotherapies to treat GCSF-secreting tumors.
Collapse
Affiliation(s)
- Israel Matos
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Maunish Barvalia
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Manreet K Chehal
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Agency. Vancouver, British Columbia, Canada
| | - Iva Kulic
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - Jessica A F D Silva
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Abhinandan Ranganathan
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Amy Short
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - Yu-Hsuan Huang
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - Erin Long
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - John J Priatel
- ME Therapeutics Inc. Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Salim Dhanji
- ME Therapeutics Inc. Vancouver, British Columbia, Canada
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, British Columbia, Canada
| | - Danielle L Krebs
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada
| | - Kenneth W Harder
- Department of Microbiology and Immunology, University of British Columbia, Life Sciences Institute, Vancouver, British Columbia, Canada.,ME Therapeutics Inc. Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Sekino Y, Teishima J, Liang G, Hinata N. Molecular mechanisms of resistance to tyrosine kinase inhibitor in clear cell renal cell carcinoma. Int J Urol 2022; 29:1419-1428. [PMID: 36122306 PMCID: PMC10087189 DOI: 10.1111/iju.15042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/25/2022] [Indexed: 12/24/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma (RCC). Loss of von Hippel-Lindau tumor suppressor gene is frequently observed in ccRCC and increases the expression of hypoxia-inducible factors and their targets, including epidermal growth factor, vascular endothelial growth factor, and platelet-derived growth factor. Tyrosine kinase inhibitors (TKIs) offer a survival benefit in metastatic renal cell carcinoma (mRCC). Recently, immune checkpoint inhibitors have been introduced in mRCC. Combination therapy with TKIs and immune checkpoint inhibitors significantly improved patient outcomes. Therefore, TKIs still play an essential role in mRCC treatment. However, the clinical utility of TKIs is compromised when primary and acquired resistance are encountered. The mechanism of resistance to TKI is not fully elucidated. Here, we comprehensively reviewed the molecular mechanisms of resistance to TKIs and a potential strategy to overcome this resistance. We outlined the involvement of angiogenesis, non-angiogenesis, epithelial-mesenchymal transition, activating bypass pathways, lysosomal sequestration, non-coding RNAs, epigenetic modifications and tumor microenvironment factors in the resistance to TKIs. Deep insight into the molecular mechanisms of resistance to TKIs will help to better understand the biology of RCC and can ultimately help in the development of more effective therapies.
Collapse
Affiliation(s)
- Yohei Sekino
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Department of Urology, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Jun Teishima
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Gangning Liang
- Department of Urology, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| | - Nobuyuki Hinata
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
8
|
Park SD, Saunders AS, Reidy MA, Bender DE, Clifton S, Morris KT. A review of granulocyte colony-stimulating factor receptor signaling and regulation with implications for cancer. Front Oncol 2022; 12:932608. [PMID: 36033452 PMCID: PMC9402976 DOI: 10.3389/fonc.2022.932608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/15/2022] [Indexed: 12/29/2022] Open
Abstract
Granulocyte colony-stimulating factor receptor (GCSFR) is a critical regulator of granulopoiesis. Studies have shown significant upregulation of GCSFR in a variety of cancers and cell types and have recognized GCSFR as a cytokine receptor capable of influencing both myeloid and non-myeloid immune cells, supporting pro-tumoral actions. This systematic review aims to summarize the available literature examining the mechanisms that control GCSFR signaling, regulation, and surface expression with emphasis on how these mechanisms may be dysregulated in cancer. Experiments with different cancer cell lines from breast cancer, bladder cancer, glioma, and neuroblastoma are used to review the biological function and underlying mechanisms of increased GCSFR expression with emphasis on actions related to tumor proliferation, migration, and metastasis, primarily acting through the JAK/STAT pathway. Evidence is also presented that demonstrates a differential physiological response to aberrant GCSFR signal transduction in different organs. The lifecycle of the receptor is also reviewed to support future work defining how this signaling axis becomes dysregulated in malignancies.
Collapse
Affiliation(s)
- Sungjin David Park
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Apryl S. Saunders
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Megan A. Reidy
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Dawn E. Bender
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Shari Clifton
- Department of Information Management, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Katherine T. Morris
- Department of Surgery, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
- *Correspondence: Katherine T. Morris,
| |
Collapse
|
9
|
Lee JE, Lee E. The Probiotic Effects of the Saccharomyces cerevisiae 28-7 Strain Isolated from Nuruk in a DSS-Induced Colitis Mouse Model. J Microbiol Biotechnol 2022; 32:877-884. [PMID: 35791078 PMCID: PMC9628919 DOI: 10.4014/jmb.2206.06035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 12/15/2022]
Abstract
Probiotics are microorganisms that can benefit host health when ingested in a live state, and lactic acid bacteria are the most common type. Among fungi, Saccharomyces boulardii (SB) is the only strain known to have a probiotic function with beneficial effects on colitis; however, information on other probiotic yeast strains is limited. Therefore, this study aimed to discover yeast strains expressing intestinal anti-inflammatory activities by exhibiting probiotic properties in dextran sodium sulfate (DSS)-induced colitis mice model. Nuruk (Korean traditional fermentation starter) containing various microbial strains was used as a source for yeast strains, and S. cerevisiae 28-7 (SC28-7) strain was selected with in vitro and in vivo characteristics to enable survival in the intestines. After 14 days of pretreatment with the yeast strains, DSS was co-administered for six days to induce colitis in mice. The results revealed that the disease activity index score was lowered by SC28-7 treatment compared to the DSS group, and the colon length and weight/length ratio were recovered in a pattern similar to that of the normal group. SC28-7 administration significantly reduced the secretion of pro-inflammatory cytokines in the serum and modified the mRNA expression of inflammatory cytokines (interleukin-1β, transforming growth factor-β, and interferon-γ) and proteins involved in gut barrier functions (mucin 2, mucin 3, zonula occludens-1, and occludin) in colon tissues. These results indicate that SC28-7 attenuates DSS-induced colon damage and inflammation, supporting its future use as a probiotic yeast for treating and preventing intestinal inflammatory diseases such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Jang Eun Lee
- Reserch Group of Traditional Food, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea,Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Eunjung Lee
- Reserch Group of Traditional Food, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea,Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea,Corresponding author Phone: +82-63-219-9413 E-mail:
| |
Collapse
|
10
|
Shoari A, Tahmasebi M, Khodabakhsh F, Cohan RA, Oghalaie A, Behdani M. Angiogenic biomolecules specific nanobodies application in cancer imaging and therapy; review and updates. Int Immunopharmacol 2022; 105:108585. [DOI: 10.1016/j.intimp.2022.108585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/05/2022]
|
11
|
Malengier-Devlies B, Metzemaekers M, Wouters C, Proost P, Matthys P. Neutrophil Homeostasis and Emergency Granulopoiesis: The Example of Systemic Juvenile Idiopathic Arthritis. Front Immunol 2021; 12:766620. [PMID: 34966386 PMCID: PMC8710701 DOI: 10.3389/fimmu.2021.766620] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are key pathogen exterminators of the innate immune system endowed with oxidative and non-oxidative defense mechanisms. More recently, a more complex role for neutrophils as decision shaping cells that instruct other leukocytes to fine-tune innate and adaptive immune responses has come into view. Under homeostatic conditions, neutrophils are short-lived cells that are continuously released from the bone marrow. Their development starts with undifferentiated hematopoietic stem cells that pass through different immature subtypes to eventually become fully equipped, mature neutrophils capable of launching fast and robust immune responses. During severe (systemic) inflammation, there is an increased need for neutrophils. The hematopoietic system rapidly adapts to this increased demand by switching from steady-state blood cell production to emergency granulopoiesis. During emergency granulopoiesis, the de novo production of neutrophils by the bone marrow and at extramedullary sites is augmented, while additional mature neutrophils are rapidly released from the marginated pools. Although neutrophils are indispensable for host protection against microorganisms, excessive activation causes tissue damage in neutrophil-rich diseases. Therefore, tight regulation of neutrophil homeostasis is imperative. In this review, we discuss the kinetics of neutrophil ontogenesis in homeostatic conditions and during emergency myelopoiesis and provide an overview of the different molecular players involved in this regulation. We substantiate this review with the example of an autoinflammatory disease, i.e. systemic juvenile idiopathic arthritis.
Collapse
Affiliation(s)
- Bert Malengier-Devlies
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mieke Metzemaekers
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Carine Wouters
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.,Division of Pediatric Rheumatology, University Hospitals Leuven, Leuven, Belgium.,European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at University Hospital Leuven, Leuven, Belgium
| | - Paul Proost
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Mouchemore KA, Anderson RL. Immunomodulatory effects of G-CSF in cancer: Therapeutic implications. Semin Immunol 2021; 54:101512. [PMID: 34763974 DOI: 10.1016/j.smim.2021.101512] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/23/2021] [Indexed: 01/04/2023]
Abstract
Numerous preclinical studies have reported a pro-tumour role for granulocyte colony-stimulating factor (G-CSF) that is predominantly mediated by neutrophils and MDSCs, the major G-CSF receptor expressing populations. In the presence of G-CSF (either tumour-derived or exogenous) these myeloid populations commonly exhibit a T cell suppressive phenotype. However, the direct effects of this cytokine on other immune lineages, such as T and NK cells, are not as well established. Herein we discuss the most recent data relating to the effect of G-CSF on the major immune populations, exclusively in the context of cancer. Recent publications have drawn attention to the other tumour-promoting effects of G-CSF on myeloid cells, including NETosis, promotion of cancer stemness and skewed differentiation of bone marrow progenitors towards myelopoiesis. Although G-CSF is safely and commonly used as a supportive therapy to prevent or treat chemotherapy-associated neutropenia in cancer patients, we also discuss the potential impacts of G-CSF on other anti-cancer treatments. Importantly, considerations for immune checkpoint blockade are highlighted, as many publications report a T cell suppressive effect of G-CSF that may diminish the effectiveness of this immunotherapy.
Collapse
Affiliation(s)
- Kellie A Mouchemore
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Robin L Anderson
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
13
|
Zhang W, Karagiannidis I, Van Vliet EDS, Yao R, Beswick EJ, Zhou A. Granulocyte colony-stimulating factor promotes an aggressive phenotype of colon and breast cancer cells with biochemical changes investigated by single-cell Raman microspectroscopy and machine learning analysis. Analyst 2021; 146:6124-6131. [PMID: 34543367 PMCID: PMC8631005 DOI: 10.1039/d1an00938a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) is produced at high levels in several cancers and is directly linked with metastasis in gastrointestinal (GI) cancers. In order to further understand the alteration of molecular compositions and biochemical features triggered by G-CSF treatment at molecular and cell levels, we sought to investigate the long term treatment of G-CSF on colon and breast cancer cells measured by label-free, non-invasive single-cell Raman microspectroscopy. Raman spectrum captures the molecule-specific spectral signatures ("fingerprints") of different biomolecules presented on cells. In this work, mouse breast cancer line 4T1 and mouse colon cancer line CT26 were treated with G-CSF for 7 weeks and subsequently analyzed by machine learning based Raman spectroscopy and gene/cytokine expression. The principal component analysis (PCA) identified the Raman bands that most significantly changed between the control and G-CSF treated cells. Notably, here we proposed the concept of aggressiveness score, which can be derived from the posterior probability of linear discriminant analysis (LDA), for quantitative spectral analysis of tumorigenic cells. The aggressiveness score was effectively applied to analyze and differentiate the overall cell biochemical changes of G-CSF-treated two model cancer cells. All these tumorigenic progressions suggested by Raman analysis were confirmed by pro-tumorigenic cytokine and gene analysis. A high correlation between gene expression data and Raman spectra highlights that the machine learning based non-invasive Raman spectroscopy offers emerging and powerful tools to better understand the regulation mechanism of cytokines in the tumor microenvironment that could lead to the discovery of new targets for cancer therapy.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Biological Engineering, Utah State University, Logan, UT 84322, USA.
| | - Ioannis Karagiannidis
- Department of Internal Medicine, Division of Gastroenterology, University of Utah School of Medicine, Salt Lake City, UT84132, USA.
| | - Eliane De Santana Van Vliet
- Department of Internal Medicine, Division of Gastroenterology, University of Utah School of Medicine, Salt Lake City, UT84132, USA.
| | - Ruoxin Yao
- Department of Internal Medicine, Division of Gastroenterology, University of Utah School of Medicine, Salt Lake City, UT84132, USA.
| | - Ellen J Beswick
- Department of Internal Medicine, Division of Gastroenterology, University of Utah School of Medicine, Salt Lake City, UT84132, USA.
| | - Anhong Zhou
- Department of Biological Engineering, Utah State University, Logan, UT 84322, USA.
| |
Collapse
|
14
|
Karagiannidis I, Salataj E, Said Abu Egal E, Beswick EJ. G-CSF in tumors: Aggressiveness, tumor microenvironment and immune cell regulation. Cytokine 2021; 142:155479. [PMID: 33677228 DOI: 10.1016/j.cyto.2021.155479] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) is a cytokine most well-known for maturation and mobilization of bone marrow neutrophils. Although it is used therapeutically to treat chemotherapy induced neutropenia, it is also highly expressed in some tumors. Case reports suggest that tumors expressing high levels of G-CSF are aggressive, more difficult to treat, and present with poor prognosis and high mortality rates. Research on this topic suggests that G-CSF has tumor-promoting effects on both tumor cells and the tumor microenvironment. G-CSF has a direct effect on tumor cells to promote tumor stem cell longevity and overall tumor cell proliferation and migration. Additionally, it may promote pro-tumorigenic immune cell phenotypes such as M2 macrophages, myeloid-derived suppressor cells, and regulatory T cells. Overall, the literature suggests a plethora of pro-tumorigenic activity that should be balanced with the therapeutic use. In this review, we present an overview of the multiple complex roles of G-CSF and G-CSFR in tumors and their microenvironment and discuss how clinical advances and strategies may open new therapeutic avenues.
Collapse
Affiliation(s)
- Ioannis Karagiannidis
- Division of Gastroenterology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, United States
| | - Eralda Salataj
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Crete, Greece
| | - Erika Said Abu Egal
- Division of Gastroenterology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, United States
| | - Ellen J Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, United States.
| |
Collapse
|
15
|
Colony-stimulating factor 3 signaling in colon and rectal cancers: Immune response and CMS classification in TCGA data. PLoS One 2021; 16:e0247233. [PMID: 33606788 PMCID: PMC7895368 DOI: 10.1371/journal.pone.0247233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/03/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer is the 2nd leading cause of cancer-related deaths in the world. The mechanisms underlying CRC development, progression, and resistance to treatment are complex and not fully understood. The immune response in the tumor microenvironment has been shown to play a significant role in many cancers, including colorectal cancer. Colony-stimulating factor 3 (CSF3) has been associated with changes to the immune environment in colorectal cancer animal models. We hypothesized that CSF3 signaling would correlate with pro-tumor tumor microenvironment changes associated with immune infiltrate and response. We utilized publicly available datasets to guide future mechanistic studies of the role CSF3 and its receptor (CSF3R) play in colorectal cancer development and progression. Here, we use bioinformatics data and mRNA from patients with colon (n = 242) or rectal (n = 92) cancers, obtained from The Cancer Genome Atlas Firehose Legacy dataset. We examined correlations of CSF3 and CSF3R expression with patient demographics, tumor stage and consensus molecular subtype classification. Gene expression correlations, cell type enrichment, Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data scores and Gene Ontology were used to analyze expression of receptor and ligand, tumor microenvironment infiltration of immune cells, and alterations in biological pathways. We found that CSF3 and CSF3R expression is highest in consensus molecular subtype 1 and consensus molecular subtype 4. Ligand and receptor expression are also correlated with changes in T cell and macrophage signatures. CSF3R significantly correlates with a large number of genes that are associated with poor colorectal cancer prognosis.
Collapse
|
16
|
Karagiannidis I, de Santana Van Vilet E, Said Abu Egal E, Phinney B, Jacenik D, Prossnitz ER, Beswick EJ. G-CSF and G-CSFR Induce a Pro-Tumorigenic Macrophage Phenotype to Promote Colon and Pancreas Tumor Growth. Cancers (Basel) 2020; 12:cancers12102868. [PMID: 33036138 PMCID: PMC7601499 DOI: 10.3390/cancers12102868] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAMs) in the gastrointestinal tumor microenvironment (TME) are known to polarize into populations exhibiting pro- or anti-tumoral activity in response to stimuli such as growth factors and cytokines. Our previous work has recognized granulocyte colony-stimulating factor (G-CSF) as a cytokine capable of influencing immune cells of the TME exhibiting pro-tumoral activity. Here, we aimed to focus on how G-CSF regulates TAM phenotype and function and the effects on gastrointestinal (GI) tumor progression. Thus, wildtype (WT) and G-CSFR-/- macrophages were examined for cytokine production, gene expression, and transcription factor activity. Adoptive transfer of WT or G-CSFR-/- macrophages into tumor-bearing mice was performed to study their influence in the progression of colon (MC38) and pancreatic (PK5L1940) tumor mouse models. Finally, the difference in cytotoxic potential between WT and G-CSFR-/- macrophages was examined both in vitro and in vivo. Our results indicate that G-CSF promotes increased IL-10 production and decreased IL-12 production, which was reversed in G-CSFR-/- macrophages for a pro-inflammatory phenotype. Furthermore, G-CSFR-/- macrophages were characterized by higher levels of NOS2 expression and NO production, which led to greater tumor related cytotoxicity both in vitro and in vivo. Our results suggest that in the absence of G-CSFR, macrophage-related tumor cytotoxicity was amplified. These findings, along with our previous reports, pinpoint G-CSF /G-CSFR as a prominent target for possible clinical applications that aim to control the TME and the GI tumor progression.
Collapse
Affiliation(s)
- Ioannis Karagiannidis
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA; (I.K.); (E.d.S.V.V.); (E.S.A.E.)
| | - Eliane de Santana Van Vilet
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA; (I.K.); (E.d.S.V.V.); (E.S.A.E.)
| | - Erika Said Abu Egal
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA; (I.K.); (E.d.S.V.V.); (E.S.A.E.)
| | - Brandon Phinney
- Division of Molecular Medicine, Department of Internal Medicine; Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Comprehensive Cancer Center; University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (B.P.); (D.J.); (E.R.P.)
| | - Damian Jacenik
- Division of Molecular Medicine, Department of Internal Medicine; Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Comprehensive Cancer Center; University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (B.P.); (D.J.); (E.R.P.)
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
| | - Eric R. Prossnitz
- Division of Molecular Medicine, Department of Internal Medicine; Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Comprehensive Cancer Center; University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (B.P.); (D.J.); (E.R.P.)
| | - Ellen J. Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA; (I.K.); (E.d.S.V.V.); (E.S.A.E.)
- Correspondence:
| |
Collapse
|
17
|
Karagiannidis I, Jerman SJ, Jacenik D, Phinney BB, Yao R, Prossnitz ER, Beswick EJ. G-CSF and G-CSFR Modulate CD4 and CD8 T Cell Responses to Promote Colon Tumor Growth and Are Potential Therapeutic Targets. Front Immunol 2020; 11:1885. [PMID: 33042110 PMCID: PMC7522314 DOI: 10.3389/fimmu.2020.01885] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/13/2020] [Indexed: 01/01/2023] Open
Abstract
Cytokines are known to shape the tumor microenvironment and although progress has been made in understanding their role in carcinogenesis, much remains to learn regarding their role in tumor growth and progression. We have identified granulocyte colony-stimulating factor (G-CSF) as one such cytokine, showing that G-CSF is linked with metastasis in human gastrointestinal tumors and neutralizing G-CSF in a mouse model of colitis-associated cancer is protective. Here, we set out to identify the role of G-CSF and its receptor, G-CSFR, in CD4+ and CD8+ T cell responses in the tumor microenvironment. MC38 colon cancer cells were injected into WT, G-CSFR-/- mice, or Rag2-/- mice. Flow cytometry, Real Time PCR and Multiplex cytokine array analysis were used for in vitro T cell phenotype analysis. Adoptive transfer of WT or G-CSFR-/- CD4+ of CD8+ T cells were performed. Mouse tumor size, cytokine expression, T cell phenotype, and cytotoxic activity were analyzed. We established that in G-CSFR-/- mice, tumor growth of MC38 colon cancer cells is significantly decreased. T cell phenotype and cytokine production were also altered, as both in vitro and in vivo approaches revealed that the G-CSF/G-CSFR stimulate IL-10-producing, FoxP3-expressing CD4+ and CD8+ T cells, whereas G-CSFR-/- T cells exhibit increased IFNγ and IL-17A production, leading to increased cytotoxic activity in the tumor microenvironment. Furthermore, peritumoral injection of recombinant IFNγ or IL-17A inhibited colon and pancreas tumor growth compared to controls. Taken together, our data reveal an unknown mechanism by which G-CSF, through its receptor G-CSFR, promotes an inhibitory Treg phenotype that limits tumor immune responses and furthermore suggest that targeting this cytokine/receptor axis could represent a novel therapeutic approach for gastrointestinal, and likely other tumors with high expression of these factors.
Collapse
Affiliation(s)
- Ioannis Karagiannidis
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Stephanie J. Jerman
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center Albuquerque, Albuquerque, NM, United States
| | - Damian Jacenik
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States,Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Brandon B. Phinney
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center Albuquerque, Albuquerque, NM, United States
| | - Ruoxin Yao
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Eric R. Prossnitz
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Ellen J. Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States,*Correspondence: Ellen J. Beswick
| |
Collapse
|
18
|
Zhao Y, Rahmy S, Liu Z, Zhang C, Lu X. Rational targeting of immunosuppressive neutrophils in cancer. Pharmacol Ther 2020; 212:107556. [PMID: 32343986 DOI: 10.1016/j.pharmthera.2020.107556] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
Neutrophils, the most abundant circulating leukocytes in human, play an indispensable role in the innate immune response to microbial infections. However, the contribution of tumor-associated neutrophils (TANs) to cancer progression and tumor immunity has been a matter of debate for decades. A higher neutrophil-to-lymphocyte ratio is associated with adverse overall survival in many solid tumors. Preclinical evidence exists to support both anti-tumor and pro-tumor activities of TANs, and TANs employ diverse mechanisms to influence tumor progression and metastasis. Here, we focus our review on the immunosuppressive mechanism of TANs and highlight how neutrophils can operate to dampen both innate and adaptive immunity to promote tumorigenesis. Here we discuss the intriguing and sometimes controversial connection between TANs and granulocytic/polymorphonuclear myeloid-derived suppressor cells (G/PMN-MDSCs). The molecular mechanisms underlying neutrophils' role in immunosuppression provide potential therapeutic targets for cancer treatment, either as monotherapies or as a part of combinatorial regimens. Therefore, we also highlight a number of neutrophil-targeting approaches that may improve the efficacy of current anticancer therapies, especially cancer immunotherapy. Currently interest is surging in the understanding and targeting of immunosuppressive neutrophils, with the goal of developing novel therapeutic strategies in the battle against cancer.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University, Shanghai 200092, China; Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sharif Rahmy
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Zhongmin Liu
- Department of Cardiac Surgery, Shanghai East Hospital, Tongji University, Shanghai 200092, China
| | - Chao Zhang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Xin Lu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA; Integrated Biomedical Sciences Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA; Tumor Microenvironment and Metastasis Program, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
19
|
Qeadan F, Bansal P, Hanson JA, Beswick EJ. The MK2 pathway is linked to G-CSF, cytokine production and metastasis in gastric cancer: a novel intercorrelation analysis approach. J Transl Med 2020; 18:137. [PMID: 32216812 PMCID: PMC7098132 DOI: 10.1186/s12967-020-02294-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background Gastric cancer is associated with chronic inflammation, but there is still much to understand about the tumor microenvironment and the underlying tumor-promoting mechanisms. The Map kinase-activated protein kinase 2 (MK2) pathway is a regulator of inflammatory cytokine production that we have been studying in gastrointestinal cancers. Here, we set out to determine the significance of this gene in gastric cancer along with its downstream mediators and if there were differences in the primary tumors with and without metastasis. Methods Human gastric cancer tissues with and without metastasis were examined for MK2 expression and cytokine profile in organ culture supernatants. Advanced statistical methods including a lower triangular correlation matrix, novel rooted correlation network, linear and logistic regression modeling along with Kruskal–Wallis testing with Sidak correction for multiple testing were applied to gain understanding of cytokines/chemokines linked to metastasis. Results The MK2 pathway is strongly linked with metastasis and a panel of cytokines. Gene expression was able to classify gastric cancer metastasis 85.7% of the time. A significant association with a panel of cytokines was found, including G-CSF, GM-CSF, Mip-1β, IFN-α, MCP-1, IL-1β, IL-6, and TNF-α. Mip-1β was found to have the strongest association with MK2 and metastasis after Sidak correction for multiple testing. Conclusions MK2 gene expression and a novel associated cytokine panel are linked to gastric cancer metastasis. G-CSF is the strongest cytokine to differentiate between metastasis and non-metastasis patients and had the lowest P value, while Mip-1β showed the strongest association with MK2 and metastasis after Sidak correction. MK2 and associated cytokines are potential biomarkers for gastric cancer metastasis. The novel intercorrelation analysis approach is a promising method for understanding the complex nature of cytokine/chemokine regulation and links to disease outcome.
Collapse
Affiliation(s)
- Fares Qeadan
- Department of Family and Preventative Medicine, University of Utah, Salt Lake City, UT, USA
| | - Pranshu Bansal
- New Mexico Oncology Hematology Consultants, Albuquerque, NM, USA
| | - Joshua A Hanson
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Ellen J Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
20
|
Critical Roles of Balanced Innate Lymphoid Cell Subsets in Intestinal Homeostasis, Chronic Inflammation, and Cancer. J Immunol Res 2019; 2019:1325181. [PMID: 31781671 PMCID: PMC6875018 DOI: 10.1155/2019/1325181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022] Open
Abstract
Innate lymphoid cells (ILCs) comprise a recently identified subset of innate immune cells that are mainly localized to mucosa-associated tissues. Although they have not yet been fully characterized, they can generally be divided into ILC1s, ILC2s, and ILC3s. ILCs and their corresponding cytokines act as important mediators of the early stages of the immune response during inflammation, tissue repair, and the maintenance of epithelial integrity. Consequently, the dysregulation of ILC subsets might promote inflammation and cancer. Numerous studies have demonstrated that these cells play an important role in maintaining the microecological balance of the small intestine; however, their specific roles in mediating inflammation in this tissue and tumorigenesis remain unclear and controversial. In this review, we focus on recent progress that has helped to gain a better understanding of the role of ILCs in intestinal homeostasis, chronic inflammation, and cancer. Further focused research on the regulation and role of ILCs in intestinal homeostasis and pathology will help to reveal valuable diagnostic and therapeutic targets for the treatment of intestinal diseases.
Collapse
|
21
|
Du R, Hu P, Liu Q, Zhang J, Deng G, Hu D, Zhang J. Granulocyte Colony-Stimulating Factor Treatment During Radiotherapy Is Associated With Survival Benefit in Patients With Lung Cancer. Technol Cancer Res Treat 2019; 17:1533033818816076. [PMID: 31122176 PMCID: PMC6295692 DOI: 10.1177/1533033818816076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Objectives: Granulocyte colony-stimulating factor, an agent commonly used for neutropenia treatment, plays an important role in cancer treatment. However, the effect of granulocyte colony-stimulating factor treatment on patient’s survival during radiation therapy in lung cancer remains unknown. Materials and Methods: A retrospective study of patients with lung cancer who underwent radiation therapy from 2012 to 2015 at Shandong Provincial Qianfoshan Hospital was performed. Granulocyte colony-stimulating factor was administered when grade 3 or 4 leukopenia and/or neutropenia occurred during radiation therapy, and no prophylactic granulocyte colony-stimulating factor was used in this study. Patients were classified into high and low granulocyte colony-stimulating factor group according the dosage of granulocyte colony-stimulating factor use during radiation therapy. The influence of granulocyte colony-stimulating factor on survival was investigated. In addition, the predict value of granulocyte colony-stimulating factor in concurrent chemoradiotherapy group and radiation therapy alone group was also evaluated, respectively. Results: A total of 231 patients were enrolled, with 56 in the high granulocyte colony-stimulating factor group and 175 in the low granulocyte colony-stimulating factor group. High dose of granulocyte colony-stimulating factor for the entire population group was associated with a favorable overall survival (hazard ratio [95% confidence interval] = 1.798 [1.260-2.568]; P = .001) and a longer progression-free survival (hazard ratio = 1.550 [1.127-2.132]; P = .002). However, compared with a lower granulocyte colony-stimulating factor, a higher granulocyte colony-stimulating factor was associated with significant better overall survival and progression-free survival in radiation therapy group, not in concurrent chemoradiotherapy group. Although there was no statistical significance in concurrent chemoradiotherapy group, the median overall survival and progression-free survival of patients in the higher granulocyte colony-stimulating factor group were longer than those in the lower group. Furthermore, the treatment strategy was also associated with the overall survival, not the progression-free survival. Conclusion: This study suggests that granulocyte colony-stimulating factor treatment during radiation therapy has favorable impact on outcome in patients with lung cancer. Besides, results showed that patients treated with concurrent chemoradiotherapy had better prognosis than those treated with radiation therapy alone.
Collapse
Affiliation(s)
- Rui Du
- 1 Department of Graduate, Weifang Medical University, Weifang, Shandong Pro, China
| | - Pingping Hu
- 2 Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong Pro, China
| | - Qiqi Liu
- 3 Department of Graduate, Shandong University, Jinan, Shandong Pro, China
| | - Jingxin Zhang
- 1 Department of Graduate, Weifang Medical University, Weifang, Shandong Pro, China
| | - Guodong Deng
- 3 Department of Graduate, Shandong University, Jinan, Shandong Pro, China
| | - Dan Hu
- 4 Department of Graduate, Taishan Medical University, Taian, Shandong Pro, China
| | - Jiandong Zhang
- 2 Department of Radiation Oncology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong Pro, China
| |
Collapse
|
22
|
CTLA4Ig-based reduced intensity conditioning and donor lymphocyte infusions for haploidentical transplantation in refractory aggressive B-cell lymphoma relapsing after an autograft: Early results from a pilot study. Exp Hematol 2019; 77:26-35.e1. [DOI: 10.1016/j.exphem.2019.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/24/2019] [Accepted: 08/14/2019] [Indexed: 12/15/2022]
|
23
|
Shi H, Lan L, Lv X, Sun L. Effect of pidotimod combined with azithromycin on children with mycoplasma pneumonia and the expression levels of IL-10 and G-CSF in serum. Exp Ther Med 2019; 18:1800-1806. [PMID: 31410140 PMCID: PMC6676206 DOI: 10.3892/etm.2019.7725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
This study explored the effect of pidotimod combined with azithromycin on children with mycoplasma pneumonia and the expression of interleukin-10 (IL-10) and granulocyte colony-stimulating factor (G-CSF) in serum. The clinical data of 149 children with mycoplasma pneumonia from May 2014 to May 2018 in Zhangqiu District Maternal and Child Health Care Hospital were collected. Among them, 70 children treated with azithromycin sequential therapy were the control group, and 79 children treated with the combination of pidotimod and azithromycin were the observation group. Double antibody sandwich enzyme-linked immunosorbent assay (ELISA) was used to determine the expression levels of IL-10 and G-CSF in serum before and after treatment. Pearson's correlation coefficient was used to analyze the correlation between IL-10 and G-CSF in serum. The total effective rate in the observation group (94.94%) was significantly higher than that in the control group (81.43%) (P<0.05). There was no significant difference in the expression levels of IL-10 and G-CSF between the two groups before treatment (P>0.05). The expression levels of IL-10 and G-CSF in the two groups after treatment were significantly lower than those before treatment (P<0.05). After treatment, the expression levels of IL-10 and G-CSF in serum in the observation group were significantly lower than those in the control group. There was a significant positive correlation between the expression levels of IL-10 and G-CSF before and after treatment in the observation group (P<0.05), and a significant positive correlation between the expression levels of IL-10 and G-CSF before and after treatment in the control group (P<0.05). Compared with sequential treatment with azithromycin alone, pidotimod combined with azithromycin significantly reduced the expression levels of IL-10 and G-CSF in serum of children with mycoplasma pneumonia, improved the curative effect and reduced the occurrence of adverse reactions, which has high application value in clinic.
Collapse
Affiliation(s)
- Hongxing Shi
- Department of Pediatrics, Zhangqiu District Maternal and Child Health Care Hospital, Jinan, Shandong 250200, P.R. China
| | - Limin Lan
- Department of Pharmacy, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Xianghong Lv
- Department of Pediatrics, The Affiliated Central Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Lizhi Sun
- Department of Medical Laboratory Diagnosis Center, Jinan Central Hospital, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
24
|
Atreya I, Kindermann M, Wirtz S. Innate lymphoid cells in intestinal cancer development. Semin Immunol 2019; 41:101267. [PMID: 30772139 DOI: 10.1016/j.smim.2019.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/25/2019] [Accepted: 02/05/2019] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is a highly prominent cause of cancer-related deaths worldwide. Although the functions of immune cells in the colorectal tumor microenvironment are complex and heterogeneous, dysregulated changes in the composition and activation state of immune cells are believed to represent key events supporting the establishment of pro- or anti-tumorigenic immune states. Recently, innate lymphoid cells (ILCs) emerged as central innate immune mediators during both gastrointestinal homeostasis and inflammatory pathologies. Hence, ILCs might also represent promising targets in the context of cancer therapy and are increasingly recognized as innate immune cells with potent immunomodulatory properties. In this review, we summarize the pleiotropic roles of the different ILC subsets for intestinal homeostasis and discuss the recent evidence on their potential involvement in the development and growth of intestinal cancers.
Collapse
Affiliation(s)
- Imke Atreya
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany
| | - Markus Kindermann
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-University, Erlangen, Germany.
| |
Collapse
|
25
|
G-CSF-induced macrophage polarization and mobilization may prevent acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2019; 54:1419-1433. [PMID: 30683906 DOI: 10.1038/s41409-019-0449-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/24/2018] [Accepted: 01/04/2019] [Indexed: 12/11/2022]
Abstract
Macrophages (MΦs) are an important immune cell population that are essential for tissue homeostasis and disease pathogenesis. MΦs are now classified as either M1, which produce pro-inflammatory cytokines, or M2, which produce antiinflammatory cytokines. The impact of granulocyte colony-stimulating factor (G-CSF) on MΦs in humans is unclear. Moreover, little is known about the association between MΦ subsets in allografts and the occurrence of acute graft-versus-host disease (aGVHD) in patients who undergo allogeneic hematopoietic stem cell transplantation (allo-HSCT). In the current study, we found that the M1/M2 ratio was markedly decreased in both G-CSF-treated bone marrow (post-BM) and G-CSF-treated peripheral blood from healthy donors. Post-BM MΦs exhibited reduced migration and increased phagocytosis. Moreover, post-BM MΦs reduced the percentage of Th1 and Tc1 lineages and increased the percentage of Th2, Tc2, and Treg lineages. Patients who received BM grafts with a higher M1/M2 ratio exhibited a higher incidence of grade 2-4 aGVHD. In summary, our data indicate that G-CSF decreases the M1/M2 ratio in BM grafts from healthy donors, which may contribute to preventing the occurrence of grade 2-4 aGVHD in patients after allo-HSCT.
Collapse
|
26
|
Yeo B, Redfern AD, Mouchemore KA, Hamilton JA, Anderson RL. The dark side of granulocyte-colony stimulating factor: a supportive therapy with potential to promote tumour progression. Clin Exp Metastasis 2018; 35:255-267. [PMID: 29968171 DOI: 10.1007/s10585-018-9917-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022]
Abstract
Granulocyte-colony stimulating factor (G-CSF) is one of several cytokines that can expand and mobilize haematopoietic precursor cells from bone marrow. In particular, G-CSF mobilizes neutrophils when the host is challenged by infection or tissue damage. Severe neutropenia, or febrile neutropenia is a life-threatening event that can be mitigated by administration of G-CSF. Consequently, G-CSF has been used to support patients undergoing chemotherapy who would otherwise require dose reduction due to neutropenia. Over the past 10-15 years it has become increasingly apparent, in preclinical tumour growth and metastasis models, that G-CSF can support tumour progression by mobilization of tumour-associated neutrophils which consequently promote tumour dissemination and metastasis. With the increasing use of G-CSF in the clinic, it is pertinent to ask if there is any evidence of a similar promotion of tumour progression in patients. Here, we have reviewed the preclinical and clinical data on the potential contribution of G-CSF to tumour progression. We conclude that, whilst the evidence for a promotion of metastasis is strong in preclinical models and that limited data indicate that high serum G-CSF levels in patients are associated with poorer prognosis, no studies published so far have revealed evidence of increased tumour progression associated with supportive G-CSF use during chemotherapy in patients. Analysis of G-CSF receptor positive cohorts within supportive trials, as well as studies of the role of G-CSF blockade in appropriate tumours in the absence of chemotherapy could yield clinically translatable findings.
Collapse
Affiliation(s)
- Belinda Yeo
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, 145 Studley Road, Heidelberg, VIC, 3084, Australia.,Austin Health, Heidelberg, VIC, 3084, Australia
| | | | - Kellie A Mouchemore
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia.,Department of Biochemistry & Molecular Biology, Monash University, Clayton, VIC, Australia
| | - John A Hamilton
- Arthritis and Inflammation Research Centre, Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, VIC, Australia
| | - Robin L Anderson
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, 145 Studley Road, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
27
|
Jeannin P, Paolini L, Adam C, Delneste Y. The roles of CSFs on the functional polarization of tumor-associated macrophages. FEBS J 2017; 285:680-699. [PMID: 29171156 DOI: 10.1111/febs.14343] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/03/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022]
Abstract
Macrophages have a central role in numerous physiological processes, such as immune defense, maintenance of tissue homeostasis, wound healing, and inflammation. Moreover, in numerous severe disorders, such as cancer or chronic inflammation, their functions can be profoundly affected. Macrophages continuously sense their environment and adapt their phenotypes and functions to the local requirements; this process is called plasticity. In addition to stress signals, metabolites, and direct cell-contact interactions with surrounding cells, numerous cytokines play a central role in controlling macrophage polarization. In this review, we will focus on three human macrophage differentiation factors: macrophage colony-stimulating factor (M-CSF), IL-34, and granulocyte M-CSF. These CSFs allow human monocyte survival, promote their differentiation into macrophages, and control macrophage polarization as they give rise to cells with different phenotype and functions. Based on recent observations, the role of granulocyte CSF on macrophage polarization is also addressed. Finally, our current knowledge on the expression of these growth factors in tumor microenvironment and their impact on the generation and polarization of tumor-associated macrophages are summarized.
Collapse
Affiliation(s)
- Pascale Jeannin
- CRCINA, INSERM, Université de Nantes, Université d'Angers, France.,Laboratory of Immunology and Allergology, University Hospital of Angers, France.,LabEx ImmunoGraftOnco, Angers, France
| | - Léa Paolini
- CRCINA, INSERM, Université de Nantes, Université d'Angers, France.,LabEx ImmunoGraftOnco, Angers, France
| | - Clement Adam
- CRCINA, INSERM, Université de Nantes, Université d'Angers, France.,LabEx ImmunoGraftOnco, Angers, France
| | - Yves Delneste
- CRCINA, INSERM, Université de Nantes, Université d'Angers, France.,Laboratory of Immunology and Allergology, University Hospital of Angers, France.,LabEx ImmunoGraftOnco, Angers, France
| |
Collapse
|
28
|
Ray AL, Berggren KL, Restrepo Cruz S, Gan GN, Beswick EJ. Inhibition of MK2 suppresses IL-1β, IL-6, and TNF-α-dependent colorectal cancer growth. Int J Cancer 2017; 142:1702-1711. [PMID: 29197088 DOI: 10.1002/ijc.31191] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 10/22/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) development and progression is associated with chronic inflammation. We have identified the MAPK-activated protein kinase 2 (MK2) pathway as a primary mediator of inflammation in CRC. MK2 signaling promotes production of proinflammatory cytokines IL-1β, IL-6 and TNF-α. These cytokines have been implicated in tumor growth, invasion and metastasis. For the first time, we investigate whether MK2 inhibition can improve outcome in two mouse models of CRC. In our azoxymethane/dextran sodium sulfate (AOM/DSS) model of colitis-associated CRC, MK2 inhibitor treatment eliminated murine tumor development. Using the implanted, syngeneic murine CRC cell line CT26, we observe significant tumor volume reduction following MK2 inhibition. Tumor cells treated with MK2 inhibitors produced 80% less IL-1β, IL-6 and TNF-α and demonstrated decreased invasion. Replenishment of downstream proinflammatory MK2-mediated cytokines (IL-1β, IL-6 and TNF-α) to tumors led to restoration of tumor proliferation and rapid tumor regrowth. These results demonstrate the importance of MK2 in driving proinflammatory cytokine production, its relevance to in vivo tumor proliferation and invasion. Inhibition of MK2 may represent an attractive therapeutic target to suppress tumor growth and progression in patients.
Collapse
Affiliation(s)
- Anita L Ray
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Kiersten L Berggren
- Department of Internal Medicine, Division of Medical Oncology, Section of Radiation Oncology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Sebastian Restrepo Cruz
- Department of Internal Medicine, Division of Medical Oncology, Section of Radiation Oncology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Gregory N Gan
- Department of Internal Medicine, Division of Medical Oncology, Section of Radiation Oncology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Ellen J Beswick
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
29
|
Lu L, Wang Y, Ou R, Feng Q, Ji L, Zheng H, Guo Y, Qi X, Kong ANT, Liu Z. DACT2 Epigenetic Stimulator Exerts Dual Efficacy for Colorectal Cancer Prevention and Treatment. Pharmacol Res 2017; 129:318-328. [PMID: 29199082 DOI: 10.1016/j.phrs.2017.11.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/30/2022]
Abstract
DACT2, a tumor suppressor gene in various tumors, is frequently down-regulated via hypermethylation. We found DACT2 gene expressions were dramatically silenced (P = 0.002, n = 8) in our clinical colorectal cancer (CRC) tissues, and TCGA data revealed DACT2 hypermethylation correlated to CRC poor prognosis (P = 0.0129, HR = 0.2153, n = 248). Thus, by screening twelve nutritional compounds, we aimed to find out an effective DACT2 epigenetic stimulator to determine whether DACT2 epigenetic restoration could reverse CRC tumorigenesis. We found that kaempferol significantly increased DACT2 expressions up to 3.47-fold in three CRC cells (HCT116, HT29, and YB5). Furthermore, kaempferol remarkably decreased DACT2 methylation (range: 19.58%-67.00%, P < 0.01), while increased unmethylated DACT2 by 13.72-fold (P < 0.01) via directly binding to DNA methyltransferases DNMT1. By epigenetic reactivating DACT2 transcription, kaempferol notably inhibited nuclear β-catenin expression to inactivate Wnt/β-catenin pathway, which consequently restricted CRC cells proliferation and migration. Moreover, in AOM/DSS-induced CRC tumorigenesis, kaempferol-demethylated DACT2 effectively decreased tumor load (range: 50.00%-73.52%, P < 0.05). By determining the chemopreventive and chemotherapeutic efficacy of a novel DACT2 demethylating stimulator, we demonstrated that DACT2 epigenetic restoration could successfully slow down and reverse CRC tumorigenesis.
Collapse
Affiliation(s)
- Linlin Lu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ying Wang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Rilan Ou
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Qian Feng
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Liyan Ji
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Hongming Zheng
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Yue Guo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Xiaoxiao Qi
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ah-Ng Tony Kong
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
30
|
|
31
|
Pan P, Kang S, Wang Y, Liu K, Oshima K, Huang YW, Zhang J, Yearsley M, Yu J, Wang LS. Black Raspberries Enhance Natural Killer Cell Infiltration into the Colon and Suppress the Progression of Colorectal Cancer. Front Immunol 2017; 8:997. [PMID: 28861089 PMCID: PMC5561013 DOI: 10.3389/fimmu.2017.00997] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/04/2017] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are an essential component of innate immunity against cancer development. Many studies have been conducted to evaluate immune-modulating effects using dietary compounds. Our laboratory has been investigating the chemopreventive potential of black raspberries (BRBs) and previously demonstrated their beneficial modulation of genetic and epigenetic biomarkers in patients with colorectal cancer (CRC). The current study investigated their potential on modulating NK cells. To avoid the excessive inflammation caused by the dextran sulfate sodium (DSS) treatment that leads to colitis, we treated the mice with overnight DSS so that it would slightly irritate the colon but still promote colon carcinogenesis with 100% incidence in both the ApcMin/+ mice and azoxymethane (AOM)-treated mice. A significant decrease of tissue-infiltrating NK cells along the progression of microadenoma-to-adenoma and adenoma-to-adenocarcinoma was observed in the ApcMin/+ /DSS and AOM/DSS mice, respectively. Depletion of NK cells significantly promoted the development of CRC, suggesting a critical role of NK cells in combating CRC progression. BRBs significantly suppressed the CRC progression and increased the number of tissue-infiltrating NK cells in both mouse models. Moreover, we further determined BRBs' effects on NK cells in the human biopsy specimens collected from our previously completed clinical trial, in which CRC patients consumed BRBs for an average of 4 weeks during a presurgical window. We observed an increased number and an enhanced cytotoxicity of NK cells by BRB intervention. The current study provides evidence that BRBs have the potential to enhance the tumor immunesurveillance of NK cells that can be beneficial in the setting of CRC prevention and treatment.
Collapse
Affiliation(s)
- Pan Pan
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Siwen Kang
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Youwei Wang
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Ka Liu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Kiyoko Oshima
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Yi-Wen Huang
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University, Columbus, OH, United States
| | - Martha Yearsley
- Department of Pathology, The Ohio State University, Columbus, OH, United States
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States.,Comprehensive Cancer Center, The James Cancer Hospital, The Ohio State University, Columbus, OH, United States
| | - Li-Shu Wang
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
32
|
Profiling of cytokines, chemokines and other soluble proteins as a potential biomarker in colorectal cancer and polyps. Cytokine 2017; 99:35-42. [PMID: 28689023 DOI: 10.1016/j.cyto.2017.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/20/2022]
Abstract
Soluble proteins including cytokines, chemokines and growth factors are small proteins that mediate and regulate immunity. They involved in the pathogenesis of many diseases including cancers. The concentration of these proteins in biological fluids (serum or plasma) and tissues in diseases may suggest pathway activation that leads to inflammatory response or disease progression. Therefore, these soluble proteins may be useful as a tool for screening, diagnosis classification between stages of disease or surveillance for therapy. Enzyme-linked immunosorbent assays (ELISA) and bioassay have been used as a gold standard in cytokine level measurements in clinical practice. However, these methods allow only single cytokine detection at a time and ineffective for screening purposes. Hence, the innovation of multiplexing technology allows measurement of many these soluble proteins simultaneously, thus allowing rapid, cost effective and better efficiency by using a minute amount of sample. In this study, we explored the profiles of key inflammatory cytokines, chemokines and other soluble proteins from the serum derived from colorectal carcinoma (CRC, n=20), colorectal polyps (P, n=20) and healthy volunteers (N, n=20) using multiplexed bead-based immunoassays. We aimed to evaluate if the levels of these soluble proteins can classify these groups of populations and explore the possible application of the soluble proteins as biomarkers in early stage screening and/or surveillance. We observed significant high IL-4, MIP-1β, FasL and TGF-β1 levels but lower levels for RANTES in P-derived serum as compared to N-derived serum. Significant high IL-8, VEGF, MIP-1β, Eotaxin and G-CSF observed in CRC-derived serum when compared to N-derived serum. Between CRC- and P-derived serum, significantly higher levels of IL-8, Eotaxin and G-CSF but lower levels for TGF-β1 were detected in CRC-derived serum. These preliminary results were obtained from small sample size and could be further validated with larger sample size cohort to produce a panel of biomarkers for CRC and P patients. Our findings might be useful in developing a disease-specific panel for biomarker screening assay. This could be used for early diagnosis and/or treatment surveillance.
Collapse
|
33
|
Wang Z, Xu L, Hu Y, Huang Y, Zhang Y, Zheng X, Wang S, Wang Y, Yu Y, Zhang M, Yuan K, Min W. miRNA let-7b modulates macrophage polarization and enhances tumor-associated macrophages to promote angiogenesis and mobility in prostate cancer. Sci Rep 2016; 6:25602. [PMID: 27157642 PMCID: PMC4860600 DOI: 10.1038/srep25602] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/18/2016] [Indexed: 11/09/2022] Open
Abstract
Macrophage polarization is a highly plastic physiological process that responds to a variety of environmental factors by changing macrophage phenotype and function. Tumor-associated macrophages (TAMs) are generally recognized as promoting tumor progression. As universal regulators, microRNAs (miRNAs) are functionally involved in numerous critical cellular processes including macrophage polarization. Let-7b, a miRNA, has differential expression patterns in inflamed tissues compared with healthy controls. However, whether and how miRNA let-7b regulates macrophage phenotype and function is unclear. In this report, we find that up-regulation of let-7b is characteristic of prostatic TAMs, and down-regulation of let-7b in TAMs leads to changes in expression profiles of inflammatory cytokines, such as IL-12, IL-23, IL-10 and TNF-α. As a result, TAMs treated with let-7b inhibitors reduce angiogenesis and prostate carcinoma (PCa) cell mobility. Let-7b may play a vital role in regulating macrophage polarization, thus modulating the prognosis of prostate cancer.
Collapse
Affiliation(s)
- Zhigang Wang
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Lu Xu
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Yinying Hu
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Yanqin Huang
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Yujuan Zhang
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Xiufen Zheng
- Departments of Surgery, Pathology, and Oncology, University of Western Ontario, London, Canada
| | - Shanshan Wang
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Yifan Wang
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Yanrong Yu
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Meng Zhang
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Keng Yuan
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
| | - Weiping Min
- Institute of Immunotherapy of Nanchang University, and Jiangxi Academy of Medical Sciences, Nanchang, China
- Jiangxi Provincial Key Laboratory of Immunotherapy, Nanchang, China
- Departments of Surgery, Pathology, and Oncology, University of Western Ontario, London, Canada
| |
Collapse
|
34
|
Yoo N, Lee HR, Shin SH, Sohn KY, Kim HJ, Han YH, Chong S, Kim MH, Yoon SY, Kim JW. PLAG (1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol) augments the therapeutic effect of pegfilgrastim on gemcitabine-induced neutropenia. Cancer Lett 2016; 377:25-31. [PMID: 27105612 DOI: 10.1016/j.canlet.2016.04.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 04/14/2016] [Accepted: 04/14/2016] [Indexed: 12/31/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) is widely used for preventing neutropenia during chemotherapy. Polyethylene glycol-conjugated granulocyte colony-stimulating factor (PEG-G-CSF, pegfilgrastim) serves the same purpose but has a longer half-life and greater stability than G-CSF. In this study, we investigated whether 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol, acetylated diglyceride (PLAG), augments the therapeutic effect of pegfilgrastim on chemotherapy-induced neutropenia. We compared neutrophil counts in four groups of mice: control mice, gemcitabine-treated mice, gemcitabine/pegfilgrastim-treated mice, and gemcitabine/pegfilgrastim/PLAG-treated mice. PLAG (50 mg/kg) was orally administered every day during the treatment course. CBC analysis showed that the group treated with PLAG experienced a dramatically increased neutrophil counts on the third day following pegfilgrastim treatment. PLAG had no effect on blood cell apoptosis and neutrophil release from bone marrow. Additionally, pegfilgrastim-induced CXCR2 expression in neutrophils was markedly decreased in PLAG-treated animals. These results suggest that PLAG plays a role in inhibiting neutrophil extravasation, giving rise to an increased number of circulating neutrophils when used with pegfilgrastim during gemcitabine treatment. These data support the potential for PLAG to be used with pegfilgrastim to treat or prevent chemotherapy-induced neutropenia by modulating neutrophil transmigration.
Collapse
Affiliation(s)
- Nina Yoo
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea; ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea
| | - Ha-Reum Lee
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea; ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea
| | - Su-Hyun Shin
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Ki-Young Sohn
- ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea
| | - Heung-Jae Kim
- ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea
| | - Yong-Hae Han
- ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea
| | - Saeho Chong
- ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea
| | - Myung-Hwan Kim
- Division of Gastroenterology, Department of Internal Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sun Young Yoon
- ENZYCHEM Lifesciences, 103-6, KAIST-ICC F741, Munjidong, Daejeon 305-732, Republic of Korea.
| | - Jae Wha Kim
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea; Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
35
|
Jiang Z, Zhang H, Wang Y, Yu B, Wang C, Liu C, Lu J, Chen F, Wang M, Yu X, Lin J, Pan X, Wang P, Zhu H. Altered Hepa1-6 cells by dimethyl sulfoxide (DMSO)-treatment induce anti-tumor immunity in vivo. Oncotarget 2016; 7:9340-52. [PMID: 26824185 PMCID: PMC4891044 DOI: 10.18632/oncotarget.7009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 01/01/2016] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy is the use of the immune system to treat cancer. Our current research proposed an optional strategy of activating immune system involving in cancer immunotherapy. When being treated with 2% DMSO in culture medium, Hepa1-6 cells showed depressed proliferation with no significant apoptosis or decreased viability. D-hep cells, Hepa1-6 cells treated with DMSO for 7 days, could restore to the higher proliferation rate in DMSO-free medium, but alteration of gene expression profile was irreversible. Interestingly, tumors from D-hep cells, not Hepa1-6 cells, regressed in wild-type C57BL/6 mice whereas D-hep cells exhibited similar tumorigenesis as Hep1–6 cells in immunodeficient mice. As expected, additional Hepa1-6 cells failed to form tumors in the D-hep-C57 mice in which D-hep cells were eliminated. Further research confirmed that D-hep-C57 mice established anti-tumor immunity against Hepa1-6 cells. Our research proposed viable tumor cells with altered biological features by DMSO-treatment could induce anti-tumor immunity in vivo.
Collapse
Affiliation(s)
- Zhengyu Jiang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China.,Department of Anesthesiology, Second Military Medical University, Shanghai, P.R. China
| | - Hongxia Zhang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Ye Wang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Bin Yu
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Chen Wang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Changcheng Liu
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Juan Lu
- Training Department, Second Military Medical University, Shanghai, P.R. China
| | - Fei Chen
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Minjun Wang
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Xinlu Yu
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| | - Jiahao Lin
- School of Clinic Medicine, Second Military Medical University, Shanghai, P.R. China
| | - Xinghua Pan
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Pin Wang
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai, P.R. China
| | - Haiying Zhu
- Department of Cell Biology, Second Military Medical University, Shanghai, P.R. China.,Center for Stem Cell and Medicine, The Graduate School, Second Military Medical University, Shanghai, P.R. China
| |
Collapse
|
36
|
G-CSF is a key modulator of MDSC and could be a potential therapeutic target in colitis-associated colorectal cancers. Protein Cell 2016; 7:130-40. [PMID: 26797765 PMCID: PMC4742385 DOI: 10.1007/s13238-015-0237-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 11/30/2015] [Indexed: 12/22/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) is an essential regulator of neutrophil trafficking and is highly expressed in multiple tumors. Myeloid derived suppressor cells (MDSCs) promote neoplastic progression through multiple mechanisms by immune suppression. Despite the findings of G-CSF function in colon cancer progression, the precise mechanism of G-CSF on MDSCs regulation and its blockade effects on tumor growth remains a worthy area of investigation. In this study we observed an overexpression of G-CSF in a mouse colitis-associated cancer (CAC) model, which was consistent with the accumulation of MDSCs in mouse colon tissues. Further in vitro studies demonstrated that G-CSF could promote MDSCs survival and activation through signal transducer and activator of transcription 3 (STAT3) signaling pathway. Moreover, compared with isotype control, anti-G-CSF mAb treatment demonstrated reduced MDSC accumulation, which led to a marked decrease in neoplasm size and number in mice. Our results indicated that G-CSF is a critical regulating molecule in the migration, proliferation and function maintenance of MDSCs, which could be a potential therapeutic target for colitis-associated cancer.
Collapse
|
37
|
Yang JZ, Zhang JQ, Sun LX. Mechanisms for T cell tolerance induced with granulocyte colony-stimulating factor. Mol Immunol 2015; 70:56-62. [PMID: 26703218 DOI: 10.1016/j.molimm.2015.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 12/15/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) has been widely accepted as a mediator of T cell tolerance. The immune modulatory effect of G-CSF on T cells is believed to be mediated exclusively through other effector cells, such as monocytes, tolerogenic dendritic cells (DC), and myeloid-derived suppressor cells. Recent advances confirmed the direct effects of G-CSF in inducing immune tolerance of T cells through the G-CSF-G-CSF receptor pathway and related molecular mechanisms. This review aims to summarize the findings associated with the direct and indirect mechanisms for T cell tolerance induced with G-CSF. The role of G-CSF in preventing graft-versus-host disease (GVHD) and in treating autoimmune diseases (ADs) is also discussed. It is conceivable that G-CSF and immune cell compositions, such as tolerogenic DC and CD4(+)CD25(+)Foxp3(+) T cells, modulated by G-CSF could become an integral part of the immunomodulatory therapies against GVHD and ADs in the future.
Collapse
Affiliation(s)
- Jian-Zhu Yang
- Department of Pathology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jin-Qiao Zhang
- Department of Hematology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li-Xia Sun
- Department of Hematology, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|