51
|
Martinez-Vidal L, Murdica V, Venegoni C, Pederzoli F, Bandini M, Necchi A, Salonia A, Alfano M. Causal contributors to tissue stiffness and clinical relevance in urology. Commun Biol 2021; 4:1011. [PMID: 34446834 PMCID: PMC8390675 DOI: 10.1038/s42003-021-02539-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 08/02/2021] [Indexed: 02/07/2023] Open
Abstract
Mechanomedicine is an emerging field focused on characterizing mechanical changes in cells and tissues coupled with a specific disease. Understanding the mechanical cues that drive disease progression, and whether tissue stiffening can precede disease development, is crucial in order to define new mechanical biomarkers to improve and develop diagnostic and prognostic tools. Classically known stromal regulators, such as fibroblasts, and more recently acknowledged factors such as the microbiome and extracellular vesicles, play a crucial role in modifications to the stroma and extracellular matrix (ECM). These modifications ultimately lead to an alteration of the mechanical properties (stiffness) of the tissue, contributing to disease onset and progression. We describe here classic and emerging mediators of ECM remodeling, and discuss state-of-the-art studies characterizing mechanical fingerprints of urological diseases, showing a general trend between increased tissue stiffness and severity of disease. Finally, we point to the clinical potential of tissue stiffness as a diagnostic and prognostic factor in the urological field, as well as a possible target for new innovative drugs.
Collapse
Affiliation(s)
- Laura Martinez-Vidal
- Vita-Salute San Raffaele University, Milan, Italy.
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy.
| | - Valentina Murdica
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Chiara Venegoni
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Filippo Pederzoli
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Marco Bandini
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Andrea Salonia
- Vita-Salute San Raffaele University, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| | - Massimo Alfano
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
52
|
lncRNA DCST1-AS1 Facilitates Oral Squamous Cell Carcinoma by Promoting M2 Macrophage Polarization through Activating NF- κB Signaling. J Immunol Res 2021; 2021:5524231. [PMID: 34414241 PMCID: PMC8369177 DOI: 10.1155/2021/5524231] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/18/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
lncRNAs are related to the progression of various diseases, including oral squamous cell carcinoma (OSCC), which is a common squamous cell carcinoma of the head and neck. Tumor-associated macrophages and tumor cells are significant components of tumor microenvironment. M2 polarization of tumor-associated macrophages is a crucial actor in tumor malignancy and metastasis. In this study, we studied the molecular mechanism of lncRNA DCST1-AS1 in OSCC. Here, we reported that DCST1-AS1 was significantly increased in OSCC cells. We found that loss of DCST1-AS1 obviously inhibited the proliferation, migration, and invasion of OSCC cells and xenograft tumor growth. Meanwhile, silencing of DCST1-AS1 also repressed the percentage of macrophages expressing M2 markers CD206 and CD11b. DCST1-AS1 shRNA enhanced the percentage of macrophages expressing M1 markers CD80 and CD11c. Then, we observed that loss of DCST1-AS1 suppressed OSCC progression via inactivating NF-κB signaling. As well established, NF-κB signaling exerts critical roles in tumor progression, and our study proved that DCST1-AS1 could regulate NF-κB signaling. We proved that blocking the NF-κB pathway using antagonists greatly downregulated OSCC progression and M2 macrophage polarization induced by the overexpression of DCST1-AS1. To sum up, we reported that DCST1-AS1 plays an important role in modulating OSCC tumorigenicity and M2 macrophage polarization through regulating the NF-κB pathway.
Collapse
|
53
|
Wang S, Zheng Y, Yang F, Zhu L, Zhu XQ, Wang ZF, Wu XL, Zhou CH, Yan JY, Hu BY, Kong B, Fu DL, Bruns C, Zhao Y, Qin LX, Dong QZ. The molecular biology of pancreatic adenocarcinoma: translational challenges and clinical perspectives. Signal Transduct Target Ther 2021; 6:249. [PMID: 34219130 PMCID: PMC8255319 DOI: 10.1038/s41392-021-00659-4] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is an increasingly common cause of cancer mortality with a tight correspondence between disease mortality and incidence. Furthermore, it is usually diagnosed at an advanced stage with a very dismal prognosis. Due to the high heterogeneity, metabolic reprogramming, and dense stromal environment associated with pancreatic cancer, patients benefit little from current conventional therapy. Recent insight into the biology and genetics of pancreatic cancer has supported its molecular classification, thus expanding clinical therapeutic options. In this review, we summarize how the biological features of pancreatic cancer and its metabolic reprogramming as well as the tumor microenvironment regulate its development and progression. We further discuss potential biomarkers for pancreatic cancer diagnosis, prediction, and surveillance based on novel liquid biopsies. We also outline recent advances in defining pancreatic cancer subtypes and subtype-specific therapeutic responses and current preclinical therapeutic models. Finally, we discuss prospects and challenges in the clinical development of pancreatic cancer therapeutics.
Collapse
Affiliation(s)
- Shun Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Yan Zheng
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Feng Yang
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Le Zhu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xiao-Qiang Zhu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhe-Fang Wang
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Xiao-Lin Wu
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Cheng-Hui Zhou
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Jia-Yan Yan
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bei-Yuan Hu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Bo Kong
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - De-Liang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Christiane Bruns
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yue Zhao
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany.
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
| | - Qiong-Zhu Dong
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
- Key laboratory of whole-period monitoring and precise intervention of digestive cancer, Shanghai Municipal Health Commission (SMHC), Shanghai, China.
| |
Collapse
|
54
|
Ahmad RS, Eubank TD, Lukomski S, Boone BA. Immune Cell Modulation of the Extracellular Matrix Contributes to the Pathogenesis of Pancreatic Cancer. Biomolecules 2021; 11:biom11060901. [PMID: 34204306 PMCID: PMC8234537 DOI: 10.3390/biom11060901] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/07/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a five-year survival rate of only 9%. PDAC is characterized by a dense, fibrotic stroma composed of extracellular matrix (ECM) proteins. This desmoplastic stroma is a hallmark of PDAC, representing a significant physical barrier that is immunosuppressive and obstructs penetration of cytotoxic chemotherapy agents into the tumor microenvironment (TME). Additionally, dense ECM promotes hypoxia, making tumor cells refractive to radiation therapy and alters their metabolism, thereby supporting proliferation and survival. In this review, we outline the significant contribution of fibrosis to the pathogenesis of pancreatic cancer, with a focus on the cross talk between immune cells and pancreatic stellate cells that contribute to ECM deposition. We emphasize the cellular mechanisms by which neutrophils and macrophages, specifically, modulate the ECM in favor of PDAC-progression. Furthermore, we investigate how activated stellate cells and ECM influence immune cells and promote immunosuppression in PDAC. Finally, we summarize therapeutic strategies that target the stroma and hinder immune cell promotion of fibrogenesis, which have unfortunately led to mixed results. An enhanced understanding of the complex interactions between the pancreatic tumor ECM and immune cells may uncover novel treatment strategies that are desperately needed for this devastating disease.
Collapse
Affiliation(s)
- Ramiz S. Ahmad
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA;
| | - Timothy D. Eubank
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Slawomir Lukomski
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Brian A. Boone
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA;
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
- Correspondence:
| |
Collapse
|
55
|
Chopra A, Zamora R, Vodovotz Y, Hodges JC, Barclay D, Brand R, Simmons RL, Lee KK, Paniccia A, Murthy P, Lotze MT, Boone BA, Zureikat AH. Baseline Plasma Inflammatory Profile Is Associated With Response to Neoadjuvant Chemotherapy in Patients With Pancreatic Adenocarcinoma. J Immunother 2021; 44:185-192. [PMID: 33935273 PMCID: PMC8102434 DOI: 10.1097/cji.0000000000000370] [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: 09/11/2020] [Accepted: 02/12/2021] [Indexed: 11/26/2022]
Abstract
Despite its increased application in pancreatic ductal adenocarcinoma (PDAC), complete response to neoadjuvant therapy (NAT) is rare. Given the critical role of host immunity in regulating cancer, we sought to correlate baseline inflammatory profiles to significant response to NAT. PDAC patients receiving NAT were classified as responders (R) or nonresponders (NR) by carbohydrate antigen 19-9 response, pathologic tumor size, and lymph node status in the resected specimen. Baseline (treatment-naive) plasma was analyzed to determine levels of 27 inflammatory mediators. Logistic regression was used to correlate individual mediators with response. Network analysis and Pearson correlation maps were derived to determine baseline inflammatory mediator profiles. Forty patients (20R and 20NR) met study criteria. The R showed significantly higher overall survival (59.4 vs. 21.25 mo, P=0.002) and disease-free survival (50.97 vs. 10.60 mo, P=0.005), compared with NR. soluble interleukin-2 receptor alpha was a significant predictor of no response to NAT (P=0.045). Analysis of inflammatory profiles using the Pearson heat map analysis followed by network analysis depicted increased inflammatory network complexity in NR compared with R (1.69 vs. 1), signifying a more robust baseline inflammatory status of NR. A panel of inflammatory mediators identified by logistic regression and Fischer score analysis was used to create a potential decision tree to predict NAT response. We demonstrate that baseline inflammatory profiles are associated with response to NAT in PDAC, and that an upregulated inflammatory status is associated with a poor response to NAT. Further analysis into the role of inflammatory mediators as predictors of chemotherapy response is warranted.
Collapse
Affiliation(s)
- Asmita Chopra
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ruben Zamora
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jacob C. Hodges
- Wolff Center of UPMC, University of Pittsburgh, Pittsburgh, PA, USA
| | - Derek Barclay
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Randall Brand
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Richard L. Simmons
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kenneth K Lee
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alessandro Paniccia
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Pranav Murthy
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael T. Lotze
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Departments of Immunology and Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian A. Boone
- Department of Surgery and Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, USA
| | - Amer H. Zureikat
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
56
|
Lu Z, Long Y, Wang Y, Wang X, Xia C, Li M, Zhang Z, He Q. Phenylboronic acid modified nanoparticles simultaneously target pancreatic cancer and its metastasis and alleviate immunosuppression. Eur J Pharm Biopharm 2021; 165:164-173. [PMID: 34020022 DOI: 10.1016/j.ejpb.2021.05.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022]
Abstract
Pancreatic ductal adenocarcinoma is one of the most lethal malignant tumors, its drug resistance, immunosuppression and metastasis makes the traditional chemotherapy and immunotherapy inefficient. Here we confirmed a 3-aminophenylboronic acid-modified low molecular weight heparin-D-α-tocopheryl succinate micellar nanoparticle (PBA-LMWH-TOS NP, PLT NP) could inhibit orthotopic pancreatic tumor and its spontaneous metastases. The small particle size and high affinity of PBA to sialic acid residue (SA) made PLT/PTX NPs significantly targeted and accumulated in both pancreatic tumor tissues and metastases. The immunosuppressive microenvironment of pancreatic tumor was most caused by the infiltration of immunosuppressive cells, mainly myeloid-derived suppressor cells (MDSCs). We first reported that P-selectin glycoprotein ligand-1 (PSGL-1) was expressed on the surfaces of MDSCs in pancreatic tumor tissues. Meanwhile, we found that LMWH could inhibit the early stage of adhesion cascade between vascular endothelial cells (VECs) and MDSCs by interfering with P-selectin/PSGL-1 binding, thus inhibiting MDSC recruitment to pancreatic tumor tissues. The therapeutic results indicated that PLT/PTX NPs could significantly improve the immune microenvironment of pancreatic tumor and inhibit spontaneous metastases. This nanosystem provides a new immune microenvironment regulation mechanism based on carrier materials in pancreatic tumor, and has high clinical application potential.
Collapse
Affiliation(s)
- Zhengze Lu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yang Long
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yashi Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xuhui Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Chunyu Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China.
| |
Collapse
|
57
|
Ma J, Shayiti F, Ma J, Wei M, Hua T, Zhang R, Su J, Chen P. Tumor-associated macrophage-derived CCL5 promotes chemotherapy resistance and metastasis in prostatic cancer. Cell Biol Int 2021; 45:2054-2062. [PMID: 34003531 DOI: 10.1002/cbin.11630] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/25/2021] [Accepted: 05/16/2021] [Indexed: 01/06/2023]
Abstract
The crosstalk between tumor microenvironment and cancer cells is emerging as a critical determinant in tumor progression. However, the underlying mechanism of tumor microenvironment-induced cancer development remains controversial. Here, our study provides evidence to suggest that tumor-associated macrophage (TAM) enrichment is found in chemoresistant prostatic tumor tissues. Those TAMs are demonstrated to promote chemoresistance and distant metastasis in prostatic cancer through secretion of CCL5. Mechanistically, TAM coculture or additional CCL5 can mediate the STAT3-dependent epithelial-mesenchymal transition process, resulting in distant metastasis in prostatic cancer. Meanwhile, activation of STAT3 induced by CCL5 can mediate upregulation of the transcription factor Nanog, leading to drug resistance. In vivo study further demonstrated that blockade of STAT3 signals significantly reverses chemoresistance and suppresses lung metastasis in colorectal tumor-bearing mice, suggesting a novel strategy for clinical prostatic cancer treatment.
Collapse
Affiliation(s)
- Jian Ma
- Urology Department, Xinjiang Medical University Affiliated Tumor Hospital, Ürümqi, Xinjiang, China
| | - Fuerhaiti Shayiti
- Urology Department, Xinjiang Medical University Affiliated Tumor Hospital, Ürümqi, Xinjiang, China
| | - Jing Ma
- Comprehensive Internal Medicine Department, First Affiliated Hospital of Xinjiang Medical University, Ürümqi, Xinjiang, China
| | - Meng Wei
- Department of Medical Analysis, Xinjiang Zhizhen Medical Laboratory Science Co., Ltd., Xinjiang, China
| | - Tingting Hua
- Department of Ultrasound, Xinjiang Medical University Affiliated Tumor Hospital, Ürümqi, Xinjiang, China
| | - Rong Zhang
- Urology Department, Xinjiang Medical University Affiliated Tumor Hospital, Ürümqi, Xinjiang, China
| | - Junyan Su
- Department of Medical, Lifehealthcare Clinical Laboratories, Beijing, China
| | - Peng Chen
- Urology Department, Xinjiang Medical University Affiliated Tumor Hospital, Ürümqi, Xinjiang, China
| |
Collapse
|
58
|
Narasimhan A, Zhong X, Au EP, Ceppa EP, Nakeeb A, House MG, Zyromski NJ, Schmidt CM, Schloss KNH, Schloss DEI, Liu Y, Jiang G, Hancock BA, Radovich M, Kays JK, Shahda S, Couch ME, Koniaris LG, Zimmers TA. Profiling of Adipose and Skeletal Muscle in Human Pancreatic Cancer Cachexia Reveals Distinct Gene Profiles with Convergent Pathways. Cancers (Basel) 2021; 13:1975. [PMID: 33923976 PMCID: PMC8073275 DOI: 10.3390/cancers13081975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/14/2021] [Indexed: 01/06/2023] Open
Abstract
The vast majority of patients with pancreatic ductal adenocarcinoma (PDAC) suffer cachexia. Although cachexia results from concurrent loss of adipose and muscle tissue, most studies focus on muscle alone. Emerging data demonstrate the prognostic value of fat loss in cachexia. Here we sought to identify the muscle and adipose gene profiles and pathways regulated in cachexia. Matched rectus abdominis muscle and subcutaneous adipose tissue were obtained at surgery from patients with benign conditions (n = 11) and patients with PDAC (n = 24). Self-reported weight loss and body composition measurements defined cachexia status. Gene profiling was done using ion proton sequencing. Results were queried against external datasets for validation. 961 DE genes were identified from muscle and 2000 from adipose tissue, demonstrating greater response of adipose than muscle. In addition to known cachexia genes such as FOXO1, novel genes from muscle, including PPP1R8 and AEN correlated with cancer weight loss. All the adipose correlated genes including SCGN and EDR17 are novel for PDAC cachexia. Pathway analysis demonstrated shared pathways but largely non-overlapping genes in both tissues. Age related muscle loss predominantly had a distinct gene profiles compared to cachexia. This analysis of matched, externally validate gene expression points to novel targets in cachexia.
Collapse
Affiliation(s)
- Ashok Narasimhan
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Xiaoling Zhong
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
| | - Ernie P. Au
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Eugene P. Ceppa
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Atilla Nakeeb
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Michael G. House
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Nicholas J. Zyromski
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - C. Max Schmidt
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Katheryn N. H. Schloss
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Daniel E. I. Schloss
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Yunlong Liu
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
| | - Guanglong Jiang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Bradley A. Hancock
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Milan Radovich
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
| | - Joshua K. Kays
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
| | - Safi Shahda
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Marion E. Couch
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Department of Otolaryngology—Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Leonidas G. Koniaris
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.N.); (X.Z.); (E.P.A.); (E.P.C.); (A.N.); (M.G.H.); (N.J.Z.); (C.M.S.); (K.N.H.S.); (D.E.I.S.); (B.A.H.); (M.R.); (J.K.K.); (L.G.K.)
- IUPUI Center for Cachexia Research Innovation and Therapy, Indianapolis, IN 46202, USA; (Y.L.); (S.S.); (M.E.C.)
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Cancer Center, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
- Department of Otolaryngology—Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
59
|
Xavier CPR, Castro I, Caires HR, Ferreira D, Cavadas B, Pereira L, Santos LL, Oliveira MJ, Vasconcelos MH. Chitinase 3-like-1 and fibronectin in the cargo of extracellular vesicles shed by human macrophages influence pancreatic cancer cellular response to gemcitabine. Cancer Lett 2021; 501:210-223. [PMID: 33212158 DOI: 10.1016/j.canlet.2020.11.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/25/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022]
Abstract
Tumour-associated macrophages have been implicated in pancreatic ductal adenocarcinoma (PDAC) therapy response and Extracellular vesicles (EVs) shed by macrophages might have a role in this process. Here, we demonstrated that large EVs released by anti-inflammatory human macrophages decreased PDAC cellular sensitivity to gemcitabine. Using proteomic analysis, chitinase 3-like-1 (CHI3L1) and fibronectin (FN1) were identified as two of the most abundant proteins in the cargo of macrophages-derived EVs. Overexpression of CHI3L1 and FN1, using recombinant human proteins, induced PDAC cellular resistance to gemcitabine through ERK (extracellular-signal-regulated kinase) activation. Inhibition of CHI3L1 and FN1 by pentoxifylline and pirfenidone, respectively, partially reverted gemcitabine resistance. In PDAC patient samples, CHI3L1 and FN1 were expressed in the stroma, associated with the high presence of macrophages. The Cancer Genome Atlas analysis revealed an association between CHI3L1 and FN1 gene expression, overall survival of PDAC patients, gemcitabine response, and macrophage infiltration. Altogether, our data identifies CHI3L1 and FN1 as potential targets for pharmacological inhibition in PDAC. Further pre-clinical in vivo work is warranted to study the possibility of repurposing pentoxifylline and pirfenidone as adjuvant therapies for PDAC treatment.
Collapse
Affiliation(s)
- Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Inês Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Hugo R Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Dylan Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Experimental Pathology and Therapeutics Group, IPO - Instituto Português de Oncologia, Porto, Portugal
| | - Bruno Cavadas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Genetic Diversity Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Luisa Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Genetic Diversity Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | - Lúcio L Santos
- Experimental Pathology and Therapeutics Group, IPO - Instituto Português de Oncologia, Porto, Portugal; ICBAS - Biomedical Sciences Institute Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria J Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal; Tumour and Microenvironment Interactions Group, INEB - Instituto Nacional de Engenharia Biomédica, Porto, Portugal
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal.
| |
Collapse
|
60
|
Moore AM, Zhou L, Cui J, Li L, Wu N, Yu A, Poddar S, Liang K, Abt ER, Kim S, Ghukasyan R, Khachatourian N, Pagano K, Elliott I, Dann AM, Riahi R, Le T, Dawson DW, Radu CG, Donahue TR. NAD + depletion by type I interferon signaling sensitizes pancreatic cancer cells to NAMPT inhibition. Proc Natl Acad Sci U S A 2021; 118:e2012469118. [PMID: 33597293 PMCID: PMC7923374 DOI: 10.1073/pnas.2012469118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Emerging evidence suggests that intratumoral interferon (IFN) signaling can trigger targetable vulnerabilities. A hallmark of pancreatic ductal adenocarcinoma (PDAC) is its extensively reprogrammed metabolic network, in which nicotinamide adenine dinucleotide (NAD) and its reduced form, NADH, are critical cofactors. Here, we show that IFN signaling, present in a subset of PDAC tumors, substantially lowers NAD(H) levels through up-regulating the expression of NAD-consuming enzymes PARP9, PARP10, and PARP14. Their individual contributions to this mechanism in PDAC have not been previously delineated. Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme in the NAD salvage pathway, a dominant source of NAD in cancer cells. We found that IFN-induced NAD consumption increased dependence upon NAMPT for its role in recycling NAM to salvage NAD pools, thus sensitizing PDAC cells to pharmacologic NAMPT inhibition. Their combination decreased PDAC cell proliferation and invasion in vitro and suppressed orthotopic tumor growth and liver metastases in vivo.
Collapse
Affiliation(s)
- Alexandra M Moore
- Department of Surgery, University of California, Los Angeles, CA 90095
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Lei Zhou
- Department of Surgery, University of California, Los Angeles, CA 90095
- Department of Pancreatic and Thyroidal Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Jing Cui
- Department of Surgery, University of California, Los Angeles, CA 90095
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430022, China
| | - Luyi Li
- Department of Surgery, University of California, Los Angeles, CA 90095
| | - Nanping Wu
- Department of Surgery, University of California, Los Angeles, CA 90095
| | - Alice Yu
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Soumya Poddar
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
- Ahmanson Translational Imaging Division, University of California, Los Angeles, CA 90095
| | - Keke Liang
- Department of Surgery, University of California, Los Angeles, CA 90095
- Department of Pancreatic and Thyroidal Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Evan R Abt
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
- Ahmanson Translational Imaging Division, University of California, Los Angeles, CA 90095
| | - Stephanie Kim
- Department of Surgery, University of California, Los Angeles, CA 90095
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Razmik Ghukasyan
- Department of Surgery, University of California, Los Angeles, CA 90095
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | | | - Kristina Pagano
- Department of Surgery, University of California, Los Angeles, CA 90095
| | - Irmina Elliott
- Department of Surgery, University of California, Los Angeles, CA 90095
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Amanda M Dann
- Department of Surgery, University of California, Los Angeles, CA 90095
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Rana Riahi
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
| | - Thuc Le
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
- Ahmanson Translational Imaging Division, University of California, Los Angeles, CA 90095
| | - David W Dawson
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| | - Caius G Radu
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095;
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
- Ahmanson Translational Imaging Division, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| | - Timothy R Donahue
- Department of Surgery, University of California, Los Angeles, CA 90095;
- David Geffen School of Medicine, University of California, Los Angeles, CA 90095
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095
- Ahmanson Translational Imaging Division, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| |
Collapse
|
61
|
Yang S, Liu Q, Liao Q. Tumor-Associated Macrophages in Pancreatic Ductal Adenocarcinoma: Origin, Polarization, Function, and Reprogramming. Front Cell Dev Biol 2021; 8:607209. [PMID: 33505964 PMCID: PMC7829544 DOI: 10.3389/fcell.2020.607209] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy. PDAC is only cured by surgical resection in its early stage, but there remains a relatively high possibility of recurrence. The development of PDAC is closely associated with the tumor microenvironment. Tumor-associated macrophages (TAMs) are one of the most abundant immune cell populations in the pancreatic tumor stroma. TAMs are inclined to M2 deviation in the tumor microenvironment, which promotes and supports tumor behaviors, including tumorigenesis, immune escape, metastasis, and chemotherapeutic resistance. Herein, we comprehensively reviewed the latest researches on the origin, polarization, functions, and reprogramming of TAMs in PDAC.
Collapse
Affiliation(s)
- Sen Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
62
|
Wu S, Pan Y, Mao Y, Chen Y, He Y. Current progress and mechanisms of bone metastasis in lung cancer: a narrative review. Transl Lung Cancer Res 2021; 10:439-451. [PMID: 33569325 PMCID: PMC7867745 DOI: 10.21037/tlcr-20-835] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lung cancer is a kind of malignant tumor with rapid progression and poor prognosis. Distant metastasis has been the main cause of mortality among lung cancer patients. Bone is one of the most common sites. Among all lung cancer patients with bone metastasis, most of them are osteolytic metastasis. Some serious clinical consequences like bone pain, pathological fractures, spinal instability, spinal cord compression and hypercalcemia occur as well. Since the severity of bone metastasis in lung cancer, it is undoubtedly necessary to know how lung cancer spread to bone, how can we diagnose it and how can we treat it. Here, we reviewed the process, possible mechanisms, diagnosis methods and current treatment of bone metastasis in lung cancer. We divided the process of bone metastasis in lung cancer into three steps: tumor invasion, tumor cell migration and invasion in bone tissue. It may be influenced by genetic factors, microenvironment and other adhesion-related factors. Imaging examination, laboratory examination, and pathological examination are used to diagnose lung cancer metastasis to bone. Surgery, radiotherapy, targeted therapy, bisphosphonate, radiation therapy and chemotherapy are the common clinical treatment methods currently. We also found some problems remained to be solved. For example, drugs for skeletal related events mainly target on osteoclasts at present, which increase the ratio of patients in osteoporosis and fractures in the long term. In all, this review provides the direction for future research on bone metastasis in lung cancer.
Collapse
Affiliation(s)
- Shengyu Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Yue Pan
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Yanyu Mao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Yu Chen
- Spine Center, Orthopedic department, Shanghai Changzheng Hospital, Shanghai, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| |
Collapse
|
63
|
Yu Y, Schuck K, Friess H, Kong B. Targeting Aggressive Fibroblasts to Enhance the Treatment of Pancreatic Cancer. Expert Opin Ther Targets 2020; 25:5-13. [PMID: 33246383 DOI: 10.1080/14728222.2021.1857727] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant cancer entities, which is characterized by abundant desmoplastic stroma. The stroma consists of extracellular matrix, infiltrating immune cells, cancer-associated fibroblasts (CAFs) and others. Depending on environmental cues, CAFs can be highly heterogeneous and play context-dependent roles in PDAC progression. AREAS COVERED In this article, we discuss the biological significance of CAFs heterogeneity (oncogenic vs. tumor-suppressive) in pancreatic carcinogenesis. In particular, the complex interaction between CAFs and infiltrating immune cells has a determinant role in defining the stromal composition. A subset of PDAC patients may benefit from anti-CAFs therapy. EXPERT OPINION Co-defined by CAFs and infiltrating immune cells, the prognostic stroma signature is clinically relevant in a subset of human PDAC. This is the patient population which may benefit from future anti-stroma or anti-CAFs therapies. To consider CAF heterogeneity is crucial for designing anti-stroma studies. Here, reliable and traceable subtype-specific markers for CAFs are urgently needed to dissect the biological impact of CAF heterogeneity on PDAC development spatiotemporally. Given the significant contribution of CAFs to immunosuppressive microenvironment of PDAC, it is conceivable to combine anti-CAFs therapy with immunotherapy. To implement a CAF-subtype specific therapy is crucially important to improve the effectiveness of current treatments including chemotherapies and immunotherapy.
Collapse
Affiliation(s)
- Yuanyuan Yu
- Department of Surgery, Klinikum Rechts Der Isar, School of Medicine, Technical University of Munich (TUM) , Munich, Germany
| | - Kathleen Schuck
- Department of Surgery, Klinikum Rechts Der Isar, School of Medicine, Technical University of Munich (TUM) , Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum Rechts Der Isar, School of Medicine, Technical University of Munich (TUM) , Munich, Germany
| | - Bo Kong
- Department of Surgery, Klinikum Rechts Der Isar, School of Medicine, Technical University of Munich (TUM) , Munich, Germany
| |
Collapse
|
64
|
Villalobos-Ayala K, Ortiz Rivera I, Alvarez C, Husain K, DeLoach D, Krystal G, Hibbs ML, Jiang K, Ghansah T. Apigenin Increases SHIP-1 Expression, Promotes Tumoricidal Macrophages and Anti-Tumor Immune Responses in Murine Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12123631. [PMID: 33291556 PMCID: PMC7761852 DOI: 10.3390/cancers12123631] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) has an extremely poor prognosis due to the expansion of immunosuppressive myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) in the inflammatory tumor microenvironment (TME), which halts the recruitment of effector immune cells and renders immunotherapy ineffective. Thus, the identification of new molecular targets that can modulate the immunosuppressive TME is warranted for PC intervention. Src Homology-2 (SH2) domain-containing Inositol 5'-Phosphatase-1 (SHIP-1) is a lipid signaling protein and a regulator of myeloid cell development and function. Herein, we used the bioflavonoid apigenin (API) to reduce inflammation in different PC models. Wild type mice harboring heterotopic or orthotopic PC were treated with API, which induced SHIP-1 expression, reduced inflammatory tumor-derived factors (TDF), increased the proportion of tumoricidal macrophages and enhanced anti-tumor immune responses, resulting in a reduction in tumor burden compared to vehicle-treated PC mice. In contrast, SHIP-1-deficient mice exhibited an increased tumor burden and displayed augmented proportions of pro-tumor macrophages. These results provide further support for the importance of SHIP-1 expression in promoting pro-tumor macrophage development in the pancreatic TME. Our findings suggest that agents augmenting SHIP-1 expression may provide novel therapeutic options for the treatment of PC.
Collapse
Affiliation(s)
- Krystal Villalobos-Ayala
- Department of Molecular Medicine, Morsani College of Medicine at the University of South Florida, Tampa, FL 33612, USA; (K.V.-A.); (I.O.R.); (C.A.); (K.H.)
| | - Ivannie Ortiz Rivera
- Department of Molecular Medicine, Morsani College of Medicine at the University of South Florida, Tampa, FL 33612, USA; (K.V.-A.); (I.O.R.); (C.A.); (K.H.)
| | - Ciara Alvarez
- Department of Molecular Medicine, Morsani College of Medicine at the University of South Florida, Tampa, FL 33612, USA; (K.V.-A.); (I.O.R.); (C.A.); (K.H.)
| | - Kazim Husain
- Department of Molecular Medicine, Morsani College of Medicine at the University of South Florida, Tampa, FL 33612, USA; (K.V.-A.); (I.O.R.); (C.A.); (K.H.)
| | - DeVon DeLoach
- Comparative Medicine at the University of South Florida, Tampa, FL 33612, USA;
| | - Gerald Krystal
- The Terry Fox Laboratory, BC Cancer, Vancouver, BC V5Z 1L3, Canada;
| | - Margaret L. Hibbs
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne 3004, Australia;
| | - Kun Jiang
- Anatomic Pathology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Tomar Ghansah
- Department of Molecular Medicine, Morsani College of Medicine at the University of South Florida, Tampa, FL 33612, USA; (K.V.-A.); (I.O.R.); (C.A.); (K.H.)
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Correspondence: ; Tel.: +1-813-974-1825
| |
Collapse
|
65
|
Zhang J, Sun H, Liu S, Huang W, Gu J, Zhao Z, Qin H, Luo L, Yang J, Fang Y, Ge J, Ni B, Wang H. Alteration of tumor-associated macrophage subtypes mediated by KRT6A in pancreatic ductal adenocarcinoma. Aging (Albany NY) 2020; 12:23217-23232. [PMID: 33221741 PMCID: PMC7746340 DOI: 10.18632/aging.104091] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 09/03/2020] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is severely affecting the health and lives of patients. Clarifying the composition and regulatory factors of tumor immune microenvironment (TIME) is helpful for the treatment of PDAC. We analyzed the unique TIMEs and gene expression patterns between PDAC and adjacent normal tissue (ANT) using Gene Expression Omnibus (GEO) to find new immunotherapy targets. The Cancer Genome Atlas (TCGA) datasets were used to elucidate the possible mechanism of which tumor-associated macrophages (TAMs) changed in PDAC. We found that the composition of TAMs subtypes, including M0, M1, and M2, was different between PDAC and ANT, which was validated in recently published single-cell RNA-seq data. Many immune cells interacted with each other to affect the TIME. There were many DEGs enriched in some pathways that could potentially change the immune cell composition. KRT6A was found to be a DEG between PDAC and ANT that overlapped with DEGs between the M0-high group and the M0-low group in TCGA datasets, and it might alter and regulate TAMs via a collection of genes including COL5A2, COL1A2, MIR3606, SPARC, and COL6A3. TAMs, which could be a target of immunotherapy, might be influenced by genes through KRT6A and indicate an undesirable prognosis in PDAC.
Collapse
Affiliation(s)
- Junfeng Zhang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| | - Hui Sun
- Department of Rheumatology, First Affiliated Hospital of Third Military Medical University, Chongqing 400038, P R China
| | - Songsong Liu
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Wenjie Huang
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, P R China
| | - Jianyou Gu
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, Guangdong Province, P R China
| | - Zhiping Zhao
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Huan Qin
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Liwen Luo
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Jiali Yang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, P R China
| | - Yongfei Fang
- Department of Rheumatology, First Affiliated Hospital of Third Military Medical University, Chongqing 400038, P R China
| | - Jiayun Ge
- Hepatopancreatobiliary Surgery Department, the Second Affiliated Hospital of Kunming Medical University, Kunming 650101, Yunnan Province, P R China
| | - Bing Ni
- Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing 400038, P R China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing 400038, P R China
- Key Laboratory of High Altitude Medicine, PLA, Chongqing 400038, P R China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 401120, P R China
| |
Collapse
|
66
|
Geng Y, Fan J, Chen L, Zhang C, Qu C, Qian L, Chen K, Meng Z, Chen Z, Wang P. A Notch-Dependent Inflammatory Feedback Circuit between Macrophages and Cancer Cells Regulates Pancreatic Cancer Metastasis. Cancer Res 2020; 81:64-76. [PMID: 33172931 DOI: 10.1158/0008-5472.can-20-0256] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 09/11/2020] [Accepted: 10/28/2020] [Indexed: 11/16/2022]
Abstract
Notch activation has been detected in pancreatic ductal adenocarcinoma (PDAC). However, its role in PDAC metastasis remains unknown. In this study, we identify a Notch-dependent feedback circuit between pancreatic cancer cells and macrophages, which contributes to PDAC metastasis. In this circuit, miR-124 regulated Notch signaling in cancer cells by directly targeting the Notch ligand Jagged 1. Autoamplified Notch signaling promoted the recruitment and activation of macrophages to a tumor-supporting M2-like phenotype via downstream IL8, CCL2, IL1α, and uPA paracrine signaling. In turn, activated macrophage-derived IL6 activated the oncogenic transcription factor STAT3 that directly repressed miR-124 genes via a conserved STAT3-binding site in their promoters, thereby promoting cancer cell epithelial-mesenchymal transition and invasion. Disrupting this circuit suppressed liver metastasis in mouse models. Thus, our study suggests that manipulation of this Notch-dependent circuit has a therapeutic potential for the treatment of PDAC metastasis. SIGNIFICANCE: This study provided potential therapeutic targets and robust preclinical evidence for PDAC treatment by interrupting feedback signaling between cancer cells and macrophages with targeted inhibitors.
Collapse
Affiliation(s)
- Yawen Geng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jie Fan
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Lianyu Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chenyue Zhang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chao Qu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling Qian
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kun Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiqiang Meng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
67
|
Yang J, Li Y, Sun Z, Zhan H. Macrophages in pancreatic cancer: An immunometabolic perspective. Cancer Lett 2020; 498:188-200. [PMID: 33122097 DOI: 10.1016/j.canlet.2020.10.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/30/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
As one of the most fatal gastrointestinal cancers, pancreatic cancer (PC) has a long-term survival rate that has shown limited improvement during recent decades and remains dismal. The poor prognosis is attributed to challenges in early detection, low opportunity for radical resection and resistance to chemotherapy and radiation. Macrophages are one of the most abundant infiltrating immune cells in PC stroma, and they can crosstalk with cancer cells, adipocytes and other stromal cells to modulate metabolism, inflammation and immune status, create an immunosuppressive tumor microenvironment (TME), and ultimately facilitate tumor initiation and progression. In this review, we summarize recent advances in our understanding of macrophage origin, distribution and polarization, as well as provide a thorough review of the role macrophages in PC carcinogenesis and development, as well as the underlying molecular mechanism. Additionally, we investigated macrophage targets in preclinical and clinical trials to evaluate their potential therapeutic value in PC.
Collapse
Affiliation(s)
- Jian Yang
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Yongzheng Li
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Zhaowei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, Shandong Province, 250012, China.
| |
Collapse
|
68
|
Han S, Zhen W, Guo T, Zou J, Li F. SETDB1 promotes glioblastoma growth via CSF-1-dependent macrophage recruitment by activating the AKT/mTOR signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:218. [PMID: 33059737 PMCID: PMC7560339 DOI: 10.1186/s13046-020-01730-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022]
Abstract
Background Glioblastoma is a common disease of the central nervous system (CNS), with high morbidity and mortality. In the infiltrate in the tumor microenvironment, tumor-associated macrophages (TAMs) are abundant, which are important factors in glioblastoma progression. However, the exact details of TAMs in glioblastoma progression have yet to be determined. Methods The clinical relevance of SET domain bifurcated 1 (SETDB1) was analyzed by immunohistochemistry, real-time PCR and Western blotting of glioblastoma tissues. SETDB1-induced cell proliferation, migration and invasion were investigated by CCK-8 assay, colony formation assay, wound healing and Transwell assay. The relationship between SETDB1 and colony stimulating factor 1 (CSF-1), as well as TAMs recruitment was examined by Western blotting, real-time PCR and syngeneic mouse model. Results Our findings showed that SETDB1 upregulated in glioblastoma and relative to poor progression. Gain and loss of function approaches showed the SETDB1 overexpression promotes cell proliferation, migration and invasion in glioblastoma cells. However, knockdown SETDB1 exerted opposite effects in vitro. Moreover, SETDB1 promotes AKT/mTOR-dependent CSF-1 induction and secretion, which leads to macrophage recruitment in the tumor, resulted in tumor growth. Conclusion Our research clarified that SETDB1 regulates of tumor microenvironment and hence presents a potential therapeutic target for treating glioblastoma.
Collapse
Affiliation(s)
- Shuai Han
- Department of Neurosurgery, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wei Zhen
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), No.33, Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning Province, PR China
| | - Tongqi Guo
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), No.33, Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning Province, PR China
| | - Jianjun Zou
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), No.33, Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning Province, PR China
| | - Fuyong Li
- Department of Neurosurgery, The People's Hospital of China Medical University (The People's Hospital of Liaoning Province), No.33, Wenyi Road, Shenhe District, Shenyang, 110016, Liaoning Province, PR China.
| |
Collapse
|
69
|
Bi Y, Shirure VS, Liu R, Cunningham C, Ding L, Meacham JM, Goedegebuure SP, George SC, Fields RC. Tumor-on-a-chip platform to interrogate the role of macrophages in tumor progression. Integr Biol (Camb) 2020; 12:221-232. [PMID: 32930334 DOI: 10.1093/intbio/zyaa017] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/20/2020] [Accepted: 08/15/2020] [Indexed: 02/06/2023]
Abstract
Tumor-infiltrating leukocytes, in particular macrophages, play an important role in tumor behavior and clinical outcome. The spectrum of macrophage subtypes ranges from antitumor 'M1'-type to protumor 'M2'-type macrophages. Tumor-associated macrophages (TAMs) typically display phenotypic features of both M1 and M2, and the population distribution is thought to be dynamic and evolves as the tumor progresses. However, our understanding of how TAMs impact the tumor microenvironment remains limited by the lack of appropriate 3D in vitro models that can capture cell-cell dynamics at high spatial and temporal resolution. Using our recently developed microphysiological 'tumor-on-a-chip' (TOC) device, we present here our findings on the impact of defined macrophage subsets on tumor behavior. The TOC device design contains three adjacent and connected chambers in which both the upper and lower chambers are loaded with tumor cells, whereas the central chamber contains a dynamic, perfused, living microvascular network. Introduction of human pancreatic or colorectal cancer cells together with M1-polarized macrophages significantly inhibited tumor growth and tumor-induced angiogenesis. Protein analysis and antibody-based neutralization studies confirmed that these effects were mediated through production of C-X-C motif chemokines (CXCL9), CXCL10 and CXCL11. By contrast, M2-macrophages mediated increased tumor cell migration into the vascularized chamber and did not inhibit tumor growth or angiogenesis. In fact, single-cell RNA sequencing showed that M2 macrophages further segregated endothelial cells into two distinct subsets, corresponding to static cells in vessels versus active cells involved in angiogenesis. The impact of M2 macrophages was mediated mostly by production of matrix metalloproteinase 7 and angiopoietin 2. In summary, our data demonstrate the utility of the TOC device to mechanistically probe biological questions in a 3D in vitro microenvironment.
Collapse
Affiliation(s)
- Ye Bi
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Ruiyang Liu
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA.,McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Cassandra Cunningham
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA.,McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - J Mark Meacham
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
70
|
Cruz AF, Rohban R, Esni F. Macrophages in the pancreas: Villains by circumstances, not necessarily by actions. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:807-824. [PMID: 32885589 PMCID: PMC7654401 DOI: 10.1002/iid3.345] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022]
Abstract
Introduction Mounting evidence suggest that macrophages play crucial roles in disease and tissue regeneration. However, despite much efforts during the past decade, our knowledge about the extent of macrophages' contribution to adult pancreatic regeneration after injury or during pancreatic disease progression is still limited. Nevertheless, it is generally accepted that some macrophage features that normally would contribute to healing and regeneration may be detrimental in pancreatic cancer. Altogether, the current literature contains conflicting reports on whether macrophages act as friends or foe in these conditions. Methods and Results In this review, we briefly review the origins of tissue resident and infiltrating macrophages and the importance of cellular crosstalking between macrophages and other resident cells in tissue regeneration. The primary objective of this review is to summarize our knowledge of the distinct roles of tissue resident and infiltrating macrophages, the impact of M1 and M2 macrophage phenotypes, and emerging evidence on macrophage crosstalking in pancreatic injury, regeneration, and disease. Conclusion Macrophages are involved with various stages of pancreatic cancer development, pancreatitis, and diabetes. Elucidating their role in these conditions will aid the development of targeted therapeutic treatments.
Collapse
Affiliation(s)
- Andrea F Cruz
- Division of Pediatric General and Thoracic Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Rokhsareh Rohban
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Farzad Esni
- Division of Pediatric General and Thoracic Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| |
Collapse
|
71
|
Huang BZ, Binder AM, Sugar CA, Chao CR, Setiawan VW, Zhang ZF. Methylation of immune-regulatory cytokine genes and pancreatic cancer outcomes. Epigenomics 2020; 12:1273-1285. [PMID: 32867538 DOI: 10.2217/epi-2019-0335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Given the immunosuppressive nature of pancreatic cancer, we investigated the relationship between epigenetic modification of immune-regulatory cytokine genes and pancreatic cancer outcomes. Materials & methods: We evaluated DNA methylation of 184 pancreatic tumor samples from The Cancer Genome Atlas for 111 CpG loci in seven cytokine genes: IL10, IL6, IL8, TGFβ1, TGFβ2, TGFβ3 and TNF. We used Cox regression to evaluate the associations between methylation and overall survival, disease-specific survival and disease progression (α = 0.05). Results: Poorer survival was associated with increased methylation in fifteen CpG probes in TGFβ1, TGFβ2, TGFβ3 and TNF. We also detected improved outcomes for three loci in IL10, IL8 and IL6. Conclusion: Epigenetic regulation of cytokine-related gene expression may be associated with pancreatic cancer outcomes.
Collapse
Affiliation(s)
- Brian Z Huang
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA.,Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA 91101, USA.,Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Alexandra M Binder
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA.,Department of Cancer Epidemiology, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Catherine A Sugar
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA.,Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Chun R Chao
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA 91101, USA
| | - Veronica Wendy Setiawan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA Fielding School of Public Health, Los Angeles, CA 90095, USA
| |
Collapse
|
72
|
Ren S, Zhang X, Hu Y, Wu J, Ju Y, Sun X, Liu Y, Shan B. Blocking the Notch signal transduction pathway promotes tumor growth in osteosarcoma by affecting polarization of TAM to M2 phenotype. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1057. [PMID: 33145276 PMCID: PMC7575992 DOI: 10.21037/atm-20-3881] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Osteosarcoma is a primary malignant tumor that seriously affects the health and life of patients. It is of great clinical significance to explore the molecular mechanism of osteosarcoma development and develop the corresponding therapeutic targets. Th1/Th2 cytokines in the normal human body are in a state of dynamic balance. When this balance is destroyed, it is related to many diseases such as a tumor, autoimmune disease, microbial infection, transplant rejection, among many others. Method The model of mouse tumor-associated macrophage (TAM) was induced by being co-cultured with inducer granulocyte-macrophage colony stimulating factor (GM-CSF) and osteosarcoma S180 cells. The Notch1 knockout mice were obtained by gene targeting technology. The distribution of M1- and M2-type TAMs in the tumor was visualized by immunofluorescence staining. And the western-blot testing was used to detect and quantified the protein level of Notch1 and Th1/Th2-type cytokines. Results In this study, the polarization of TAMs to the M2 phenotype occurred after coculture with osteosarcoma S180 cells and secretion level Th1/Th2-type cytokines changed. Also, the expression level of Notch1 reduced significantly. Further, the critical transcription factor Notch1 of the Notch signaling pathway was knocked out in mice. The tumor volume of Notch1 knockout mice was significantly more extensive than of the control mice. The results of microstructural observation on tumor showed that M2-type TAMs infiltrated into tumor increased with increased expression of Th2-type cytokines, but M1-type TAMs reduced with reduced expression of Th1-type cytokines. Conclusions According to our results, the Notch signal transduction pathway participates in tumor occurrence and growth with a negative role by maintaining Th1/Th2 balance.
Collapse
Affiliation(s)
- Shuguang Ren
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiangmei Zhang
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yueyang Hu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianhua Wu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingchao Ju
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xu Sun
- Hospital of Stomatology Hebei Medical University, Shijiazhuang, China
| | - Yunjiang Liu
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Baoen Shan
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
73
|
Nasrollahzadeh E, Razi S, Keshavarz-Fathi M, Mazzone M, Rezaei N. Pro-tumorigenic functions of macrophages at the primary, invasive and metastatic tumor site. Cancer Immunol Immunother 2020; 69:1673-1697. [PMID: 32500231 DOI: 10.1007/s00262-020-02616-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment (TME) not only facilitates cancer progression from the early formation to distant metastasis, but also it differs itself from time to time alongside the tumor evolution. Tumor-associated macrophages (TAMs), whether as pre-existing tissue-resident macrophages or recruited monocytes, are an inseparable part of this microenvironment. As their parents are broadly classified into a dichotomic, simplistic M1 and M2 subtypes, TAMs also exert paradoxical and diverse phenotypes as they are settled in different regions of TME and receive different microenvironmental signals. Briefly, M1 macrophages induce an inflammatory precancerous niche and flame the early oncogenic mutations, whereas their M2 counterparts are reprogrammed to release various growth factors and providing an immunosuppressive state in TME as long as abetting hypoxic cancer cells to set up a new vasculature. Further, they mediate stromal micro-invasion and co-migrate with invasive cancer cells to invade the vascular wall and neural sheath, while another subtype of TAMs prepares suitable niches much earlier than metastatic cells arrive at the target tissues. Accordingly, at the neoplastic transformation, during the benign-to-malignant transition and through the metastatic cascade, macrophages are involved in shaping the primary, micro-invasive and pre-metastatic TMEs. Whether their behavioral plasticity is derived from distinct genotypes or is fueled by microenvironmental cues, it could define these cells as remarkably interesting therapeutic targets.
Collapse
Affiliation(s)
- Elaheh Nasrollahzadeh
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer Biology, VIB, KU Leuven, Louvain, B3000, Belgium
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
74
|
Li F, Guo H, Wang Y, Liu B, Zhou H. Profiles of tumor-infiltrating immune cells and prognostic genes associated with the microenvironment of bladder cancer. Int Immunopharmacol 2020; 85:106641. [PMID: 32470882 DOI: 10.1016/j.intimp.2020.106641] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022]
Abstract
The immune microenvironment in bladder cancer (BC) and its significance still remain poorly understood. The present work aims to investigate tumor-infiltrating immune cells (TIICs) and prognostic genes associated with the tumor microenvironment (TME) of BC. The immune and stromal scores of BC samples from The Cancer Genome Atlas database were downloaded from the ESTIMATE website. Based on these scores, BC samples were assigned to the high and low score groups and 429 intersecting differentially expressed genes were identified. Functional enrichment analysis further revealed that these genes dramatically participated in the immune-related biological processes and signaling pathways. Two TME-related genes, angiotensin II receptor type 2 (AGTR2) and sclerostin domain containing 1 (SOSTDC1), were identified to establish an immune-related risk model using Cox regression analyses. Intriguingly, patients with high-risk scores had poor outcomes (p < 0.001). The areas under the curve for the risk model in predicting 3- and 5-year survival rates were 0.692 and 0.707, respectively. Kaplan-Meier survival analysis showed that the expression of AGTR2 and SOSTDC1 significantly correlated with the overall survival of BC patients. Additionally, 22 TIICs in the BC microenvironment were analyzed with the CIBERSORT algorithm. This study indicated that the effective components of TME affected the clinical outcomes of BC patients and might provide a basis for the development of new immunotherapies for BC patients.
Collapse
Affiliation(s)
- Faping Li
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Hui Guo
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, Jilin, China
| | - Bin Liu
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Honglan Zhou
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China.
| |
Collapse
|
75
|
Chandrakesan P, Panneerselvam J, May R, Weygant N, Qu D, Berry WR, Pitts K, Stanger BZ, Rao CV, Bronze MS, Houchen CW. DCLK1-Isoform2 Alternative Splice Variant Promotes Pancreatic Tumor Immunosuppressive M2-Macrophage Polarization. Mol Cancer Ther 2020; 19:1539-1549. [PMID: 32371580 DOI: 10.1158/1535-7163.mct-19-0776] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/20/2019] [Accepted: 04/21/2020] [Indexed: 12/24/2022]
Abstract
Tumor-associated M2-macrophages are one of the most abundant immunosuppressive cell types in the pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment (TME). However, the molecular mechanisms responsible for the generation of M2-macrophages are unclear. Here, we demonstrated that overexpression of DCLK1-isoform2 in AsPC1 and MIA PaCa2 cells resulted in the polarization of M1-macrophages toward an M2 phenotype via secreted chemokines/cytokines. These M2-macrophages enhanced parental PDAC cell migration, invasion, and self-renewal, and this was associated with increased expression of Snail and Slug. We observed distinct expression of Dclk-isoform2, marked infiltration of M2-macrophages, and a marginal increase of CD8+ T cells in 20-week-old KPCY mice pancreas compared with 5 weeks old. Utilizing an autochthonous mouse model of pancreatic adenocarcinoma, we observed distinct immunoreactive Dclk1 and arginase1 in tissues where CD8+ T-cell infiltration was low and observed a paucity of DCLK1 and arginase1 staining where CD8+ T-cell infiltration was high. Finally, we found that DCLK1-isoform2 tumor-educated M2-macrophages inhibit CD8+ T-cell proliferation and granzyme-B activation. Inhibition of DCLK1 in an organoid coculture system enhanced CD8+ T-cell activation and associated organoid death. We conclude that DCLK1-isoform2 is a novel initiator of alternate macrophage activation that contributes to the immunosuppression observed in the PDAC TME. These data suggest that tumor DCLK1-isoform2 may be an attractive target for PDAC therapy, either alone or in conjunction with immunotherapeutic strategies.
Collapse
Affiliation(s)
- Parthasarathy Chandrakesan
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma. .,OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Janani Panneerselvam
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Randal May
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Nathaniel Weygant
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Dongfeng Qu
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - William R Berry
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kamille Pitts
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Ben Z Stanger
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chinthalapally V Rao
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| | - Michael S Bronze
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Courtney W Houchen
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma. .,OU Cancer Institute, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.,Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma
| |
Collapse
|
76
|
Salaymeh Y, Farago M, Sebban S, Shalom B, Pikarsky E, Katzav S. Vav1 and mutant K-Ras synergize in the early development of pancreatic ductal adenocarcinoma in mice. Life Sci Alliance 2020; 3:e202000661. [PMID: 32277014 PMCID: PMC7156281 DOI: 10.26508/lsa.202000661] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023] Open
Abstract
To explore the contribution of Vav1, a hematopoietic signal transducer, to pancreatic ductal adenocarcinoma (PDAC) development, we generated transgenic mouse lines expressing, Vav1, K-RasG12D, or both K-RasG12D and Vav1 in pancreatic acinar cells. Co-expression of Vav1 and K-RasG12D synergistically enhanced acinar-to-ductal metaplasia (ADM) formation, far exceeding the number of lesions developed in K-RasG12D mice. Mice expressing only Vav1 did not develop ADM. Moreover, the incidence of PDAC in K-RasG12D/Vav1 was significantly higher than in K-RasG12D mice. Discontinuing Vav1 expression in K-RasG12D/Vav1 mice elicited a marked regression of malignant lesions in the pancreas, demonstrating Vav1 is required for generation and maintenance of ADM. Rac1-GTP levels in the K-RasG12D/Vav1 mice pancreas clearly demonstrated an increase in Rac1 activity. Treatment of K-RasG12D and K-RasG12D/Vav1 mice with azathioprine, an immune-suppressor drug which inhibits Vav1's activity as a GDP/GTP exchange factor, dramatically reduced the number of malignant lesions. These results suggest that Vav1 plays a role in the development of PDAC when co-expressed with K-RasG12D via its activity as a GEF for Rac1GTPase.
Collapse
Affiliation(s)
- Yaser Salaymeh
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Marganit Farago
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Shulamit Sebban
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Batel Shalom
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Eli Pikarsky
- The Lautenberg Center for Immunology and Cancer Research and Department of Pathology, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Shulamit Katzav
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
77
|
Wang B, Zhang W, Zhou X, Liu M, Hou X, Cheng Z, Chen D. Development of dual-targeted nano-dandelion based on an oligomeric hyaluronic acid polymer targeting tumor-associated macrophages for combination therapy of non-small cell lung cancer. Drug Deliv 2020; 26:1265-1279. [PMID: 31777307 PMCID: PMC6896416 DOI: 10.1080/10717544.2019.1693707] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In this study, the novel carrier materials were screened to structure targeting nano-micelles (named ‘nano-dandelion’) for synchronous delivery of curcumin (Cur) and baicalin (Bai), which could effectively overcome the tumor resistance. Mannose (Man) was found to bind better to CD206 receptors on the surface of tumor-associated macrophages (TAMs), thereby increasing the number of nano-dandelion engulfed by TAMs. Furthermore, oligomeric hyaluronic acid (oHA) was able to target CD44 receptors, resulting in recruitment of a higher number of nano-dandelion to locate and engulf tumor cells. The disulfide bond (S–S) in 3,3′-dithiodipropionic acid (DA) could be broken by the high concentration of glutathione (GSH) in the tumor microenvironment (TME). Based on this, we selected DA to connect hydrophobic fragments (quercetin, Que) and oHA. A reduction-sensitive amphiphilic carrier material, quercetin–dithiodipropionic acid–oligomeric hyaluronic acid–mannose–ferulic acid (Que–S–S–oHA–Man–FA; QHMF) was fabricated and synthesized by 1H NMR. Next, QHMF self-assembled into nano-dandelion, i.e. encapsulated Cur and Bai in water. Critical experimental conditions in the preparation process of nano-dandelion that could affect its final properties were explored. Nano-dandelion with a small particle size (121.0 ± 15 nm) and good normal distribution (PI = 0.129) could easily enter tumor tissue through vascular barrier. In addition, nano-dandelion with a suitable surface potential (–20.33 ± 4.02 mV) could remain stable for a long duration. Furthermore, good cellular penetration and tumor cytotoxicity of nano-dandelion were demonstrated through in vitro cellular studies. Finally, effective antitumor activity and reduced side effects were confirmed through in vivo antitumor experiments in A549 tumor-bearing nude mice.
Collapse
Affiliation(s)
- Bingjie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| | - Wei Zhang
- Department of Radiotherapy, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, PR China
| | - Xiudi Zhou
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China.,Department of Pharmacy, Binzhou People's Hospital, Binzhou, PR China
| | - Mengna Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| | - Xiaoya Hou
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| | - Ziting Cheng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| | - Daquan Chen
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| |
Collapse
|
78
|
Hajiasgharzadeh K, Somi MH, Sadigh-Eteghad S, Mokhtarzadeh A, Shanehbandi D, Mansoori B, Mohammadi A, Doustvandi MA, Baradaran B. The dual role of alpha7 nicotinic acetylcholine receptor in inflammation-associated gastrointestinal cancers. Heliyon 2020; 6:e03611. [PMID: 32215331 PMCID: PMC7090353 DOI: 10.1016/j.heliyon.2020.e03611] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 12/10/2019] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
Alpha7 nicotinic acetylcholine receptor (α7nAChR) is one of the main subtypes of nAChRs that modulates various cancer-related properties including proliferative, anti-apoptotic, pro-angiogenic and pro-metastatic activities in most of the cancers. It also plays a crucial role in inflammation control through the cholinergic anti-inflammatory pathway in numerous pathophysiological contexts. Such diverse physiological and pathological functions that initiate from this receptor may have significant impacts in determining the outcome of different cancers. Various tissues of gastrointestinal (GI) cancers such as gastric, colorectal, pancreatic and liver cancers have shown the up-regulated expression of α7nAChR as compared to normal adjacent tissues. According to the well-established connection between inflammation and tumorigenesis in the digestive system, there are mounting studies demonstrated either stimulatory or inhibitory effects of α7nAChR signaling in the development of GI cancers. To date, the precise underlying mechanisms related to this receptor in patients with GI cancers have not been fully elucidated. Regarding the paradoxical modulatory effects of this receptor in carcinogenesis, in this review, we aim to summarize the accumulated evidence about the involvement of α7nAChR in inflammation-associated GI cancers. It seems that the complex influences of α7nAChR may be a promising target in designing novel strategies in the treatment of such pathologic conditions.
Collapse
Affiliation(s)
| | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Sadigh-Eteghad
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
79
|
Bazzichetto C, Conciatori F, Luchini C, Simionato F, Santoro R, Vaccaro V, Corbo V, Falcone I, Ferretti G, Cognetti F, Melisi D, Scarpa A, Ciuffreda L, Milella M. From Genetic Alterations to Tumor Microenvironment: The Ariadne's String in Pancreatic Cancer. Cells 2020; 9:309. [PMID: 32012917 PMCID: PMC7072496 DOI: 10.3390/cells9020309] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/18/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
The threatening notoriety of pancreatic cancer mainly arises from its negligible early diagnosis, highly aggressive progression, failure of conventional therapeutic options and consequent very poor prognosis. The most important driver genes of pancreatic cancer are the oncogene KRAS and the tumor suppressors TP53, CDKN2A, and SMAD4. Although the presence of few drivers, several signaling pathways are involved in the oncogenesis of this cancer type, some of them with promising targets for precision oncology. Pancreatic cancer is recognized as one of immunosuppressive phenotype cancer: it is characterized by a fibrotic-desmoplastic stroma, in which there is an intensive cross-talk between several cellular (e.g., fibroblasts, myeloid cells, lymphocytes, endothelial, and myeloid cells) and acellular (collagen, fibronectin, and soluble factors) components. In this review; we aim to describe the current knowledge of the genetic/biological landscape of pancreatic cancer and the composition of its tumor microenvironment; in order to better direct in the intrinsic labyrinth of this complex tumor type. Indeed; disentangling the genetic and molecular characteristics of cancer cells and the environment in which they evolve may represent the crucial step towards more effective therapeutic strategies.
Collapse
Affiliation(s)
- Chiara Bazzichetto
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Fabiana Conciatori
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Francesca Simionato
- Division of Oncology, University of Verona, 37126 Verona, Italy; (F.S.); (M.M.)
| | - Raffaela Santoro
- Medicine-Digestive Molecular Clinical Oncology Research Unit, University of Verona, 37126 Verona, Italy; (R.S.); (D.M.)
| | - Vanja Vaccaro
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Vincenzo Corbo
- ARC-Net Research Centre, University and Hospital Trust of Verona, 37126 Verona, Italy; (V.C.); (A.S.)
| | - Italia Falcone
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Gianluigi Ferretti
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Francesco Cognetti
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (C.B.); (V.V.); (I.F.); (G.F.); (F.C.)
| | - Davide Melisi
- Medicine-Digestive Molecular Clinical Oncology Research Unit, University of Verona, 37126 Verona, Italy; (R.S.); (D.M.)
| | - Aldo Scarpa
- ARC-Net Research Centre, University and Hospital Trust of Verona, 37126 Verona, Italy; (V.C.); (A.S.)
| | - Ludovica Ciuffreda
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Michele Milella
- Division of Oncology, University of Verona, 37126 Verona, Italy; (F.S.); (M.M.)
| |
Collapse
|
80
|
Xu Z, Zhang Y, Xu M, Zheng X, Lin M, Pan J, Ye C, Deng Y, Jiang C, Lin Y, Lu X, Chi P. Demethylation and Overexpression of CSF2 are Involved in Immune Response, Chemotherapy Resistance, and Poor Prognosis in Colorectal Cancer. Onco Targets Ther 2019; 12:11255-11269. [PMID: 31908491 PMCID: PMC6927609 DOI: 10.2147/ott.s216829] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/13/2019] [Indexed: 12/21/2022] Open
Abstract
Purpose This study aimed to evaluate the role of colony-stimulating factor 2 (CSF2) in chemotherapy resistance, prognosis, and immune response and to identify its possible mechanisms underlying drug resistance. Methods Drug-resistant cell lines were obtained by successively increasing drug concentration. RNA-Seq was performed to screen hub genes. CSF2 expression was analyzed via immunohistochemistry. Moreover, The Cancer Genome Atlas (TCGA), Tumor Immune Estimation Resource (TIMER) dataset, and R2 platform were used to explore the correlations among CSF2 expression, prognosis, and immune response. Results RNA-Seq indicated that microRNAs in cancer, P53 signaling pathway, and cell cycle were associated with FOLFOX chemotherapy resistance. Protein-protein interaction (PPI), molecular complex detection (MOCDE), and qRT-PCR analysis verified CSF2 as the hub gene in chemotherapy resistance. Moreover, CSF2 expression was lower in the normal tissue than in the cancerous tissue (P<0.05). Higher expression of CSF2 was associated with poor OS and DFS in colon cancer patients (P<0.05). We further found similar results in the Oncomine database and R2 platform (P<0.05). A higher expression of CSF2 in the CRC tissue may be caused by demethylation, which was verified using the TCGA datasets. Moreover, GSEA demonstrated that CSF2 was associated with immune response, which was consistent with results reported using TIMER datasets. Conclusion CSF2 is a novel biomarker and a prognostic factor for the survival of CRC patients affecting the immune response, and an overexpression of CSF2 in CRC patients may be caused by DNA demethylation.
Collapse
Affiliation(s)
- Zongbin Xu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Yiyi Zhang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Meifang Xu
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Xin Zheng
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Mengxin Lin
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Jie Pan
- Department of Emergency Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Chengwei Ye
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Yu Deng
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Caiyun Jiang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Yu Lin
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Xingrong Lu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Pan Chi
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| |
Collapse
|
81
|
Benzing C, Lam H, Tsang CM, Rimmer A, Arroyo-Berdugo Y, Calle Y, Wells CM. TIMP-2 secreted by monocyte-like cells is a potent suppressor of invadopodia formation in pancreatic cancer cells. BMC Cancer 2019; 19:1214. [PMID: 31836008 PMCID: PMC6911299 DOI: 10.1186/s12885-019-6429-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/03/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Monocytes are a major component of the tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC). However, the complex interactions between tumor cells and monocytes and their role in tumor invasion have not been fully established. METHODS To specifically test the impact of interaction on invasive potential two PDAC cell lines PaTu8902 and CFPAC-1 were selected on their ability to form invasive adhesions, otherwise known as invadopodia and invade in a spheroid invasion assay. RESULTS Interestingly when the PDAC cells were co-cultured with undifferentiated THP1 monocyte-like cells invadopodia formation was significantly suppressed. Moreover, conditioned media of THP1 cells (CM) was also able to suppress invadopodia formation. Further investigation revealed that both tissue inhibitor of metalloproteinase (TIMP) 1 and 2 were present in the CM. However, suppression of invadopodia formation was found that was specific to TIMP2 activity. CONCLUSIONS Our findings indicate that TIMP2 levels in the tumour microenvironment may have prognostic value in patients with PDAC. Furthermore, activation of TIMP2 expressing monocytes in the primary tumour could present a potential therapeutic opportunity to suppress cell invasion in PDAC.
Collapse
Affiliation(s)
- Christian Benzing
- School of Cancer and Pharmaceutical Sciences, New Hunts House, Kings College London, London, SE1 1UL, UK.,Department of Surgery, Campus Charité-Mitte and Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hoyin Lam
- School of Cancer and Pharmaceutical Sciences, New Hunts House, Kings College London, London, SE1 1UL, UK
| | - Chi Man Tsang
- Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hongkong, Hong Kong SAR
| | - Alexander Rimmer
- School of Cancer and Pharmaceutical Sciences, New Hunts House, Kings College London, London, SE1 1UL, UK
| | | | - Yolanda Calle
- Department of Life Sciences, University of Roehampton, London, UK
| | - Claire M Wells
- School of Cancer and Pharmaceutical Sciences, New Hunts House, Kings College London, London, SE1 1UL, UK.
| |
Collapse
|
82
|
Shi J, Huo R, Li N, Li H, Zhai T, Li H, Shen B, Ye J, Fu R, Di W. CYR61, a potential biomarker of tumor inflammatory response in epithelial ovarian cancer microenvironment of tumor progress. BMC Cancer 2019; 19:1140. [PMID: 31766991 PMCID: PMC6878653 DOI: 10.1186/s12885-019-6321-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/31/2019] [Indexed: 01/14/2023] Open
Abstract
Background Recent studies have found that inflammatory response is involved in the pathogenesis of ovarian cancer. Advanced ovarian cancer is often presented with ascites that is rich in cytokines, inflammatory factors or cancer cells. Therefore, it is important to study the microenvironment of ascites in order to further clarify the occurrence and progression of ovarian cancer. As a pro-inflammatory factor, the Cyr61 expression patterns are inconsistent in human tumors. Although it has been reported that Cyr61 is related to the progression of ovarian cancer, its specific mechanism is not yet clear. This study sought to evaluate the Cyr61 levels of ascites, serum and different tissues of ovarian cancer to explore the potential association of Cyr61with the tumor-associated inflammatory microenvironment of EOC. Methods Tumor specimens were procured from patients with ovarian serous cystadenocarcinoma and ovarian serous cystadenoma. Cyr61 and IL-6 levels of serum or ascites were determined by ELISA (Enzyme-Linked ImmunoSorbent Assay), while Cyr61 expressions of different ovarian tumor tissues were evaluated by IHC (Immunohistochemistry). Then the correlation of Cyr61 level in ascites with clinicopathologic features was analyzed. And other laboratory data were obtained from medical records. Results Both in ascites and serum, significantly higher Cyr61 levels were found in ovarian serous cystadenocarcinoma. In malignant ascites, higher Cyr61 level of ovarian serous cystadenocarcinoma was more closely associated with FIGO stage, initial tumor size > 10 cm and the residual tumor size. And the increased IL-6 level was linearly related to Cyr61 level. Moreover, the serum levels of Cyr61, IL-6 and CRP in advanced stage of ovarian cancer were much higher than those in early stage. Lastly, the IHC data demonstrate that Cyr61 expression of ovarian serous adenocarcinoma was higher than that of ovarian serous cystadenoma, but it was lower than the paired metastatic lesions. Conclusions As a pro-inflammatory factor, increased ascites Cyr61 level is associated with FIGO stage, initial tumor size > 10 cm and the residual tumor size. Moreover, serum Cyr61 may be used as a potential marker for EOC inflammatory response. Finally, Cyr61 may be involved in the process of tumor metastasis and progression by producing IL-6 and CRP in the EOC inflammatory microenvironment.
Collapse
Affiliation(s)
- Jun Shi
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, People's Republic of China
| | - Rongfen Huo
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Ningli Li
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Haichuan Li
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Tianhang Zhai
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Huidan Li
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Baihua Shen
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Jing Ye
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, People's Republic of China
| | - Ruojin Fu
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, People's Republic of China
| | - Wen Di
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China. .,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
83
|
Gao Y, Fang P, Li WJ, Zhang J, Wang GP, Jiang DF, Chen FP. LncRNA NEAT1 sponges miR-214 to regulate M2 macrophage polarization by regulation of B7-H3 in multiple myeloma. Mol Immunol 2019; 117:20-28. [PMID: 31731055 DOI: 10.1016/j.molimm.2019.10.026] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND LncRNA NEAT1 was associated with the tumorigenesis of multiple myeloma (MM). However, the mechanisms of M2 macrophage polarization involved with NEAT1 in MM are still unknown. METHODS Bone marrow samples, multiple myeloma cells RPMI 8226 and monocyte cell line THP-1 were used in this study. The expression of NEAT1 and miR-214 was modified by transfection with the shNEAT1 or miR-214 inhibitor. The expression of NEAT1, miR-214 and B7-H3 in MM patient tissues and cells was analyzed by RT-qPCR. ELISA assay was used to determine the release of B7-H3 in the supernatant of cell culture. The patient survival curve was analyzed using Kaplan-Meier method. The macrophage polarization markers were examined by RT-qPCR and western blotting. The interaction between NEAT1, miR-214 and B7-H3 was analyzed by Dual-Luciferase reporter and RIP assays. AG490 was used to block the JAK2/STAT3 signaling. Co-culture of THP-1 and RPMI 8226 cells was used for macrophage polarization. RESULTS NEAT1 and B7-H3 were up-regulated, but miR-214 was obviously down-regulated in MM patients. B7-H3, NEAT1 and miR-214 were associated with overall survival time of MM patients. NEAT1 silencing induced miR-214 and inhibited the expression and release of B7-H3 and then suppressed M2 macrophage polarization via inhibiting the JAK2/STAT3 signaling. NEAT1 directly targeted miR-214, and miR-214 directly bound to B7-H3. MiR-214 inhibitor reversed the down-regulation and release of B7-H3 and M2 macrophage polarization caused by shNEAT1. The specific JAK2/STAT3 signaling inhibitor AG490 abrogated M2 macrophage polarization. CONCLUSION NEAT1 promoted M2 macrophage polarization by sponging miR-214 and then regulating B7-H3, thus accelerating MM progression via the JAK2/STAT3 signaling pathway. Our study revealed novel mechanisms of M2 macrophage polarization and provided new potential clinical therapeutic targets for MM.
Collapse
Affiliation(s)
- Yin Gao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Peng Fang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Wen-Jin Li
- Department of Hematology, JiangXi PingXiang People's Hospital, Pingxiang 337000, Jiangxi Province, P.R. China
| | - Jian Zhang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410003, Hunan Province, P.R. China
| | - Guang-Ping Wang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Duan-Feng Jiang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410003, Hunan Province, P.R. China
| | - Fang-Ping Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China; Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410003, Hunan Province, P.R. China.
| |
Collapse
|
84
|
Sun D, Luo T, Dong P, Zhang N, Chen J, Zhang S, Dong L, Janssen HLA, Zhang S. M2-polarized tumor-associated macrophages promote epithelial-mesenchymal transition via activation of the AKT3/PRAS40 signaling pathway in intrahepatic cholangiocarcinoma. J Cell Biochem 2019; 121:2828-2838. [PMID: 31692069 DOI: 10.1002/jcb.29514] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022]
Abstract
Tumor-associated macrophages (TAMs) have been considered as a major component of the tumor microenvironment. However, the crosstalk between M2-polarized tumor-associated macrophages (M2-TAMs) and intrahepatic cholangiocarcinoma (ICC) remains undetermined. In the present study, we aimed to clarify the role of M2-TAMs in ICC and the underlying mechanism. The in vitro assay demonstrated M2-TAMs promoted epithelial-mesenchymal transition (EMT) of ICC cells, resulting in enhanced cell invasion and metastasis ability. Moreover, M2-TAMs modulated the microenvironment of ICC by increasing the secretion of cytokines (GM-CSF, tumor necrosis factor-α [TNF-α], ICAM-1, interleukin-6 [IL-6], etc) and chemokines (CCL1, CCL3, etc). In addition, p-AKT (Ser473) and p-PRAS40 (Thr246) were upregulated in ICC cells when cocultured with M2-TAMs or treated with M2-TAMs secreted core cytokines (GM-CSF, TNF-α, ICAM-1, and IL-6). Consistently, AKT3 silencing (but not AKT1 silencing and AKT2 silencing) markedly inhibited phosphorylation of AKT and PRAS40 of ICC cells and inhibited the EMT process when cocultured with M2-TAMs. Taken together, the current data indicated that M2-TAMs promoted ICC cells EMT, partially through increasing secretion of cytokines and chemokines, thus modulating the microenvironment and activating the AKT3/PRAS40 signaling pathway.
Collapse
Affiliation(s)
- Dalong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China.,Department of Gastroenterology and Hepatology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Tiancheng Luo
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China
| | - Pingping Dong
- Department of Surgery, Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ningping Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China
| | - Jing Chen
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuncai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Institute of Liver Disease, Shanghai, China
| | - Harry L A Janssen
- Toronto Center for Liver Disease, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
85
|
Ponomarev AV, Shubina IZ. Insights Into Mechanisms of Tumor and Immune System Interaction: Association With Wound Healing. Front Oncol 2019; 9:1115. [PMID: 31709183 PMCID: PMC6823879 DOI: 10.3389/fonc.2019.01115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022] Open
Abstract
A large number of studies have presented a great deal of information about tumor and immune system interaction. Nevertheless, the problem of tumor evasion from the immune reaction is still difficult to resolve. Understanding the ways in which immunosuppressive tumor microenvironment develops and maintains its potential is of utmost importance to ensure the best use of the suppressed immune functions. The study presents a review covering the data on tumor-associated antigens, mechanisms of tumor evasion from the immune reactions, and search for common immunosuppressive processes of tumor growth and normal wound healing. The study discusses the important role of monocytes/macrophages in the regulation of immune system reactions. We suggest that the simultaneous actions of growth factors and pro-inflammatory cytokines may result in the suppression of the immune system. The study describes intracellular signaling molecules that take part in the regulation of the myeloid cell functions. If the hypothesis is proved correct, the indicated interaction of cytokines could be regarded as a prospective target for antitumor therapy.
Collapse
Affiliation(s)
| | - Irina Zh Shubina
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| |
Collapse
|
86
|
Brunetti O, Gnoni A, Licchetta A, Longo V, Calabrese A, Argentiero A, Delcuratolo S, Solimando AG, Casadei-Gardini A, Silvestris N. Predictive and Prognostic Factors in HCC Patients Treated with Sorafenib. ACTA ACUST UNITED AC 2019; 55:medicina55100707. [PMID: 31640191 PMCID: PMC6843290 DOI: 10.3390/medicina55100707] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022]
Abstract
Sorafenib is an oral kinase inhibitor that enhances survival in patients affected by advanced hepatocellular carcinoma (HCC). According to the results of two registrative trials, this drug represents a gold quality standard in the first line treatment of advanced HCC. Recently, lenvatinib showed similar results in terms of survival in a non-inferiority randomized trial study considering the same subset of patients. Unlike other targeted therapies, predictive and prognostic markers in HCC patients treated with sorafenib are lacking. Their identification could help clinicians in the daily management of these patients, mostly in light of the new therapeutic options available in the first.
Collapse
Affiliation(s)
- Oronzo Brunetti
- Medical Oncology Unit, National Cancer Research Centre, IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy.
| | - Antonio Gnoni
- Medical Oncology Unit, "S. Cuore di Gesù" Hospital, 73014 Gallipoli, Italy.
| | | | - Vito Longo
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy.
| | - Angela Calabrese
- Radiology Unit, National Cancer Research Centre, IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy.
| | - Antonella Argentiero
- Medical Oncology Unit, National Cancer Research Centre, IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy.
| | - Sabina Delcuratolo
- Scientific Direction, National Cancer Research Centre, IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy.
| | - Antonio Giovanni Solimando
- Medical Oncology Unit, National Cancer Research Centre, IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy.
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine "G. Baccelli", University of Bari Medical School, 70124 Bari, Italy.
| | - Andrea Casadei-Gardini
- Department of Medical Oncology, IstitutoScientifico Romagnolo per Lo Studio e Cura Dei Tumori (IRST) IRCCS, 47014 Meldola, Italy.
- Department of Oncology and Haematology, University Hospital of Modena, 41125 Modena, Italy.
| | - Nicola Silvestris
- Medical Oncology Unit, National Cancer Research Centre, IRCCS Istituto Tumori "Giovanni Paolo II", 70124 Bari, Italy.
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", 70124 Bari, Italy.
| |
Collapse
|
87
|
Argentiero A, Calabrese A, Solimando AG, Notaristefano A, Panarelli MM, Brunetti O. Bone metastasis as primary presentation of pancreatic ductal adenocarcinoma: A case report and literature review. Clin Case Rep 2019; 7:1972-1976. [PMID: 31624620 PMCID: PMC6787833 DOI: 10.1002/ccr3.2412] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/27/2019] [Accepted: 08/13/2019] [Indexed: 12/19/2022] Open
Abstract
PDAC bone metastases represent a clinical challenge characterized by multifaceted biological entity.
Collapse
Affiliation(s)
- Antonella Argentiero
- Medical Oncology UnitNational Cancer Research CentreIstituto Tumori "Giovanni Paolo II"BariItaly
| | - Angela Calabrese
- Radiology UnitNational Cancer Research CentreIstituto Tumori "Giovanni Paolo II"BariItaly
| | - Antonio Giovanni Solimando
- Medical Oncology UnitNational Cancer Research CentreIstituto Tumori "Giovanni Paolo II"BariItaly
- Department of Biomedical Sciences and Human OncologySection of Internal Medicine “G. Baccelli”University of Bari Medical SchoolBariItaly
| | | | | | - Oronzo Brunetti
- Medical Oncology UnitNational Cancer Research CentreIstituto Tumori "Giovanni Paolo II"BariItaly
| |
Collapse
|
88
|
Postoperative Monocyte Count Change Is a Better Predictor of Survival Than Preoperative Monocyte Count in Esophageal Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2702719. [PMID: 31485440 PMCID: PMC6710746 DOI: 10.1155/2019/2702719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/14/2019] [Indexed: 02/05/2023]
Abstract
In esophageal squamous cell carcinoma, an elevated preoperative absolute monocyte count (Pre-AMC) is reported to be a predictor of survival, but the clinical application of postoperative absolute monocyte count change (AMCc) remains unknown. The present study was designed to investigate the prognostic value of AMCc in ESCC. 686 patients of ESCC after radical surgery without preoperative adjuvant therapy were enrolled. The Pre-AMC and AMCc were recorded within one week before the operation and one week after surgery. We considered the median of Pre-AMC as the optimal cut-off value to evaluate the relationship between Pre-AMC and patient survival. AMCc was defined as AMCc increased (higher than Pre-AMC) and AMCc decreased (lower than Pre-AMC). Demographic and clinical characteristics, disease-free survival (DFS), and overall survival (OS) were statistically analyzed. Multivariate analysis revealed that AMCc was a better independent prognostic factor for both OS (P = 0.002, HR = 0.614, 95% CI 0.450-0.837) and DFS (P = 0.023, HR = 0.656, 95% CI 0.456-0.943) than Pre-AMC which was only an independent prognostic factor for OS (P = 0.033, HR = 2.031, 95% CI 1.058-3.898). AMCc could be a better prognostic factor than Pre-AMC in patients with ESCC. AMCc decrease predicts worse OS and DFS in ESCC undergoing curative resection.
Collapse
|
89
|
The Importance of a Conjoint Analysis of Tumor-Associated Macrophages and Immune Checkpoints in Pancreatic Cancer. Pancreas 2019; 48:904-912. [PMID: 31268976 DOI: 10.1097/mpa.0000000000001364] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Tumor-associated macrophages are dominant players in establishing the inmmunosuppressive microenvironment in pancreatic ductal adenocarcinoma (PDAC). Immune checkpoint inhibitor monotherapy has achieved limited clinical effectiveness. To date, the interaction of macrophages and checkpoint regulators and their correlation with clinicopathologic characteristics in PDAC have been largely unavailable. METHODS Macrophages and immune checkpoint expression were assessed by immunohistochemistry from 80 PDAC samples. Clinicopathologic features and the prognostic value of each marker were evaluated. In vitro changes in the expression of immune markers in cocultured macrophages and PDAC cells were detected by Western blot and immunosorbance assays. RESULTS The macrophages marker CD163 and the checkpoint marker programmed death-ligand 1 (PD-L1) remained as the independent prognostic factors for overall survival (hazard ratio, 2.543; P = 0.017 and hazard ratio, 2.389; P = 0.021). Furthermore, integrated analysis of CD163 and PD-L1 served as more optimal indicators of survival (P = 0.000). In vitro coculture of macrophages and PDAC cells significantly increased the expression of CD163 and PD-L1, compared with monocultured counterpart (P < 0.05). CONCLUSIONS Combined analysis of CD163 and PD-L1 was enhanced indicators of survival in PDAC patients. The interaction of macrophages and immune checkpoints implied the value of the combination therapy.
Collapse
|
90
|
Argentiero A, De Summa S, Di Fonte R, Iacobazzi RM, Porcelli L, Da Vià M, Brunetti O, Azzariti A, Silvestris N, Solimando AG. Gene Expression Comparison between the Lymph Node-Positive and -Negative Reveals a Peculiar Immune Microenvironment Signature and a Theranostic Role for WNT Targeting in Pancreatic Ductal Adenocarcinoma: A Pilot Study. Cancers (Basel) 2019; 11:cancers11070942. [PMID: 31277479 PMCID: PMC6678707 DOI: 10.3390/cancers11070942] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/25/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022] Open
Abstract
Over the past several years there has been much debate with regards to the prognostic and clinical significance of pancreatic ductal adenocarcinoma (PDAC) with lymph nodes metastasis. The PDAC gene expression knowledge and the biologic alterations underlying the lymph node involvement convey a clinical implication in dealing with the theranostic window. To this end, we provide an original bioinformatic dissection of the gene expression differences of PDAC according to the nodal involvement from a large public available dataset. Comprehensive transcriptomic analysis from 143 RNA-seq patient's derived samples indicated that WNT increased activation and a peculiar immune microenvironment identify subjects with nodal involvement. In frame of this thinking, we validated the WNT pathway role in increasing the likelihood of lymphatic dissemination in vitro. Moreover, we demonstrated for the first time in a PDAC model the potential therapeutic window that XAV-939-a specific WNT pathway inhibitor-has in re-educating a tumor-permissive immune system. Finally, we outline the potential implication on bystander molecular drivers exerted by WNT molecular inhibition, providing a picture of the proteomic oncogenic landscape changes elicited by XAV-939 on PDAC cells and their clinical implication. Our findings hold the promise to identify novel immune-based therapeutic strategies targeting WNT to enhance PDAC cytotoxicity and restore anti-PDAC immunity in node-positive disease.
Collapse
Affiliation(s)
- Antonella Argentiero
- Medical Oncology Unit, IRCCS Cancer Institute "Giovanni Paolo II" of Bari, 70124 Bari, Italy
| | - Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Cancer Institute "Giovanni Paolo II", 70124 Bari, Italy
| | - Roberta Di Fonte
- Experimental Pharmacology Laboratory, IRCCS Cancer Institute "Giovanni Paolo II", 70124 Bari, Italy
| | - Rosa Maria Iacobazzi
- Experimental Pharmacology Laboratory, IRCCS Cancer Institute "Giovanni Paolo II", 70124 Bari, Italy
| | - Letizia Porcelli
- Experimental Pharmacology Laboratory, IRCCS Cancer Institute "Giovanni Paolo II", 70124 Bari, Italy
| | - Matteo Da Vià
- Department of Internal Medicine II, Interdisciplinary Center for Clinical Research Laboratory, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Oronzo Brunetti
- Medical Oncology Unit, The Hospital Mons. R. Dimiccoli, 76121 Barletta (Bat), Italy
| | - Amalia Azzariti
- Experimental Pharmacology Laboratory, IRCCS Cancer Institute "Giovanni Paolo II", 70124 Bari, Italy
| | - Nicola Silvestris
- Scientific Direction, IRCCS Cancer Institute "Giovanni Paolo II", 70124 Bari, Italy
| | - Antonio Giovanni Solimando
- Department of Internal Medicine II, Interdisciplinary Center for Clinical Research Laboratory, University Hospital of Würzburg, 97080 Würzburg, Germany.
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine 'G. Baccelli', University of Bari Medical School, 70124 Bari, Italy.
| |
Collapse
|
91
|
Lehmann JS, Rughwani P, Kolenovic M, Ji S, Sun B. LEGENDplex™: Bead-assisted multiplex cytokine profiling by flow cytometry. Methods Enzymol 2019; 629:151-176. [PMID: 31727238 DOI: 10.1016/bs.mie.2019.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the past two decades there have been tremendous advances in our understanding of tumor immunology, which have in turn led to new and exciting immunology-based therapeutics. However, further research is needed into the dynamics and regulation of the immune response in the tumor microenvironment in order to achieve the full potential of these agents in treating all cancer patients. Defining the role of cytokines, chemokines, and other soluble mediators will be essential to this endeavor. This chapter describes, in detail, the technical protocol and applicability of LEGENDplex™ bead-based multiplex assays in quantifying these critical signaling molecules.
Collapse
|
92
|
Pei Y, Chen L, Huang Y, Wang J, Feng J, Xu M, Chen Y, Song Q, Jiang G, Gu X, Zhang Q, Gao X, Chen J. Sequential Targeting TGF-β Signaling and KRAS Mutation Increases Therapeutic Efficacy in Pancreatic Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900631. [PMID: 31033217 DOI: 10.1002/smll.201900631] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/15/2019] [Indexed: 06/09/2023]
Abstract
Pancreatic cancer is a highly aggressive malignancy that strongly resists extant treatments. The failure of existing therapies is majorly attributed to the tough tumor microenvironment (TME) limiting drug access and the undruggable targets of tumor cells. The formation of suppressive TME is regulated by transforming growth factor beta (TGF-β) signaling, while the poor response and short survival of almost 90% of pancreatic cancer patients results from the oncogenic KRAS mutation. Hence, simultaneously targeting both the TGF-β and KRAS pathways might dismantle the obstacles of pancreatic cancer therapy. Here, a novel sequential-targeting strategy is developed, in which antifibrotic fraxinellone-loaded CGKRK-modified nanoparticles (Frax-NP-CGKRK) are constructed to regulate TGF-β signaling and siRNA-loaded lipid-coated calcium phosphate (LCP) biomimetic nanoparticles (siKras-LCP-ApoE3) are applied to interfere with the oncogenic KRAS. Frax-NP-CGKRK successfully targets the tumor sites through the recognition of overexpressed heparan sulfate proteoglycan, reverses the activated cancer-associated fibroblasts (CAFs), attenuates the dense stroma barrier, and enhances tumor blood perfusion. Afterward, siKras-LCP-ApoE3 is efficiently internalized by the tumor cells through macropinocytosis and specifically silencing KRAS mutation. Compared with gemcitabine, this sequential-targeting strategy significantly elongates the lifespans of pancreatic tumor-bearing animals, hence providing a promising approach for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Yuanyuan Pei
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P. R. China
| | - Liang Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P. R. China
| | - Yukun Huang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P. R. China
| | - Jiahao Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P. R. China
| | - Jingxian Feng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P. R. China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P. R. China
| | - Yu Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P. R. China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Xiao Gu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Qian Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P. R. China
| | - Jun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P. R. China
- Department of Pharmacy, Pudong Hospital, Fudan University, 2800 Gongwei Road, Shanghai, 201399, P. R. China
| |
Collapse
|
93
|
Salmaninejad A, Valilou SF, Soltani A, Ahmadi S, Abarghan YJ, Rosengren RJ, Sahebkar A. Tumor-associated macrophages: role in cancer development and therapeutic implications. Cell Oncol (Dordr) 2019; 42:591-608. [PMID: 31144271 DOI: 10.1007/s13402-019-00453-z] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are known to play important roles in the initiation and progression of human cancers, as well as in angiogenesis. TAMs are considered as main components of the tumor microenvironment. Targeting TAMs may serve as a therapeutic strategy for the treatment of cancer. In this review, the signaling pathways, origin, function, polarization and clinical application of TAMs are discussed. The role of TAMs in tumor initiation, progression, angiogenesis, invasion and metastasis are also emphasized. In addition, a variety of clinical and pre-clinical approaches to target TAMs are discussed. CONCLUSIONS Clinical therapeutic approaches that show most promise include blocking the extravasation of TAMs along with using TAMs as diagnostic biomarkers for cancer progression. The targeting of TAMs in a variety of clinical settings appears to be a promising strategy for decreasing metastasis formation and for improving patient outcome.
Collapse
Affiliation(s)
- Arash Salmaninejad
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Medical Genetics Research Center, Student Research Committee, Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Farajzadeh Valilou
- Medical Genetics Network (MeGeNe), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Arash Soltani
- Medical Genetics Research Center, Student Research Committee, Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sepideh Ahmadi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yousef Jafari Abarghan
- Medical Genetics Research Center, Student Research Committee, Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rhonda J Rosengren
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran. .,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, P.O. box: 91779-48564, Mashhad, Iran.
| |
Collapse
|
94
|
Huang H, Li J, Hu WJ, Chen C, Luo HQ, Tang XD, Zhou KY, Zhong WT, Li XY. The serum level of CC chemokine ligand 18 correlates with the prognosis of non-small cell lung cancer. Int J Biol Markers 2019; 34:156-162. [DOI: 10.1177/1724600819829758] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Background: CC chemokine ligand 18 (CCL18) is a chemotactic cytokine involved in the pathogenesis and progression of various cancers. Our previous research showed that the expression of CCL18 is obviously higher in non-small cell lung cancer (NSCLC) than in the adjacent normal tissues, suggesting its role in NSCLC. Methods: We further examined the serum level of CCL18 in 80 NSCLC patients with enzyme-linked immunosorbent assay and simultaneously analyzed the survival curve of these patients by the Kaplan–Meier method, and then utilized a log-rank test to evaluate the correlation of CCL18 expression with the malignant progression of NSCLC. Results: Our results showed that the median serum concentration of CCL18 was significantly elevated to 436.11 ng/mL in NSCLC patients compared to 41.97 ng/ml in healthy people ( P<0.01), which was also positively related to the expression of lung cancer biomarkers carcinoma–embryonic antigen and cytokeratin fragment antigen 21-1. Moreover, correlation analysis showed that an increased level of serum CCL18 was associated with a worse survival time in NSCLC patients. Conclusion: Our findings suggest that the serum CCL18 level of NSCLC patients was negatively correlated with the prognosis, thus suggesting that CCL18 may serve as a potential circulating biomarker for NSCLC diagnosis.
Collapse
Affiliation(s)
- Hui Huang
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, P.R. China
| | - Jing Li
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, P.R. China
| | - Wen-jia Hu
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, P.R. China
| | - Chen Chen
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, P.R. China
| | - Hai-qing Luo
- Center of Oncology, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, P.R. China
| | - Xu-dong Tang
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, P.R. China
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, P.R. China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, P.R. China
| | - Ke-yuan Zhou
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, P.R. China
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, P.R. China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, P.R. China
| | - Wang-tao Zhong
- Department of Neurology, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, P.R. China
| | - Xiang-yong Li
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, P.R. China
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, P.R. China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, P.R. China
| |
Collapse
|
95
|
20-Hydroxy-3-Oxolupan-28-Oic Acid Attenuates Inflammatory Responses by Regulating PI3K⁻Akt and MAPKs Signaling Pathways in LPS-Stimulated RAW264.7 Macrophages. Molecules 2019; 24:molecules24030386. [PMID: 30678231 PMCID: PMC6385096 DOI: 10.3390/molecules24030386] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/16/2022] Open
Abstract
20-Hydroxy-3-oxolupan-28-oic acid (HOA), a lupane-type triterpene, was obtained from the leaves of Mahonia bealei, which is described in the Chinese Pharmacopeia as a remedy for inflammation and related diseases. The anti-inflammatory mechanisms of HOA, however, have not yet been fully elucidated. Therefore, the objective of this study was to characterize the molecular mechanisms of HOA in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. HOA suppressed the release of nitric oxide (NO), pro-inflammatory cytokine tumor necrosis factor α (TNF-α), and interleukin 6 (IL-6) in LPS-stimulated RAW264.7 macrophages without affecting cell viability. Quantitative real-time reverse-transcription polymerase chain reaction (RT-qPCR) analysis indicated that HOA also suppressed the gene expression of inducible NO synthase (iNOS), TNF-α, and IL-6. Further analyses demonstrated that HOA inhibited the phosphorylation of upstream signaling molecules, including p85, PDK1, Akt, IκBα, ERK, and JNK, as well as the nuclear translocation of nuclear factor κB (NF-κB) p65. Interestingly, HOA had no effect on the LPS-induced nuclear translocation of activator protein 1 (AP-1). Taken together, these results suggest that HOA inhibits the production of cytokine by downregulating iNOS, TNF-α, and IL-6 gene expression via the downregulation of phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein kinases (MAPKs), and the inhibition of NF-κB activation. Our findings indicate that HOA could potentially be used as an anti-inflammatory agent for medical use.
Collapse
|
96
|
Obr AE, Kumar S, Chang YJ, Bulatowicz JJ, Barnes BJ, Birge RB, Lazzarino DA, Gallagher E, LeRoith D, Wood TL. Insulin-like growth factor receptor signaling in breast tumor epithelium protects cells from endoplasmic reticulum stress and regulates the tumor microenvironment. Breast Cancer Res 2018; 20:138. [PMID: 30458886 PMCID: PMC6245538 DOI: 10.1186/s13058-018-1063-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Early analyses of human breast cancer identified high expression of the insulin-like growth factor type 1 receptor (IGF-1R) correlated with hormone receptor positive breast cancer and associated with a favorable prognosis, whereas low expression of IGF-1R correlated with triple negative breast cancer (TNBC). We previously demonstrated that the IGF-1R acts as a tumor and metastasis suppressor in the Wnt1 mouse model of TNBC. The mechanisms for how reduced IGF-1R contributes to TNBC phenotypes is unknown. METHODS We analyzed the METABRIC dataset to further stratify IGF-1R expression with patient survival and specific parameters of TNBC. To investigate molecular events associated with the loss of IGF-1R function in breast tumor cells, we inhibited IGF-1R in human cell lines using an IGF-1R blocking antibody and analyzed MMTV-Wnt1-mediated mouse tumors with reduced IGF-1R function through expression of a dominant-negative transgene. RESULTS Our analysis of the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset revealed association between low IGF-1R and reduced overall patient survival. IGF-1R expression was inversely correlated with patient survival even within hormone receptor-positive breast cancers, indicating reduced overall patient survival with low IGF-1R was not due simply to low IGF-1R expression within TNBCs. Inhibiting IGF-1R in either mouse or human tumor epithelial cells increased reactive oxygen species (ROS) production and activation of the endoplasmic reticulum stress response. IGF-1R inhibition in tumor epithelial cells elevated interleukin (IL)-6 and C-C motif chemokine ligand 2 (CCL2) expression, which was reversed by ROS scavenging. Moreover, the Wnt1/dnIGF-1R primary tumors displayed a tumor-promoting immune phenotype. The increased CCL2 promoted an influx of CD11b+ monocytes into the primary tumor that also had increased matrix metalloproteinase (MMP)-2, MMP-3, and MMP-9 expression. Increased MMP activity in the tumor stroma was associated with enhanced matrix remodeling and collagen deposition. Further analysis of the METABRIC dataset revealed an increase in IL-6, CCL2, and MMP-9 expression in patients with low IGF-1R, consistent with our mouse tumor model and data in human breast cancer cell lines. CONCLUSIONS Our data support the hypothesis that reduction of IGF-1R function increases cellular stress and cytokine production to promote an aggressive tumor microenvironment through infiltration of immune cells and matrix remodeling.
Collapse
Affiliation(s)
- Alison E Obr
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Sushil Kumar
- Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Yun-Juan Chang
- Office of Advance Research Computing, Rutgers-New Jersey Medical School, Newark, NJ, 07102, USA
| | - Joseph J Bulatowicz
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Betsy J Barnes
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Raymond B Birge
- Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Deborah A Lazzarino
- Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA
| | - Emily Gallagher
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Icahn Sinai School of Medicine at Mt. Sinai, New York, NY, 10029, USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Icahn Sinai School of Medicine at Mt. Sinai, New York, NY, 10029, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, 07101, USA.
| |
Collapse
|
97
|
Tan X, Zhang Z, Yao H, Shen L. RETRACTED: Tim-4 promotes the growth of colorectal cancer by activating angiogenesis and recruiting tumor-associated macrophages via the PI3K/AKT/mTOR signaling pathway. Cancer Lett 2018; 436:119-128. [PMID: 30118845 DOI: 10.1016/j.canlet.2018.08.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/14/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the authors. After publication, the authors noted unreliable data in the experimental validations. Specifically, representative images of IHC staining for CD31 and α-SMA in CT26-Vector and CT26-Tim-4 tumors in Fig. 2C and the expression of MMP9, VEGF and LOX in CT26-Tim-4 and CT26-Vector tumors was assessed by immunohistochemistry in Fig. 2E; incorrect photos were provided due to file placement confusion. All authors “agree with this retraction and deeply regret these errors and apologize to the editorial board and readers for any inconvenience caused.” Additionally, after publication, the journal was made aware of comments in relation to this article (https://pubpeer.com/publications/D191AE022DDD7B810564039CB16395). The authors have not responded to the journal request to respond to these comments.
Collapse
Affiliation(s)
- Xiao Tan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Zhongqiang Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Hongliang Yao
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
98
|
Gieseler F, Plattfaut C, Quecke T, Freund A, Ungefroren H, Ender F. Heterogeneity of microvesicles from cancer cell lines under inflammatory stimulation with TNF-α. Cell Biol Int 2018; 42:1533-1544. [PMID: 30080276 DOI: 10.1002/cbin.11040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/29/2018] [Indexed: 12/11/2022]
Abstract
Microvesicles (MVs) represent a subgroup of extracellular vesicles (EVs) emerging from various cells by blebbing of their outer membrane. Therefore, they share features such as membrane composition and antigenicity with their parental cells. Released by many immune and tumor cells, MVs act as intercellular messengers, account for horizontal gene transfer and can activate the coagulation system. With the aim to investigate their relevance for tumor cell biology, we characterized MVs released by human tumor cell lines of various origins in the absence or presence of TNF-α. After stimulation, we used the combination of low and high-speed centrifugation to enrich MVs from cell culture supernatants. We analyzed the presentation of phosphatidylserine (PS) and tissue factor (TF) activity on the cell surface and investigated their potency to induce tumor cell migration. In all tumor cell lines, TNF-α stimulation enhanced the release of MVs. While the expression of PS was universally increased, an elevated activity of procoagulant TF could be detected on MVs from lung, pancreatic, and colon carcinoma, but not from breast and ovarian cancer cell lines. Functionally, TNF-α stimulation significantly increased the potency of MVs to induce tumor cell migration. In conclusion, inflammatory conditions promote the release of MVs with increased procoagulant activity from tumor cell lines in vitro. PS-containing and TF-expressing MVs may account for systemic activation of the coagulation system as seen in cancer patients and, since they induce tumor cell migration, they may serve as biomarkers for tumor progression.
Collapse
Affiliation(s)
- Frank Gieseler
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| | - Corinna Plattfaut
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| | - Tabea Quecke
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| | - Annika Freund
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| | - Hendrik Ungefroren
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany.,Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Kiel, 24105, Germany
| | - Fanny Ender
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| |
Collapse
|