51
|
Nie S, Huili Y, Yao A, Liu J, Wang Y, Wang L, Zhang L, Kang S, Cao F. Identification of subtypes of clear cell renal cell carcinoma and construction of a prognostic model based on fatty acid metabolism genes. Front Genet 2022; 13:1013178. [PMID: 36186450 PMCID: PMC9523225 DOI: 10.3389/fgene.2022.1013178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The effects of fatty acid metabolism in many tumors have been widely reported. Due to the diversity of lipid synthesis, uptake, and transformation in clear cell renal cell carcinoma (ccRCC) cells, many studies have shown that ccRCC is associated with fatty acid metabolism. The study aimed was to explore the impact of fatty acid metabolism genes on the prognosis and immunotherapy of ccRCC.Methods: Two subtypes were distinguished by unsupervised clustering analysis based on the expression of 309 fatty acid metabolism genes. A prognostic model was constructed by lasso algorithm and multivariate COX regression analysis using fatty acid metabolism genes as the signatures. The tumor microenvironment between subtypes and between risk groups was further analyzed. The International Cancer Genome Consortium cohort was used for external validation of the model.Results: The analysis showed that subtype B had a poorer prognosis and a higher degree of immune infiltration. The high-risk group had a poorer prognosis and higher tumor microenvironment scores. The nomogram could accurately predict patient survival.Conclusion: Fatty acid metabolism may affect the prognosis and immune infiltration of patients with ccRCC. The analysis was performed to understand the potential role of fatty acid metabolism genes in the immune infiltration and prognosis of patients. These findings have implications for individualized treatment, prognosis, and immunization for patients with ccRCC.
Collapse
Affiliation(s)
- Shiwen Nie
- North China University of Science and Technology, Tangshan, China
| | - Youlong Huili
- North China University of Science and Technology, Tangshan, China
| | - Anliang Yao
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Jian Liu
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Yong Wang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Lei Wang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Liguo Zhang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Shaosan Kang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Fenghong Cao
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
- *Correspondence: Fenghong Cao,
| |
Collapse
|
52
|
Zou R, Jiang Q, Jin T, Chen M, Yao L, Ding H. Pan-cancer analyses and molecular subtypes based on the cancer-associated fibroblast landscape and tumor microenvironment infiltration characterization reveal clinical outcome and immunotherapy response in epithelial ovarian cancer. Front Immunol 2022; 13:956224. [PMID: 36032075 PMCID: PMC9402225 DOI: 10.3389/fimmu.2022.956224] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background Cancer-associated fibroblasts (CAFs) are essential components of the tumor microenvironment (TME). These cells play a supportive role throughout cancer progression. Their ability to modulate the immune system has also been noted. However, there has been limited investigation of CAFs in the TME of epithelial ovarian cancer (EOC). Methods We comprehensively evaluated the CAF landscape and its association with gene alterations, clinical features, prognostic value, and immune cell infiltration at the pan-cancer level using multi-omic data from The Cancer Genome Atlas (TCGA). The CAF contents were characterized by CAF scores based on the expression levels of seven CAF markers using the R package “GSVA.” Next, we identified the molecular subtypes defined by CAF markers and constructed a CAF riskscore system using principal component analysis in the EOC cohort. The correlation between CAF riskscore and TME cell infiltration was investigated. The ability of the CAF riskscore to predict prognosis and immunotherapy response was also examined. Results CAF components were involved in multiple immune-related processes, including transforming growth factor (TGF)-β signaling, IL2-STAT signaling, inflammatory responses, and Interleukin (IL) 2-signal transducer and activator of transcription (STAT) signaling. Considering the positive correlation between CAF scores and macrophages, neutrophils, and mast cells, CAFs may exert immunosuppressive effects in both pan-cancer and ovarian cancer cohorts, which may explain accelerated tumor progression and poor outcomes. Notably, two distinct CAF molecular subtypes were defined in the EOC cohort. Low CAF riskscores were characterized by favorable overall survival (OS) and higher efficacy of immunotherapy. Furthermore, 24 key genes were identified in CAF subtypes. These genes were significantly upregulated in EOC and showed a strong correlation with CAF markers. Conclusions Identifying CAF subtypes provides insights into EOC heterogeneity. The CAF riskscore system can predict prognosis and select patients who may benefit from immunotherapy. The mechanism of interactions between key genes, CAF markers, and associated cancer-promoting effects needs to be further elucidated.
Collapse
Affiliation(s)
- Ruoyao Zou
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qidi Jiang
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Tianqiang Jin
- Department of General Surgery, ShengJing Hospital of China Medical University, Shenyang, China
| | - Mo Chen
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Liangqing Yao
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- *Correspondence: Hongda Ding, ; Liangqing Yao,
| | - Hongda Ding
- Department of General Surgery, ShengJing Hospital of China Medical University, Shenyang, China
- *Correspondence: Hongda Ding, ; Liangqing Yao,
| |
Collapse
|
53
|
Kumar A, Taghi Khani A, Sanchez Ortiz A, Swaminathan S. GM-CSF: A Double-Edged Sword in Cancer Immunotherapy. Front Immunol 2022; 13:901277. [PMID: 35865534 PMCID: PMC9294178 DOI: 10.3389/fimmu.2022.901277] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/06/2022] [Indexed: 12/23/2022] Open
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine that drives the generation of myeloid cell subsets including neutrophils, monocytes, macrophages, and dendritic cells in response to stress, infections, and cancers. By modulating the functions of innate immune cells that serve as a bridge to activate adaptive immune responses, GM-CSF globally impacts host immune surveillance under pathologic conditions. As with other soluble mediators of immunity, too much or too little GM-CSF has been found to promote cancer aggressiveness. While too little GM-CSF prevents the appropriate production of innate immune cells and subsequent activation of adaptive anti-cancer immune responses, too much of GM-CSF can exhaust immune cells and promote cancer growth. The consequences of GM-CSF signaling in cancer progression are a function of the levels of GM-CSF, the cancer type, and the tumor microenvironment. In this review, we first discuss the secretion of GM-CSF, signaling downstream of the GM-CSF receptor, and GM-CSF’s role in modulating myeloid cell homeostasis. We then outline GM-CSF’s anti-tumorigenic and pro-tumorigenic effects both on the malignant cells and on the non-malignant immune and other cells in the tumor microenvironment. We provide examples of current clinical and preclinical strategies that harness GM-CSF’s anti-cancer potential while minimizing its deleterious effects. We describe the challenges in achieving the Goldilocks effect during administration of GM-CSF-based therapies to patients with cancer. Finally, we provide insights into how technologies that map the immune microenvironment spatially and temporally may be leveraged to intelligently harness GM-CSF for treatment of malignancies.
Collapse
Affiliation(s)
- Anil Kumar
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Adeleh Taghi Khani
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Ashly Sanchez Ortiz
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
| | - Srividya Swaminathan
- Department of Systems Biology, Beckman Research Institute of City of Hope, Monrovia, CA, United States
- Department of Hematological Malignancies, Beckman Research Institute of City of Hope, Monrovia, CA, United States
- *Correspondence: Srividya Swaminathan,
| |
Collapse
|
54
|
Liu Q, Sun W, Zhang H. Roles and new Insights of Macrophages in the Tumor Microenvironment of Thyroid Cancer. Front Pharmacol 2022; 13:875384. [PMID: 35479325 PMCID: PMC9035491 DOI: 10.3389/fphar.2022.875384] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/21/2022] [Indexed: 12/17/2022] Open
Abstract
Although most thyroid cancers have a good and predictable prognosis, the anaplastic, medullary, and refractory thyroid cancers still prone to recurrence and metastasis, resulting in poor prognosis. Although a number of newly developed targeted therapies have begun to be indicated for the above types of thyroid cancer in recent years, their ability to improve overall survival remain hindered by low efficacy. As the largest component of immune cells in tumor microenvironment, tumor-associated macrophages play a key role in the invasion and metastasis of thyroid cancer. There is much evidence that the immune system, tumor microenvironment and cancer stem cell interactions may revolutionize traditional therapeutic directions. Tumor-associated macrophages have been extensively studied in a variety of tumors, however, research on the relationship between thyroid cancer and macrophages is still insufficient. In this review, we summarize the functions of tumor-associated macrophages in different types of thyroid cancer, their cytokines or chemokines effect on thyroid cancer and the mechanisms that promote tumor proliferation and migration. In addition, we discuss the mechanisms by which tumor-associated macrophages maintain the stemness of thyroid cancer and potential strategies for targeting tumor-associated macrophages to treat thyroid cancer.
Collapse
Affiliation(s)
| | | | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
55
|
Abstract
Tumour-associated macrophages (TAMs) constitute a plastic and heterogeneous cell population of the tumour microenvironment (TME) that can account for up to 50% of solid tumours. TAMs heterogeneous are associated with different cancer types and stages, different stimulation of bioactive molecules and different TME, which are crucial drivers of tumour progression, metastasis and resistance to therapy. In this context, understanding the sources and regulatory mechanisms of TAM heterogeneity and searching for novel therapies targeting TAM subpopulations are essential for future studies. In this review, we discuss emerging evidence highlighting the redefinition of TAM heterogeneity from three different directions: origins, phenotypes and functions. We notably focus on the causes and consequences of TAM heterogeneity which have implications for the evolution of therapeutic strategies that targeted the subpopulations of TAMs.
Collapse
|
56
|
Parsons TM, Buelow KL, Hanna A, Brake MA, Poma C, Hosch SE, Westrick RJ, Villa-Diaz LG, Wilson GD, Madlambayan GJ. Intratumoural haematopoietic stem and progenitor cell differentiation into M2 macrophages facilitates the regrowth of solid tumours after radiation therapy. Br J Cancer 2022; 126:927-936. [PMID: 34931040 PMCID: PMC8927108 DOI: 10.1038/s41416-021-01652-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 11/03/2021] [Accepted: 11/22/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Bone-marrow-derived haematopoietic stem and progenitor cells (HSPCs) are a prominent part of the highly complex tumour microenvironment (TME) where they localise within tumours and maintain haematopoietic potency. Understanding the role HSPCs play in tumour growth and response to radiation therapy (RT) may lead to improved patient treatments and outcomes. METHODS We used a mouse model of non-small cell lung carcinoma where tumours were exposed to RT regimens alone or in combination with GW2580, a pharmacological inhibitor of colony stimulating factor (CSF)-1 receptor. RT-PCR, western blotting and immunohistochemistry were used to quantify expression levels of factors that affect HSPC differentiation. DsRed+ HSPC intratumoural activity was tracked using flow cytometry and confocal microscopy. RESULTS We demonstrated that CSF-1 is enhanced in the TME following exposure to RT. CSF-1 signaling induced intratumoural HSPC differentiation into M2 polarised tumour-associated macrophages (TAMs), aiding in post-RT tumour survival and regrowth. In contrast, hyperfractionated/pulsed radiation therapy (PRT) and GW2580 ablated this process resulting in improved tumour killing and mouse survival. CONCLUSIONS Tumours coopt intratumoural HSPC fate determination via CSF-1 signaling to overcome the effects of RT. Thus, limiting intratumoural HSPC activity represents an attractive strategy for improving the clinical treatment of solid tumours.
Collapse
Affiliation(s)
- Tyler M Parsons
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
- Department of Radiation Oncology, Beaumont Research Institute, Royal Oak, MI, USA
| | - Katie L Buelow
- Department of Radiation Oncology, Beaumont Research Institute, Royal Oak, MI, USA
| | - Alaa Hanna
- Department of Radiation Oncology, Beaumont Research Institute, Royal Oak, MI, USA
| | - Marisa A Brake
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Crystal Poma
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Sarah E Hosch
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
| | - Randal J Westrick
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
- Department of Bioengineering, Oakland University, Rochester, MI, USA
- Oakland University Center for Data Science and Big Data Analytics, Rochester, MI, USA
| | - Luis G Villa-Diaz
- Department of Biological Sciences, Oakland University, Rochester, MI, USA
- Department of Bioengineering, Oakland University, Rochester, MI, USA
| | - George D Wilson
- Department of Radiation Oncology, Beaumont Research Institute, Royal Oak, MI, USA
| | - Gerard J Madlambayan
- Department of Biological Sciences, Oakland University, Rochester, MI, USA.
- Department of Bioengineering, Oakland University, Rochester, MI, USA.
| |
Collapse
|
57
|
Rah B, Rather RA, Bhat GR, Baba AB, Mushtaq I, Farooq M, Yousuf T, Dar SB, Parveen S, Hassan R, Mohammad F, Qassim I, Bhat A, Ali S, Zargar MH, Afroze D. JAK/STAT Signaling: Molecular Targets, Therapeutic Opportunities, and Limitations of Targeted Inhibitions in Solid Malignancies. Front Pharmacol 2022; 13:821344. [PMID: 35401182 PMCID: PMC8987160 DOI: 10.3389/fphar.2022.821344] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
JAK/STAT signaling pathway is one of the important regulatory signaling cascades for the myriad of cellular processes initiated by various types of ligands such as growth factors, hormones, and cytokines. The physiological processes regulated by JAK/STAT signaling are immune regulation, cell proliferation, cell survival, apoptosis and hematopoiesis of myeloid and non-myeloid cells. Dysregulation of JAK/STAT signaling is reported in various immunological disorders, hematological and other solid malignancies through various oncogenic activation mutations in receptors, downstream mediators, and associated transcriptional factors such as STATs. STATs typically have a dual role when explored in the context of cancer. While several members of the STAT family are involved in malignancies, however, a few members which include STAT3 and STAT5 are linked to tumor initiation and progression. Other STAT members such as STAT1 and STAT2 are pivotal for antitumor defense and maintenance of an effective and long-term immune response through evolutionarily conserved programs. The effects of JAK/STAT signaling and the persistent activation of STATs in tumor cell survival; proliferation and invasion have made the JAK/STAT pathway an ideal target for drug development and cancer therapy. Therefore, understanding the intricate JAK/STAT signaling in the pathogenesis of solid malignancies needs extensive research. A better understanding of the functionally redundant roles of JAKs and STATs may provide a rationale for improving existing cancer therapies which have deleterious effects on normal cells and to identifying novel targets for therapeutic intervention in solid malignancies.
Collapse
|
58
|
M1 macrophage-derived exosomes and their key molecule lncRNA HOTTIP suppress head and neck squamous cell carcinoma progression by upregulating the TLR5/NF-κB pathway. Cell Death Dis 2022; 13:183. [PMID: 35210436 PMCID: PMC8873565 DOI: 10.1038/s41419-022-04640-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 01/28/2022] [Accepted: 02/09/2022] [Indexed: 12/29/2022]
Abstract
Exosomes serve as a crucial mode of communication between tumor-associated macrophages (TAMs) and cancer cells. This study attempted to explore the function of M1-derived exosomes and clarify their specific mechanism in head and neck squamous cell carcinoma (HNSCC). Moreover, the functional roles of M1-derived exosomes and their key molecule long noncoding RNA (lncRNA) HOXA transcript at the distal tip (HOTTIP) in HNSCC were investigated by conducting a series of in vitro and in vivo experiments. The dual-luciferase test was utilized to clarify the binding capacities between HOTTIP/mRNA and miRNAs. Accordingly, HOTTIP was found to be upregulated in M1-derived exosomes. Meanwhile, the in vitro experiments indicated that M1 exosomes suppressed proliferation, migration and invasion but induced apoptosis of cancer cells. This function was noted to be enhanced by HOTTIP-overexpressed M1 exosomes but was weakened by HOTTIP-knockdown ones, indicating that HOTTIP serves as a key molecule in M1 exosomes. Therefore, the function of HOTTIP in cancer cells was explored, for which overexpression of HOTTIP was found to inhibit proliferation, migration and invasion but induced apoptosis of cancer cells in vitro. A mechanism study further showed that M1 exosomes and HOTTIP activated the TLR5/NF-κB signaling pathway by competitively sponging miR-19a-3p and miR-19b-3p. Furthermore, cancer cells expressing HOTTIP were noted to induce the polarization of both local M1 and M2 macrophages; however, M1 exosomes were observed to reprogram local TAMs into M1 macrophages. More importantly, both cancer cells expressing HOTTIP and M1 exosomes reeducated circulating monocytes to express the M1 phenotype. The corresponding data demonstrated that the M1 exosomal lncRNA HOTTIP suppresses HNSCC progression by upregulating the TLR5/NF-κB signaling pathway through competitively sponging miR-19a-3p and miR-19b-3p. In particular, M1 exosomes and HOTTIP induce the polarization of M1 in circulating monocytes, thus providing novel insight into HNSCC immunotherapy.
Collapse
|
59
|
Cao H, Wang D, Gao R, Feng Y, Chen L. Qi Ling decreases paclitaxel resistance in the human prostate cancer by reversing tumor-associated macrophages function. Aging (Albany NY) 2022; 14:1812-1821. [PMID: 35193986 PMCID: PMC8908933 DOI: 10.18632/aging.203904] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/08/2022] [Indexed: 06/14/2023]
Abstract
Tumor-associated macrophages (TAMs) are critical immune cells infiltrated into tumor. In present study, we evaluated the effects of Qi Ling (QL), a traditional Chinese medicine on paclitaxel resistance in prostate cancer cells and explored the underlying mechanisms. We administrated QL to rats and collected the serum from QL-treated rats (QL-serum). We established the co-culture system of TAMs/paclitaxel resistant prostate cancer cells. We treated the TAMs with QL-serum and measured the viability of paclitaxel resistant prostate cancer cells after exposing to paclitaxel. We monitored the expression of M1 and M2 markers, the expression and activation of IL-6/STAT3 signaling pathways in TAMs after QL treatment. We treated TAMs with QL-serum together with interleukin (IL)-6, measured the expression of M1 and M2 markers, and the viability of paclitaxel resistant prostate cancer cells. In co-culture system, QL-serum-treated TAMs decreased the paclitaxel resistance in the human prostate cancer cells. QL-serum treatment significantly up-regulated the expression of M1 markers inducible nitric oxide synthase and tumor necrosis factor α while decreased the expression of M2 markers IL-10 and chemokine (C-C motif) ligand 22. QL-serum suppressed the activation of IL-6/ signal transducer and activator of transcription 3 signaling pathway. All these effects of QL-serum were abolished in the presence of IL-6. Qi Ling re-programmed TAMs and decreases paclitaxel resistance in prostate cancer cells.
Collapse
Affiliation(s)
- Hongwen Cao
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, Xuhui, Shanghai 200032, China
| | - Dan Wang
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, Xuhui, Shanghai 200032, China
| | - Renjie Gao
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, Xuhui, Shanghai 200032, China
| | - Yigeng Feng
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, Xuhui, Shanghai 200032, China
| | - Lei Chen
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, Xuhui, Shanghai 200032, China
| |
Collapse
|
60
|
Albracht SP. Immunotherapy with GcMAF revisited - A critical overview of the research of Nobuto Yamamoto. Cancer Treat Res Commun 2022; 31:100537. [PMID: 35217488 DOI: 10.1016/j.ctarc.2022.100537] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 06/14/2023]
Abstract
This overview describes the research of Nobutu Yamamoto (Philadelphia) concerning immunotherapy with GcMAF for patients with cancer and for patients infected with pathogenic envelope viruses. GcMAF (Group-specific component Macrophage-Activating Factor) is a mammalian protein with an incredible potency to directly activate macrophages. Since the late 1980s Yamamoto's investigations were published in numerous journals but in order to understand the details of his research, a minute survey of many of his patents was required. But even then, regrettably, a precise description of his experiments was sometimes lacking. This overview tries to summarize all of Yamamoto's research on GcMAF, as well as some selected more recent papers from other investigators, who tried to verify and/or reproduce Yamamoto's reports. In my opinion the most important result of the GcMAF research deserves widespread renewed attention: human GcMAF injections (100 ng per week, intramuscular or intravenous) can help to cure patients with a great variety of cancers as well as patients infected with pathogenic envelope viruses like the human immunodeficiency virus 1 (HIV-1), influenza, measles and rubella (and maybe also SARS-CoV-2). From Yamamoto's data it can be calculated that GcMAF is a near-stoichiometric activator of macrophages. Yamamoto monitored the progress of his immunotherapy via the serum level of an enzyme called nagalase (α-N-acetylgalactosaminidase activity at pH 6). I have extensively discussed the properties and potential catalytic site of this enzyme activity in an Appendix entitled: "Search for the potential active site of the latent α-N-acetylgalactosaminidase activity in the glycoproteins of some envelope viruses".
Collapse
Affiliation(s)
- Simon Pj Albracht
- Biochemist, retired from the Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
61
|
Oncolytic adenovirus inhibits malignant ascites of advanced ovarian cancer by reprogramming the ascitic immune microenvironment. Mol Ther Oncolytics 2021; 23:488-500. [PMID: 34901391 PMCID: PMC8637216 DOI: 10.1016/j.omto.2021.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 11/07/2021] [Indexed: 11/21/2022] Open
|
62
|
Salomaa T, Pemmari T, Määttä J, Kummola L, Salonen N, González-Rodríguez M, Parviainen L, Hiihtola L, Vähätupa M, Järvinen TAH, Junttila IS. IL-13Rα1 Suppresses Tumor Progression in Two-stage Skin Carcinogenesis Model by Regulating Regulatory T Cells. J Invest Dermatol 2021; 142:1565-1575.e17. [PMID: 34808240 DOI: 10.1016/j.jid.2021.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 12/19/2022]
Abstract
Type 2-inflammation-related cytokine Interleukin (IL)-13 plays a protective role in experimental papilloma induction in mice. To understand mechanisms by which IL-13 contributes to papilloma formation we utilized IL-13Rα1 knockout (KO) mice in widely used DMBA/TPA two-stage skin carcinogenesis protocol that mimics the development of Squamous Cell Carcinoma (SCC). KO mice developed more papillomas and significantly faster than wild-type (WT) mice. Papilloma development reduced Tregs in WT mice, but substantially less in KO mice. In line with this, IL-2 and IL-10 levels decreased in WT mice, but not in KO mice. Furthermore, systemic IL-5 and Thymic Stromal Lymphopoietin (TSLP) levels were elevated, while IL-22 was decreased during papilloma formation in the skin of KO mice. Polymorphonuclear Myeloid-derived suppressor cells (PMN-MDSCs) were decreased in the KO mice at the early phase of papilloma induction. We demonstrate that IL-13Rα1 protects from papilloma development in chemically induced skin carcinogenesis and our results provide further insights into the protective role of functional IL-4 and IL-13 signaling via type II IL-4R in tumor development.
Collapse
Affiliation(s)
- Tanja Salomaa
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Fimlab Laboratories, Tampere, Finland
| | - Toini Pemmari
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Juuso Määttä
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Laura Kummola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Fimlab Laboratories, Tampere, Finland
| | - Niklas Salonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Liisa Parviainen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Lotta Hiihtola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maria Vähätupa
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tero A H Järvinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Tampere University Hospital, Tampere, Finland
| | - Ilkka S Junttila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Fimlab Laboratories, Tampere, Finland.
| |
Collapse
|
63
|
MicroRNAs: Their Role in Metabolism, Tumor Microenvironment, and Therapeutic Implications in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13225604. [PMID: 34830755 PMCID: PMC8615702 DOI: 10.3390/cancers13225604] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/30/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Head and neck squamous cell carcinoma (HNSCC), which arises from the oral epithelium, is one of the most common cancers worldwide. Despite excellent diagnosis and treatment improvements, the mortality rate associated with HNSCC is still extremely high. Current data suggest that dysregulation of exosomes and metabolic abnormalities are involved in the initiation and progression of HNSCC. Thus, approaches for targeting exosomes in the tumor microenvironment and metabolic reprogramming pathways represent potential therapeutic strategies. Moreover, some miRNAs are thought to have significant functions in regulating the progression of HNSCC. The present article aims to summarize the current knowledge concerning the important miRNAs in both exosomes and cancer metabolism, as well as discuss future perspectives regarding their future diagnostic potential and treatment recommendations. Abstract MicroRNAs (miRNAs) are endogenous small non-coding RNA molecules that negatively regulate gene expression by binding to target mRNAs. Deregulated miRNAs can act as either oncogenic miRNAs or tumor suppressor miRNAs in controlling proliferation, differentiation, apoptosis, metastasis, epithelial–mesenchymal transition, and immune responses, which are all involved in the carcinogenesis process of HNSCC. Recent findings have shown that metabolic reprogramming is an important hallmark of cancer, which is necessary for malignant transformation and tumor development. Some reprogrammed metabolisms are believed to be required for HNSCC against an unfavorable tumor microenvironment (TME). The TME is composed of various cell types embedded in the altered extracellular matrix, among which exosomes, secreted by cancer cells, are one of the most important factors. Tumor-derived exosomes reshape the tumor microenvironment and play a crucial role in cell-to-cell communication during HNSCC development. Exosomes encapsulate many biomolecules, including miRNAs, circulate in body fluids, and can transmit intercellular regulatory messages to nearby and distant sites, which indicates that exosomal miRNAs have the potential to become non-invasive biomarkers. This review aims to clarify the functions of diverse miRNAs in HNSCC metabolic reprogramming and tumor-derived exosomes. In addition, it also emphasizes the potential role of miRNA as a biomarker in the diagnosis, prognosis, and treatment of HNSCC cancer.
Collapse
|
64
|
Zhang W, Yao S, Huang H, Zhou H, Zhou H, Wei Q, Bian T, Sun H, Li X, Zhang J, Liu Y. Molecular subtypes based on ferroptosis-related genes and tumor microenvironment infiltration characterization in lung adenocarcinoma. Oncoimmunology 2021; 10:1959977. [PMID: 34527427 PMCID: PMC8437492 DOI: 10.1080/2162402x.2021.1959977] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Recently, several molecular subtypes with different prognosis have been found in lung adenocarcinoma (LUAD). However, the characteristics of the ferroptosis molecular subtypes and the associated tumor microenvironment (TME) cell infiltration have not been fully studied in LUAD. Using 1160 lung adenocarcinoma samples, we explored the molecular subtypes mediated by ferroptosis-related genes, along with the associated TME cell infiltration. The ferroptosis score was constructed using the least absolute shrinkage and selection operator regression (LASSO) method to quantify the ferroptosis characteristics of a single tumor. Three different molecular subtypes related to ferroptosis, with different prognoses, were identified in LUAD. Analysis of TME cell infiltration revealed immune heterogeneity among the three subtypes. Cluster A was characterized by immunosuppression and was associated with stromal activation. Cluster C was characterized by a large number of immune cells infiltrating the TME, promoting tumor immune response, and it was significantly enriched in immune activation-related signaling pathways. Relatively less infiltration of immune cells was a feature of cluster B. The ferroptosis score can predict tumor subtype, immunity and prognosis. A low ferroptosis score was characterized by immune activation and good prognosis, as seen in the cluster C subtype. Relative immunosuppression and poor prognosis were the characteristics of a high ferroptosis score, as seen in cluster A and B subtypes. At the same time, the anti-PD-1/L1 immunotherapy cohort demonstrated that a low ferroptosis score was associated with higher efficacy of immunotherapy. The ferroptosis score is a promising biomarker that could be of great significance to determine the prognosis, molecular subtypes, TME cell infiltration characteristics and immunotherapy effects in patients with LUAD.
Collapse
Affiliation(s)
- Weiju Zhang
- Department of Pathology, Affiliated Hospital of Nantong University and Medical School of Nantong University, Nantong, China
| | - Sumei Yao
- Department Of Respiratory, Nantong First People ' s Hospital, Second Affiliated Hospital of Nantong University, Nantong, China
| | - Hua Huang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hao Zhou
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Haomiao Zhou
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Qishuang Wei
- Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Tingting Bian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hui Sun
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoli Li
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University and Medical School of Nantong University, Nantong, China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University and Medical School of Nantong University, Nantong, China
| |
Collapse
|
65
|
Yang S, Zhao W, Zhu M, Hu H, Wang W, Zang Z, Jin M, Bi J, Huang J, Liu C, Li X, Yin P, Li N. Tumor Temporal Proteome Profiling Reveals the Immunological Triple Offensive Induced by Synthetic Anti-Cancer Salmonella. Front Immunol 2021; 12:712936. [PMID: 34489962 PMCID: PMC8417115 DOI: 10.3389/fimmu.2021.712936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/03/2021] [Indexed: 01/30/2023] Open
Abstract
The engineered “obligate” anaerobic Salmonella typhimurium strain YB1 shows a prominent ability to repress tumor growth and metastasis, which has great potential as a novel cancer immunotherapy. However, the antitumor mechanism of YB1 remains unelucidated. To resolve the proteome dynamics induced by the engineered bacteria, we applied tumor temporal proteome profiling on murine bladder tumors after intravenous injection of either YB1 or PBS as a negative control. Our data suggests that during the two weeks treatment of YB1 injections, the cured tumors experienced three distinct phases of the immune response. Two days after injection, the innate immune response was activated, particularly the complement and blood coagulation pathways. In the meantime, the phagocytosis was initiated. The professional phagocytes such as macrophages and neutrophils were recruited, especially the infiltration of iNOS+ and CD68+ cells was enhanced. Seven days after injection, substantial amount of T cells was observed at the invasion margin of the tumor. As a result, the tumor shrunk significantly. Overall, the temporal proteome profiling can systematically reveal the YB1 induced immune responses in tumor, showing great promise for elucidating the mechanism of bacteria-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Shuxin Yang
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wenjuan Zhao
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Muchun Zhu
- Guangdong-Hong Kong-Macao Joint Laboratory of Human-Machine Intelligence-Synergy Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huijuan Hu
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weijie Wang
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhongsheng Zang
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Meiling Jin
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jiacheng Bi
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jiandong Huang
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chenli Liu
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xuefei Li
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Peng Yin
- Guangdong-Hong Kong-Macao Joint Laboratory of Human-Machine Intelligence-Synergy Systems, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Nan Li
- Chinese Academy of Sciences (CAS) Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
66
|
Long Term Response to Circulating Angiogenic Cells, Unstimulated or Atherosclerotic Pre-Conditioned, in Critical Limb Ischemic Mice. Biomedicines 2021; 9:biomedicines9091147. [PMID: 34572333 PMCID: PMC8469527 DOI: 10.3390/biomedicines9091147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 01/05/2023] Open
Abstract
Critical limb ischemia (CLI), the most severe form of peripheral artery disease, results from the blockade of peripheral vessels, usually correlated to atherosclerosis. Currently, endovascular and surgical revascularization strategies cannot be applied to all patients due to related comorbidities, and even so, most patients require re-intervention or amputation within a year. Circulating angiogenic cells (CACs) constitute a good alternative as CLI cell therapy due to their vascular regenerative potential, although the mechanisms of action of these cells, as well as their response to pathological conditions, remain unclear. Previously, we have shown that CACs enhance angiogenesis/arteriogenesis from the first days of administration in CLI mice. Also, the incubation ex vivo of these cells with factors secreted by atherosclerotic plaques promotes their activation and mobilization. Herein, we have evaluated the long-term effect of CACs administration in CLI mice, whether pre-stimulated or not with atherosclerotic factors. Remarkably, mice receiving CACs and moreover, pre-stimulated CACs, presented the highest blood flow recovery, lower progression of ischemic symptoms, and decrease of immune cells recruitment. In addition, many proteins potentially involved, like CD44 or matrix metalloproteinase 9 (MMP9), up-regulated in response to ischemia and decreased after CACs administration, were identified by a quantitative proteomics approach. Overall, our data suggest that pre-stimulation of CACs with atherosclerotic factors might potentiate the regenerative properties of these cells in vivo.
Collapse
|
67
|
Khoobchandani M, Khan A, Katti KK, Thipe VC, Al-Yasiri AY, MohanDoss DKD, Nicholl MB, Lugão AB, Hans CP, Katti KV. Green nanotechnology of MGF-AuNPs for immunomodulatory intervention in prostate cancer therapy. Sci Rep 2021; 11:16797. [PMID: 34408231 PMCID: PMC8373987 DOI: 10.1038/s41598-021-96224-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Men with castration-resistant prostate cancer (CRPC) face poor prognosis and increased risk of treatment-incurred adverse effects resulting in one of the highest mortalities among patient population globally. Immune cells act as double-edged sword depending on the tumor microenvironment, which leads to increased infiltration of pro-tumor (M2) macrophages. Development of new immunomodulatory therapeutic agents capable of targeting the tumor microenvironment, and hence orchestrating the transformation of pro-tumor M2 macrophages to anti-tumor M1, would substantially improve treatment outcomes of CRPC patients. We report, herein, Mangiferin functionalized gold nanoparticulate agent (MGF-AuNPs) and its immunomodulatory characteristics in treating prostate cancer. We provide evidence of immunomodulatory intervention of MGF-AuNPs in prostate cancers through observations of enhanced levels of anti-tumor cytokines (IL-12 and TNF-α) with concomitant reductions in the levels of pro-tumor cytokines (IL-10 and IL-6). In the MGF-AuNPs treated groups, IL-12 was elevated to ten-fold while TNF-α was elevated to about 50-fold, while IL-10 and IL-6 were reduced by two-fold. Ability of MGF-AuNPs to target splenic macrophages is invoked via targeting of NF-kB signaling pathway. Finally, therapeutic efficacy of MGF-AuNPs, in treating prostate cancer in vivo in tumor bearing mice, is described taking into consideration various immunomodulatory interventions triggered by this green nanotechnology-based nanomedicine agent.
Collapse
Affiliation(s)
- Menka Khoobchandani
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO, 65212, USA
- Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Ave, St. Louis, MO, 63108, USA
| | - Aslam Khan
- Department of Biochemistry, University of Missouri, Columbia, MO, 65212, USA
| | - Kavita K Katti
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO, 65212, USA
| | - Velaphi C Thipe
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear, IPEN/CNEN-SP, Butantã, São Paulo, SP, Brasil
| | - Amal Y Al-Yasiri
- Nuclear Science and Engineering Institute (NSEI), University of Missouri, Columbia, MO, 65211, USA
- College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - Darsha K D MohanDoss
- Dhanvantari Nano Ayushadi Pvt Ltd, No. 8/34, Neelakanta Mehta Street, T. Nagar, Chennai, 600017, India
| | | | - Ademar B Lugão
- Laboratório de Ecotoxicologia, Centro de Química e Meio Ambiente, Instituto de Pesquisas Energéticas e Nucleares (IPEN), Comissão Nacional de Energia Nuclear, IPEN/CNEN-SP, Butantã, São Paulo, SP, Brasil
| | - Chetan P Hans
- Department of Medicine-Cardiology, University of Missouri, Columbia, MO, 65212, USA
| | - Kattesh V Katti
- Department of Radiology, Institute of Green Nanotechnology, University of Missouri, Columbia, MO, 65212, USA.
- Department of Physics, University of Missouri, Columbia, MO, 65212, USA.
- University of Missouri Research Reactor (MURR), University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
68
|
Moeini P, Niedźwiedzka-Rystwej P. Tumor-Associated Macrophages: Combination of Therapies, the Approach to Improve Cancer Treatment. Int J Mol Sci 2021; 22:ijms22137239. [PMID: 34281293 PMCID: PMC8269174 DOI: 10.3390/ijms22137239] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Macrophages are one of the most important cells of the innate immune system and are known for their ability to engulf and digest foreign substances, including cellular debris and tumor cells. They can convert into tumor-associated macrophages (TAMs) when mature macrophages are recruited into the tumor microenvironment. Their role in cancer progression, metastasis, and therapy failure is of special note. The aim of this review is to understand how the presence of TAMs are both advantageous and disadvantageous in the immune system.
Collapse
Affiliation(s)
- Pedram Moeini
- Plant Virology Research Center, Shiraz University, Shiraz 71441-65186, Iran;
| | | |
Collapse
|
69
|
Lavy M, Gauttier V, Poirier N, Barillé-Nion S, Blanquart C. Specialized Pro-Resolving Mediators Mitigate Cancer-Related Inflammation: Role of Tumor-Associated Macrophages and Therapeutic Opportunities. Front Immunol 2021; 12:702785. [PMID: 34276698 PMCID: PMC8278519 DOI: 10.3389/fimmu.2021.702785] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammation is a fundamental physiological response orchestrated by innate immune cells to restore tissue homeostasis. Specialized pro-resolving mediators (SPMs) are involved in active resolution of inflammation but when inflammation is incomplete, chronic inflammation creates a favorable environment that fuels carcinogenesis and cancer progression. Conventional cancer therapy also strengthens cancer-related inflammation by inducing massive tumor cell death that activate surrounding immune-infiltrating cells such as tumor-associated macrophages (TAMs). Macrophages are key actors of both inflammation and its active resolution due to their plastic phenotype. In line with this high plasticity, macrophages can be hijacked by cancer cells to support tumor progression and immune escape, or therapy resistance. Impaired resolution of cancer-associated inflammation supported by TAMs may thus reinforces tumor progression. From this perspective, recent evidence suggests that stimulating macrophage's pro-resolving functions using SPMs can promote inflammation resolution in cancer and improve anticancer treatments. Thus, TAMs' re-education toward an antitumor phenotype by using SPMs opens a new line of attack in cancer treatment. Here, we review SPMs' anticancer capacities with special attention regarding their effects on TAMs. We further discuss how this new therapeutic approach could be envisioned in cancer therapy.
Collapse
|
70
|
Orsi M, Palmai-Pallag M, Yakoub Y, Ibouraadaten S, De Beukelaer M, Bouzin C, Bearzatto B, Ambroise J, Gala JL, Brusa D, Lison D, Huaux F. Monocytic Ontogeny of Regenerated Macrophages Characterizes the Mesotheliomagenic Responses to Carbon Nanotubes. Front Immunol 2021; 12:666107. [PMID: 34194430 PMCID: PMC8236701 DOI: 10.3389/fimmu.2021.666107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/28/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages are not only derived from circulating blood monocytes or embryonic precursors but also expand by proliferation. The origin determines macrophage fate and functions in steady state and pathological conditions. Macrophages predominantly infiltrate fibre-induced mesothelioma tumors and contribute to cancer development. Here, we revealed their ontogeny by comparing the response to needle-like mesotheliomagenic carbon nanotubes (CNT-7) with tangled-like non-mesotheliomagenic CNT-T. In a rat peritoneal cavity model of mesothelioma, both CNT induced a rapid macrophage disappearance reaction (MDR) of MHCIIlow resident macrophages generating an empty niche available for macrophage repopulation. Macrophage depletion after mesotheliomagenic CNT-7 was followed by a substantial inflammatory reaction, and macrophage replenishment completed after 7 days. Thirty days after non-mesotheliomagenic CNT-T, macrophage repopulation was still incomplete and accompanied by a limited inflammatory reaction. Cell depletion experiments, flow cytometry and RNA-seq analysis demonstrated that, after mesotheliomagenic CNT-7 exposure, resident macrophages were mainly replaced by an influx of monocytes, which differentiated locally into MHCIIhigh inflammatory macrophages. In contrast, the low inflammatory response induced by CNT-T was associated by the accumulation of self-renewing MHCIIlow macrophages that initially derive from monocytes. In conclusion, the mesotheliomagenic response to CNT specifically relies on macrophage niche recolonization by monocyte-derived inflammatory macrophages. In contrast, the apparent homeostasis after non-mesotheliomagenic CNT treatment involves a macrophage regeneration by proliferation. Macrophage depletion and repopulation are thus decisive events characterizing the carcinogenic activity of particles and fibres.
Collapse
Affiliation(s)
- Micaela Orsi
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Mihaly Palmai-Pallag
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Yousof Yakoub
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Saloua Ibouraadaten
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Michèle De Beukelaer
- Imaging Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Caroline Bouzin
- Imaging Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Bertrand Bearzatto
- Center for Applied Molecular Technologies, Institute of Experimental and Clinical Research (IREC), Cliniques Universitaires Saint-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Jérôme Ambroise
- Center for Applied Molecular Technologies, Institute of Experimental and Clinical Research (IREC), Cliniques Universitaires Saint-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Jean-Luc Gala
- Center for Applied Molecular Technologies, Institute of Experimental and Clinical Research (IREC), Cliniques Universitaires Saint-Luc and Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Davide Brusa
- Flow Cytometry Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Dominique Lison
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
71
|
Fang Y, He Y, Wu C, Zhang M, Gu Z, Zhang J, Liu E, Xu Q, Asrorov AM, Huang Y. Magnetism-mediated targeting hyperthermia-immunotherapy in "cold" tumor with CSF1R inhibitor. Am J Cancer Res 2021; 11:6860-6872. [PMID: 34093858 PMCID: PMC8171105 DOI: 10.7150/thno.57511] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/05/2021] [Indexed: 12/23/2022] Open
Abstract
Background: Immunotherapy has profoundly changed the landscape of cancer management and represented the most significant breakthrough. Yet, it is a formidable challenge that the majority of cancers - the so-called “cold” tumors - poorly respond to immunotherapy. To find a general immunoregulatory modality that can be applied to a broad spectrum of cancers is an urgent need. Methods: Magnetic hyperthermia (MHT) possesses promise in cancer therapy. We develop a safe and effective therapeutic strategy by using magnetism-mediated targeting MHT-immunotherapy in “cold” colon cancer. A magnetic liposomal system modified with cell-penetrating TAT peptide was developed for targeted delivery of a CSF1R inhibitor (BLZ945), which can block the CSF1-CSF1R pathway and reduce M2 macrophages. The targeted delivery strategy is characterized by its magnetic navigation and TAT-promoting intratumoral penetration. Results: The liposomes (termed TAT-BLZmlips) can induce ICD and cause excessive CRT exposure on the cell surface, which transmits an “eat-me” signal to DCs to elicit immunity. The combination of MHT and BLZ945 can repolarize M2 macrophages in the tumor microenvironment to relieve immunosuppression, normalize the tumor blood vessels, and promote T-lymphocyte infiltration. The antitumor effector CD8+ T cells were increased after treatment. Conclusion: This work demonstrated that TAT-BLZmlips with magnetic navigation and MHT can remodel tumor microenvironment and activate immune responses and memory, thus inhibiting tumor growth and recurrence.
Collapse
|
72
|
Liu X, Zheng S, Peng Y, Zhuang J, Yang Y, Xu Y, Guan G. Construction of the Prediction Model for Locally Advanced Rectal Cancer Following Neoadjuvant Chemoradiotherapy Based on Pretreatment Tumor-Infiltrating Macrophage-Associated Biomarkers. Onco Targets Ther 2021; 14:2599-2610. [PMID: 33880038 PMCID: PMC8053511 DOI: 10.2147/ott.s297263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/06/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose To assess the value of macrophage-related biomarkers (CD163, CD68, MCSF, and CCL2) for predicting the response to neo-chemoradiotherapy (NCRT) and the prognosis of locally advanced rectal cancer (LARC). Methods We enrolled 191 patients who underwent neoadjuvant chemoradiotherapy and radical resection between 2011 and 2015. Tumor tissues were collected before NCRT with a colonoscope and post-surgery and were subjected to immunohistochemical analysis. Results The expression levels of macrophage-related biomarkers (CD163, CD68, MCSF, and CCL2) were lower in the pathological complete response (pCR) group when compared with the non-pCR group (all P<0.05). Based on X-tile plots, we divided the tumors in two groups and found that lower pre-NCRT/post-surgical CD163, CD68, MCSF, CCL2 scores correlated with improved DFS. Cox regression analysis demonstrated that pre-NCRT CD163 (HR=1.008, 95% CI 1.003-1.013, P=0.003) and MCSF (HR=2.187, 95% CI 1.343-3.564, P=0.002) scores were independent predictors of DFS. Based on Cox multivariate analysis, we constructed a risk score model with a powerful ability to predict pCR in LARC patients. Moreover, COX regression analysis was performed to explore the role of the risk score in LARC patients. The results demonstrated that tumor size (HR=1.291, P=0.041), worse pathological TNM stage (HR=1.789, P=0.005, and higher risk score (HR=1.084, P<0.001) were significantly associated with impaired disease-free survival. Based on the above results, a nomogram and decision curve analysis were generated. Conclusion The expression levels of macrophage-related biomarkers CD163, CD68, MCSF, and CCL2 were associated with chemoradiotherapy resistance and prognosis in LARC patients following NCRT. A risk score model was constructed which could be used to predict LARC outcome.
Collapse
Affiliation(s)
- Xing Liu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| | - Shuping Zheng
- Public Technology Service Center, Fujian Medical University, Fuzhou, People's Republic of China
| | - Yong Peng
- Department of General Surgery, Nanjing BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jinfu Zhuang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Yuanfeng Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Yunlu Xu
- Public Technology Service Center, Fujian Medical University, Fuzhou, People's Republic of China
| | - Guoxian Guan
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| |
Collapse
|
73
|
Prat M, Salon M, Allain T, Dubreuil O, Noël G, Preisser L, Jean B, Cassard L, Lemée F, Tabah-Fish I, Pipy B, Jeannin P, Prost JF, Barret JM, Coste A. Murlentamab, a Low Fucosylated Anti-Müllerian Hormone Type II Receptor (AMHRII) Antibody, Exhibits Anti-Tumor Activity through Tumor-Associated Macrophage Reprogrammation and T Cell Activation. Cancers (Basel) 2021; 13:cancers13081845. [PMID: 33924378 PMCID: PMC8070390 DOI: 10.3390/cancers13081845] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/19/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs in healthy adults but is also re-expressed in ovarian, colorectal and lung cancers. In this context, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody, currently in clinical trial. Preliminary data suggest that murlentamab anti-tumor activity involves immune response activation. Thus, in vitro experiments were performed to precisely characterize the murlentamab effect on the human immune system. We show that murlentamab treatment is associated with evidences of innate and adaptive immune cell activation in cancer patient samples. Moreover, we demonstrate that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells promotes a polarization switch of both naïve and tumor-associated macrophages towards an anti-tumor M1-like phenotype. Our work also supports that, through macrophage reeducation, murlentamab activates an anti-tumor adaptive immune response. Finally, the combination of murlentamab with pembrolizumab confirmed novel clinical perspectives of murlentamab association with checkpoint inhibitors and other immuno-modulators. Abstract AMHRII, the anti-Müllerian hormone receptor, is selectively expressed in normal sexual organs but is also re-expressed in gynecologic cancers. Hence, we developed murlentamab, a humanized glyco-engineered anti-AMHRII monoclonal antibody currently in clinical trial. Low-fucosylated antibodies are known to increase the antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) potency of effector cells, but some preliminary results suggest a more global murlentamab-dependent activation of the immune system. In this context, we demonstrate here that the murlentamab opsonization of AMHRII-expressing ovarian tumor cells, in the presence of unstimulated- or tumor-associated macrophage (TAM)-like macrophages, significantly promotes macrophage-mediated ADCC and shifts the whole microenvironment towards a pro-inflammatory and anti-tumoral status, thus triggering anti-tumor activity. We also report that murlentamab orients both unstimulated- and TAM-like macrophages to an M1-like phenotype characterized by a strong expression of co-stimulation markers, pro-inflammatory cytokines and chemokines, favoring T cell recruitment and activation. Moreover, we show that murlentamab treatment shifts CD4+ Th1/Th2 balance towards a Th1 response and activates CD8+ T cells. Altogether, these results suggest that murlentamab, through naïve macrophage orientation and TAM reprogrammation, stimulates the anti-tumor adaptive immune response. Those mechanisms might contribute to the sustained clinical benefit observed in advanced cancer patients treated with murlentamab. Finally, the enhanced murlentamab activity in combination with pembrolizumab opens new therapeutic perspectives.
Collapse
Affiliation(s)
- Mélissa Prat
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
| | - Marie Salon
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
| | - Thibault Allain
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
| | - Olivier Dubreuil
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Grégory Noël
- Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium;
| | - Laurence Preisser
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, 49000 Angers, France; (L.P.); (P.J.)
| | - Bérangère Jean
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Lydie Cassard
- Laboratory of Immunomonitoring in Oncology, Gustave Roussy, 94905 Villejuif, France;
| | - Fanny Lemée
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Isabelle Tabah-Fish
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Bernard Pipy
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
| | - Pascale Jeannin
- Univ Angers, Université de Nantes, CHU Angers, Inserm, CRCINA, SFR ICAT, 49000 Angers, France; (L.P.); (P.J.)
| | - Jean-François Prost
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Jean-Marc Barret
- GamaMabs Pharma, 31106 Toulouse, France; (O.D.); (B.J.); (F.L.); (J.-F.P.); (J.-M.B.)
| | - Agnès Coste
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31062 Toulouse, France; (M.P.); (M.S.); (T.A.); (B.P.)
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, UPS, 31100 Toulouse, France
- Correspondence: ; Tel.: +33-534-609-501
| |
Collapse
|
74
|
Li Z, Zhao R, Yang W, Li C, Huang J, Wen Z, Du G, Jiang L. PLCG2 as a potential indicator of tumor microenvironment remodeling in soft tissue sarcoma. Medicine (Baltimore) 2021; 100:e25008. [PMID: 33725976 PMCID: PMC7982206 DOI: 10.1097/md.0000000000025008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/11/2021] [Indexed: 01/05/2023] Open
Abstract
The tumor microenvironment (TME) plays an important role in the occurrence and development of soft tissue sarcoma (STS). A number of studies have shown that to inhibit tumor growth, the TME can be remodeled into an environment unsuitable for tumor proliferation. However, a lack of understanding exists regarding the dynamic regulation of TME.In this study, we used CIBERSORT and ESTIMATE calculation methods from the Cancer Genome Atlas (TCGA) database to calculate the proportion of tumor infiltrating immune cells (TICs) and the number of immune and stromal components in 263 STS samples. Differential expression genes (DEGs) shared by Immune Score and Stromal Score were obtained via difference analysis. Univariate Cox regression analysis and construction of protein-protein interaction (PPI) networks were applied to the DEGs.Through intersection analysis of univariate COX and PPI, PLCG2 was determined as the indicator. Further analysis showed that PLCG2 expression was positively correlated with the survival of STS patients. Gene set enrichment analysis (GSEA) showed that genes in the highly expressed PLCG2 group were enriched in immune-related activities. In the low-expression PLCG2 group, genes were enriched in the E2F, G2M, and MYC pathways. Difference analysis and correlation analysis showed that CD8+ T cells, gamma delta T cells, monocytes, and M1 macrophages were positively correlated with PLCG2 expression, indicating that PLCG2 may represent the immune status of TME.Therefore, the level of PLCG2 may aid in determining the prognosis of STS patients, especially the status of TME. These data provide additional insights into the remodeling of TME.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gang Du
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning
| | - Lingling Jiang
- Department of Anesthesiology, The second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
75
|
Blondy T, d'Almeida SM, Briolay T, Tabiasco J, Meiller C, Chéné AL, Cellerin L, Deshayes S, Delneste Y, Fonteneau JF, Boisgerault N, Bennouna J, Grégoire M, Jean D, Blanquart C. Involvement of the M-CSF/IL-34/CSF-1R pathway in malignant pleural mesothelioma. J Immunother Cancer 2021; 8:jitc-2019-000182. [PMID: 32581053 PMCID: PMC7319783 DOI: 10.1136/jitc-2019-000182] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Background Malignant pleural mesothelioma (MPM) is a rare and aggressive cancer related to asbestos exposure. The tumor microenvironment content, particularly the presence of macrophages, was described as crucial for the development of the disease. This work aimed at studying the involvement of the M-CSF (CSF-1)/IL-34/CSF-1R pathway in the formation of macrophages in MPM, using samples from patients. Methods Pleural effusions (PEs), frozen tumors, primary MPM cells and MPM cell lines used in this study belong to biocollections associated with clinical databases. Cytokine expressions were studied using real-time PCR and ELISA. The Cancer Genome Atlas database was used to confirm our results on an independent cohort. An original three-dimensional (3D) coculture model including MPM cells, monocytes from healthy donors and a tumor antigen-specific cytotoxic CD8 T cell clone was used. Results We observed that high interleukin (IL)-34 levels in PE were significantly associated with a shorter survival of patients. In tumors, expression of CSF1 was correlated with ‘M2-like macrophages’ markers, whereas this was not the case with IL34 expression, suggesting two distinct modes of action of these cytokines. Expression of IL34 was higher in MPM cells compared with primary mesothelial cells. Particularly, high expression of IL34 was observed in MPM cells with an alteration of CDKN2A. Finally, using 3D coculture model, we demonstrated the direct involvement of MPM cells in the formation of immunosuppressive macrophages, through activation of the colony stimulating factor-1 receptor (CSF1-R) pathway, causing the inhibition of cytotoxicity of tumor antigen-specific CD8+ T cells. Conclusions The M-CSF/IL-34/CSF-1R pathway seems strongly implicated in MPM and could constitute a therapeutic target to act on immunosuppression and to support immunotherapeutic strategies.
Collapse
Affiliation(s)
- Thibaut Blondy
- Université de Nantes, CNRS, INSERM, CRCINA, F-44000 Nantes, France
| | - Sènan Mickael d'Almeida
- Université d'Angers, INSERM, CRCINA, F-49000 Angers, France.,Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland.,Flow Cytometry Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tina Briolay
- Université de Nantes, CNRS, INSERM, CRCINA, F-44000 Nantes, France
| | - Julie Tabiasco
- Université d'Angers, INSERM, CRCINA, F-49000 Angers, France
| | - Clément Meiller
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors, F-75006, Paris, France
| | - Anne-Laure Chéné
- Université de Nantes, CNRS, INSERM, CRCINA, F-44000 Nantes, France.,Service d'Oncologie Médicale Thoracique et Digestive, Hopital Nord Laennec, Nantes, Pays de la Loire, France
| | - Laurent Cellerin
- Université de Nantes, CNRS, INSERM, CRCINA, F-44000 Nantes, France.,Service d'Oncologie Médicale Thoracique et Digestive, Hopital Nord Laennec, Nantes, Pays de la Loire, France
| | - Sophie Deshayes
- Université de Nantes, CNRS, INSERM, CRCINA, F-44000 Nantes, France
| | - Yves Delneste
- Université d'Angers, INSERM, CRCINA, F-49000 Angers, France.,CHU Angers, Laboratoire d'Immunologie et Allergologie, F-49000 Angers, France
| | | | | | - Jaafar Bennouna
- Université de Nantes, CNRS, INSERM, CRCINA, F-44000 Nantes, France.,CHU de Nantes, oncologie thoracique et oncologie digestive, 5, allée de l'Île Gloriette, 44093 Nantes, France
| | - Marc Grégoire
- Université de Nantes, CNRS, INSERM, CRCINA, F-44000 Nantes, France
| | - Didier Jean
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université de Paris, Functional Genomics of Solid Tumors, F-75006, Paris, France
| | | |
Collapse
|
76
|
Ang AD, Vissers MCM, Burgess ER, Currie MJ, Dachs GU. Gene and Protein Expression Is Altered by Ascorbate Availability in Murine Macrophages Cultured under Tumour-Like Conditions. Antioxidants (Basel) 2021; 10:antiox10030430. [PMID: 33799728 PMCID: PMC7998289 DOI: 10.3390/antiox10030430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 01/01/2023] Open
Abstract
Tumour-associated macrophages (TAMs) are ubiquitously present in tumours and commonly associated with poor prognosis. In immune cells, ascorbate affects epigenetic regulation, differentiation and phenotype via its co-factor activity for the 2-oxoglutarate dependent dioxygenase enzymes. Here, we determined the effect of ascorbate on TAM development in response to tumour microenvironmental cues. Naïve murine bone marrow monocytes were cultured with Lewis Lung Carcinoma conditioned media (LLCM) or macrophage colony-stimulating factor (MCSF) to encourage the development into tumour-associated macrophages. Cells were stimulated with hypoxia (1% O2), with or without ascorbate (500 µM) supplementation. Cells and media were harvested for gene, cell surface marker and protein analyses. LLCM supported bone marrow monocyte growth with >90% of cells staining CD11b+F4/80+, indicative of monocytes/macrophages. LLCM-grown cells showed increased expression of M2-like and TAM genes compared to MCSF-grown cells, which further increased with hypoxia. In LLCM-grown cells, ascorbate supplementation was associated with increased F4/80 cell surface expression, and altered gene expression and protein secretion. Our study shows that ascorbate modifies monocyte phenotype when grown under tumour microenvironmental conditions, but this was not clearly associated with either a pro- or anti-tumour phenotype, and reflects a complex and nuanced response of macrophages to ascorbate. Overall, ascorbate supplementation clearly has molecular consequences for TAMs, but functional and clinical consequences remain unknown.
Collapse
Affiliation(s)
- Abel D. Ang
- Mackenzie Cancer Research Group, Department of Pathology & Biomedical Science, University of Otago Christchurch, Christchurch 8140, New Zealand; (A.D.A.); (E.R.B.); (M.J.C.)
| | - Margreet C. M. Vissers
- Centre for Free Radical Research, Department of Pathology & Biomedical Science, University of Otago Christchurch, Christchurch 8140, New Zealand;
| | - Eleanor R. Burgess
- Mackenzie Cancer Research Group, Department of Pathology & Biomedical Science, University of Otago Christchurch, Christchurch 8140, New Zealand; (A.D.A.); (E.R.B.); (M.J.C.)
| | - Margaret J. Currie
- Mackenzie Cancer Research Group, Department of Pathology & Biomedical Science, University of Otago Christchurch, Christchurch 8140, New Zealand; (A.D.A.); (E.R.B.); (M.J.C.)
| | - Gabi U. Dachs
- Mackenzie Cancer Research Group, Department of Pathology & Biomedical Science, University of Otago Christchurch, Christchurch 8140, New Zealand; (A.D.A.); (E.R.B.); (M.J.C.)
- Correspondence:
| |
Collapse
|
77
|
Targeting Innate Immunity in Cancer Therapy. Vaccines (Basel) 2021; 9:vaccines9020138. [PMID: 33572196 PMCID: PMC7916062 DOI: 10.3390/vaccines9020138] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 12/21/2022] Open
Abstract
The majority of current cancer immunotherapy strategies target and potentiate antitumor adaptive immune responses. Unfortunately, the efficacy of these treatments has been limited to a fraction of patients within a subset of tumor types, with an aggregate response rate of approximately 20% to date across all malignancies. The success of therapeutic inhibition of programmed death protein 1 (PD-1), protein death ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) with immune checkpoint inhibitors (ICI) has been limited to “hot” tumors characterized by preexisting T cell infiltration, whereas “cold” tumors, which lack T cell infiltration, have not achieved durable benefit. There are several mechanisms by which “cold” tumors fail to generate spontaneous immune infiltration, which converge upon the generation of an immunosuppressive tumor microenvironment (TME). The role of the innate immune system in tumor immunosurveillance and generation of antitumor immune responses has been long recognized. In recent years, novel strategies to target innate immunity in cancer therapy have emerged, including therapeutic stimulation of pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs); the DNA sensing cGAS/STING pathway; nucleotide-binding oligomerization domain-like receptors (NLRs), such as NLRP3; and the retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs). In addition, therapeutic modulation of key innate immune cell types, such as macrophages and natural killer cells, has been investigated. Herein, we review therapeutic approaches to activate innate immunity within the TME to enhance antitumor immune responses, with the goal of disease eradication in “cold” tumors. In addition, we discuss rational immune-oncology combination strategies that activate both innate and adaptive immunity, with the potential to enhance the efficacy of current immunotherapeutic approaches.
Collapse
|
78
|
Choi YW, Kim YH, Oh SY, Suh KW, Kim Y, Lee G, Yoon JE, Park SS, Lee Y, Park YJ, Kim HS, Park SH, Kim J, Park TJ. Senescent Tumor Cells Build a Cytokine Shield in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002497. [PMID: 33643790 PMCID: PMC7887594 DOI: 10.1002/advs.202002497] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/09/2020] [Indexed: 05/25/2023]
Abstract
Cellular senescence can either support or inhibit cancer progression. Here, it is shown that intratumoral infiltration of CD8+ T cells is negatively associated with the proportion of senescent tumor cells in colorectal cancer (CRC). Gene expression analysis reveals increased expression of C-X-C motif chemokine ligand 12 (CXCL12) and colony stimulating factor 1 (CSF1) in senescent tumor cells. Senescent tumor cells inhibit CD8+ T cell infiltration by secreting a high concentration of CXCL12, which induces a loss of CXCR4 in T cells that result in impaired directional migration. CSF1 from senescent tumor cells enhance monocyte differentiation into M2 macrophages, which inhibit CD8+ T cell activation. Neutralization of CXCL12/CSF1 increases the effect of anti-PD1 antibody in allograft tumors. Furthermore, inhibition of CXCL12 from senescent tumor cells enhances T cell infiltration and results in reducing the number and size of tumors in azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced CRC. These findings suggest senescent tumor cells generate a cytokine barrier protecting nonsenescent tumor cells from immune attack and provide a new target for overcoming the immunotherapy resistance of CRC.
Collapse
Affiliation(s)
- Yong Won Choi
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwon16499Korea
- Department of Hematology–OncologyAjou University School of MedicineSuwon16499Korea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwon16499Korea
| | - Young Hwa Kim
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwon16499Korea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwon16499Korea
| | - Seung Yeop Oh
- Department of SurgeryAjou University School of MedicineSuwon16499Korea
| | - Kwang Wook Suh
- Department of SurgeryAjou University School of MedicineSuwon16499Korea
| | - Young‐Sam Kim
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwon16499Korea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwon16499Korea
| | - Ga‐Yeon Lee
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwon16499Korea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwon16499Korea
| | - Jung Eun Yoon
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwon16499Korea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwon16499Korea
| | - Soon Sang Park
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwon16499Korea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwon16499Korea
| | - Young‐Kyoung Lee
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwon16499Korea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwon16499Korea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwon16499Korea
| | - Yoo Jung Park
- Department of Hematology–OncologyAjou University School of MedicineSuwon16499Korea
| | - Hong Seok Kim
- Department of Molecular MedicineInha University School of MedicineIncheon22212Korea
| | - So Hyun Park
- Department of PathologyAjou University School of MedicineSuwon16499Korea
| | - Jang‐Hee Kim
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwon16499Korea
- Department of PathologyAjou University School of MedicineSuwon16499Korea
| | - Tae Jun Park
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwon16499Korea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwon16499Korea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwon16499Korea
| |
Collapse
|
79
|
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: 33.3] [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
|
80
|
Soyama T, Sakuragi A, Oishi D, Kimura Y, Aoki H, Nomoto A, Yano S, Nishie H, Kataoka H, Aoyama M. Photodynamic therapy exploiting the anti-tumor activity of mannose-conjugated chlorin e6 reduced M2-like tumor-associated macrophages. Transl Oncol 2021; 14:101005. [PMID: 33401079 PMCID: PMC7785959 DOI: 10.1016/j.tranon.2020.101005] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 02/06/2023] Open
Abstract
M2-like tumor-associated macrophages (M2-TAMs) in cancer tissues are intimately involved in cancer immunosuppression in addition to growth, invasion, angiogenesis, and metastasis. Hence, considerable attention has been focused on cancer immunotherapies targeting M2-TAMs. However, systemic therapies inhibit TAMs as well as other macrophages important for normal immune responses throughout the body. To stimulate tumor immunity with fewer side effects, we targeted M2-TAMs using photodynamic therapy (PDT), which damages cells via a nontoxic photosensitizer with harmless laser irradiation. We synthesized a light-sensitive compound, mannose-conjugated chlorin e6 (M-chlorin e6), which targets mannose receptors highly expressed on M2-TAMs. M-chlorin e6 accumulated more in tumor tissue than normal skin tissue of syngeneic model mice and was more rapidly excreted than the second-generation photosensitizer talaporfin sodium. Furthermore, M-chlorin e6 PDT significantly reduced the volume and weight of tumor tissue. Flow cytometric analysis revealed that M-chlorin e6 PDT decreased the proportion of M2-TAMs and increased that of anti-tumor macrophages, M1-like TAMs. M-chlorin e6 PDT also directly damaged and killed cancer cells in vitro. Our data indicate that M-chlorin e6 is a promising new therapeutic agent for cancer PDT.
Collapse
Affiliation(s)
- Tatsuki Soyama
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| | - Akira Sakuragi
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| | - Daisuke Oishi
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| | - Yuka Kimura
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| | - Hiromasa Aoki
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan
| | - Akihiro Nomoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Shigenobu Yano
- KYOUSEI Science Center for Life and Nature, Nara Women's University, Kitauoya-Higashimachi, Nara 630-8506, Japan
| | - Hirotada Nishie
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Nagoya 467-8601, Japan
| | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Nagoya 467-8601, Japan
| | - Mineyoshi Aoyama
- Department of Pathobiology, Nagoya City University Graduate School of Pharmaceutical Sciences, 3-1 Tanabe-dori, Mizuho-ku, Nagoya 467-8603, Japan.
| |
Collapse
|
81
|
Muñoz-Garcia J, Cochonneau D, Télétchéa S, Moranton E, Lanoe D, Brion R, Lézot F, Heymann MF, Heymann D. The twin cytokines interleukin-34 and CSF-1: masterful conductors of macrophage homeostasis. Theranostics 2021; 11:1568-1593. [PMID: 33408768 PMCID: PMC7778581 DOI: 10.7150/thno.50683] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/03/2020] [Indexed: 12/19/2022] Open
Abstract
Macrophages are specialized cells that control tissue homeostasis. They include non-resident and tissue-resident macrophage populations which are characterized by the expression of particular cell surface markers and the secretion of molecules with a wide range of biological functions. The differentiation and polarization of macrophages relies on specific growth factors and their receptors. Macrophage-colony stimulating factor (CSF-1) and interleukine-34 (IL-34), also known as "twin" cytokines, are part of this regluatory landscape. CSF-1 and IL-34 share a common receptor, the macrophage-colony stimulating factor receptor (CSF-1R), which is activated in a similar way by both factors and turns on identical signaling pathways. However, there is some discrete differential activation leading to specific activities. In this review, we disscuss recent progress in understanding of the role of the twin cytokines in macrophage differentiation, from their interaction with CSF-1R and the activation of signaling pathways, to their implication in macrophage polarization of non-resident and tissue-resident macrophages. A special focus on IL-34, its involvement in pathophsyiological contexts, and its potential as a theranostic target for macrophage therapy will be proposed.
Collapse
Affiliation(s)
- Javier Muñoz-Garcia
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
- SATT Ouest Valorisation, Nantes, France
| | - Denis Cochonneau
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
| | | | - Emilie Moranton
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
| | - Didier Lanoe
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
| | - Régis Brion
- Université de Nantes, INSERM, U1238, Nantes, France
| | | | | | - Dominique Heymann
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| |
Collapse
|
82
|
Almahariq MF, Quinn TJ, Kesarwani P, Kant S, Miller CR, Chinnaiyan P. Inhibition of Colony-Stimulating Factor-1 Receptor Enhances the Efficacy of Radiotherapy and Reduces Immune Suppression in Glioblastoma. In Vivo 2021; 35:119-129. [PMID: 33402457 PMCID: PMC7880776 DOI: 10.21873/invivo.12239] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/17/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022]
Abstract
AIM To use inhibition of colony-stimulating factor-1 receptor (CSF-1R) to target tumor-associated macrophages (TAMs) and improve the efficacy of radiotherapy in glioblastoma (GBM). MATERIALS AND METHODS The CSF-1R inhibitor BLZ-945 was used to examine the impact of CSF-1R inhibition on M2 polarization in vitro. Using an orthotopic, immunocompetent GBM model, mice were treated with vehicle, RT, BLZ-945, or RT plus BLZ-945. RESULTS BLZ-945 reduced M2 polarization in vitro. BLZ-945 alone did not improve median overall survival (mOS=29 days) compared to control mice (mOS=27 days). RT improved survival (mOS=45 days; p=0.02), while RT plus BLZ-945 led to the longest survival (mOS=not reached; p=0.005). Resected tumors had a relatively large population of M2 TAMs in GBM at baseline, which was increased in response to RT. BLZ-945 reduced RT-induced M2 infiltration. CONCLUSION Inhibition of CSF-1R improved response to RT in the treatment of GBM and may represent a promising strategy to improve RT-induced antitumor immune responses.
Collapse
Affiliation(s)
| | - Thomas J Quinn
- Department of Radiation Oncology, Beaumont Health, Royal Oak, MI, U.S.A
| | - Pravin Kesarwani
- Department of Radiation Oncology, Beaumont Health, Royal Oak, MI, U.S.A
| | - Shiva Kant
- Department of Radiation Oncology, Beaumont Health, Royal Oak, MI, U.S.A
| | - C Ryan Miller
- Division of Neuropathology, Department of Pathology, O'Neal Comprehensive Cancer Center, Comprehensive Neuroscience Center, University of Alabama School of Medicine, Birmingham, AL, U.S.A
| | - Prakash Chinnaiyan
- Department of Radiation Oncology, Beaumont Health, Royal Oak, MI, U.S.A.;
- Oakland University William Beaumont School of Medicine, Rochester, MI, U.S.A
| |
Collapse
|
83
|
Correlation between prognostic indicator AHNAK2 and immune infiltrates in lung adenocarcinoma. Int Immunopharmacol 2020; 90:107134. [PMID: 33168407 DOI: 10.1016/j.intimp.2020.107134] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is among the most aggressive malignant tumors in humans. Although AHNAK nucleoprotein 2 (AHNAK2) is considered a new oncogene, the function of the AHNAK2 in LUAD remains unknown. METHODS Oncomine, Tumor Immune Estimation Resource (TIMER), and Human Protein Atlas databases were used to investigate AHNAK2 expression in LUAD. Gene Expression Profiling Interactive Analysis and Kaplan-Meier plotter databases were employed to elucidate the relationship between AHNAK2 and survival time. Data of The Cancer Genome Atlas were downloaded to analyze the correlation between AHNAK2 and clinicopathological parameters. We then immunohistochemically stained tissue chips to further confirm the correlation and conducted Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and protein-protein interaction network analyses to explore the possible functional mechanism of AHNAK2. Finally, we investigated the relationship between AHNAK2 and tumor infiltrating immune cells (TIICs). RESULTS AHNAK2 gene was significantly overexpressed in LUAD tumor tissues and an independent prognostic indicator of LUAD patients. The expression of AHNAK2 was related to disease stage, differentiation, tumor size and lymph node metastasis. We found AHNAK2 expression was mainly positively correlated with cell adhesion-related pathways and negatively correlated with oxidative phosphorylation and amino acid metabolism. AHNAK2 expression was also negatively correlated with activated B cell, activated CD8 + T cell, and immature B cell infiltrates and positively correlated with central memory CD4 + T cell, tumor-associated macrophage, M1 macrophage, and M2 macrophage infiltrates. CONCLUSION Our findings provide strong evidence of AHNAK2 expression as a prognostic indicator related to TIICs in LUAD.
Collapse
|
84
|
Castañeda-Reyes ED, Perea-Flores MDJ, Davila-Ortiz G, Lee Y, Gonzalez de Mejia E. Development, Characterization and Use of Liposomes as Amphipathic Transporters of Bioactive Compounds for Melanoma Treatment and Reduction of Skin Inflammation: A Review. Int J Nanomedicine 2020; 15:7627-7650. [PMID: 33116492 PMCID: PMC7549499 DOI: 10.2147/ijn.s263516] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022] Open
Abstract
The skin is the largest organ in the human body, providing a barrier to the external environment. It is composed of three layers: epidermis, dermis and hypodermis. The most external epidermis is exposed to stress factors that may lead to skin conditions such as photo-aging and skin cancer. Some treatments for skin disease utilize the incorporation of drugs or bioactive compounds into nanocarriers known as liposomes. Liposomes are membranes whose sizes range from nano to micrometers and are composed mostly of phospholipids and cholesterol, forming similar structures to cell membranes. Thus, skin treatments with liposomes have lower toxicity in comparison to traditional treatment routes such as parenteral and oral. Furthermore, addition of edge activators to the liposomes decreases the rigidity of the bilayer structure making it deformable, thereby improving skin permeability. Liposomes are composed of an aqueous core and a lipidic bilayer, which confers their amphiphilic property. Thus, they can carry hydrophobic and hydrophilic compounds, even simultaneously. Current applications of these nanocarriers are mainly in the cosmetic and pharmaceutic industries. Nevertheless, new research has revealed promising results regarding the effectiveness of liposomes for transporting bioactive compounds through the skin. Liposomes have been well studied; however, additional research is needed on the efficacy of liposomes loaded with bioactive peptides for skin delivery. The objective of this review is to provide an up-to-date description of existing techniques for the development of liposomes and their use as transporters of bioactive compounds in skin conditions such as melanoma and skin inflammation. Furthermore, to gain an understanding of the behavior of liposomes during the process of skin delivery of bioactive compounds into skin cells.
Collapse
Affiliation(s)
- Erick Damian Castañeda-Reyes
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Unidad Profesional Adolfo Lopez Mateos, Ciudad De México, 07738, México.,Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Maria de Jesús Perea-Flores
- Centro de Nanociencias y Micro y Nanotecnologías, Instituto Politécnico Nacional (IPN), Unidad Profesional Adolfo López Mateos, Ciudad De México 07738, México
| | - Gloria Davila-Ortiz
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Unidad Profesional Adolfo Lopez Mateos, Ciudad De México, 07738, México
| | - Youngsoo Lee
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Elvira Gonzalez de Mejia
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| |
Collapse
|
85
|
Gregory KJ, Morin SM, Kubosiak A, Ser‐Dolansky J, Schalet BJ, Jerry DJ, Schneider SS. The use of patient-derived breast tissue explants to study macrophage polarization and the effects of environmental chemical exposure. Immunol Cell Biol 2020; 98:883-896. [PMID: 32713010 PMCID: PMC7754397 DOI: 10.1111/imcb.12381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 06/23/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022]
Abstract
Ex vivo mammary explant systems are an excellent model to study interactions between epithelium and stromal cell types because they contain physiologically relevant heterotypic interactions in the background of genetically diverse patients. The intact human mammary tissue, termed patient-derived explant (PDE), can be used to investigate cellular responses to a wide variety of external stimuli in situ. For this study, we examined the impact of cytokines or environmental chemicals on macrophage phenotypes. We demonstrate that we can polarize macrophages within human breast tissue PDEs toward M1 or M2 through the addition of interferon-γ (IFNγ) + lipopolysaccharide (LPS) or interleukin (IL)-4 + IL-13, respectively. Elevated expression levels of M(IFNγ + LPS) markers (HLADRA and CXCL10) or M(IL-4 + IL-13) markers (CD209 and CCL18) were observed in cytokine-treated tissues. We also examined the impact of the endocrine-disrupting chemical, benzophenone-3, on PDEs and measured significant, yet varying effects on macrophage polarization. Furthermore, a subset of the PDEs respond to IL-4 + IL-13 through downregulation of E-cadherin and upregulation of vimentin which is reminiscent of epithelial-to-mesenchymal transition (EMT) changes. Finally, we were able to show immortalized nonmalignant breast epithelial cells can exhibit EMT characteristics when exposed to growth factors secreted by M(IL-4 + IL-13) macrophages. Taken together, the PDE model system is an outstanding preclinical model to study early tissue-resident immune responses and effects on epithelial and stromal responses to stimuli found both endogenously in the breast and exogenously as a result of exposures.
Collapse
Affiliation(s)
- Kelly J Gregory
- Pioneer Valley Life Sciences InstituteSpringfieldMA01199USA
- Biology DepartmentUniversity of MassachusettsAmherstMA01003USA
| | | | | | | | - Benjamin J Schalet
- Department of SurgeryUniversity of Massachusetts Medical School/BaystateSpringfieldMA01199USA
| | - D Joseph Jerry
- Pioneer Valley Life Sciences InstituteSpringfieldMA01199USA
- Veterinary and Animal SciencesUniversity of MassachusettsAmherstMA01003USA
| | - Sallie S Schneider
- Pioneer Valley Life Sciences InstituteSpringfieldMA01199USA
- Veterinary and Animal SciencesUniversity of MassachusettsAmherstMA01003USA
- Department of SurgeryUniversity of Massachusetts Medical School/BaystateSpringfieldMA01199USA
| |
Collapse
|
86
|
Jia Y, Liu L, Shan B. Future of immune checkpoint inhibitors: focus on tumor immune microenvironment. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1095. [PMID: 33145314 PMCID: PMC7575936 DOI: 10.21037/atm-20-3735] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immunotherapy has become a powerful clinical strategy in cancer treatment. Immune checkpoint inhibitors (ICIs) have opened a new era for cancer immunotherapy. Nowadays, the number of immunotherapy drug approvals has increased, with numerous treatment options in clinical and preclinical development. However, there remain some obstacles to improve the efficacy of ICIs further. The tumor immune microenvironment (TIME) consists of cancer cell, immune cells and cytokines, et cetera. The dynamics of TIME determine the efficacies of ICIs. Although the ICIs showed manageable toxicity, immune-related adverse effects (irAEs) are still unignorable for clinicians. Since some primary resistance mechanisms exist in TIME, ICIs can only show effects in individual cancer patients. Even for the patients who responded, acquired resistance will occur to neutralize the effect of ICIs. Understanding how to increase the response rates and overcome the resistance to various classes of ICIs is the key to improving clinical efficacy. Besides the novel ICIs in development, there are some approaches to establish combination therapies are underway to improve further the efficacies of ICIs in treating cancer patients. Here, we describe the complicated TIME and state quo of ICIs to prospect the future of ICIs in cancer treatment.
Collapse
Affiliation(s)
- Yunlong Jia
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Hebei Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Baoen Shan
- Hebei Cancer Institute, Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Research Center, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
87
|
Ye J, Yang Y, Jin J, Ji M, Gao Y, Feng Y, Wang H, Chen X, Liu Y. Targeted delivery of chlorogenic acid by mannosylated liposomes to effectively promote the polarization of TAMs for the treatment of glioblastoma. Bioact Mater 2020; 5:694-708. [PMID: 32478203 PMCID: PMC7248290 DOI: 10.1016/j.bioactmat.2020.05.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor-associated macrophages (TAMs) generally display an immunosuppressive M2 phenotype and promote tumor progression and metastasis, suggesting their potential value as a target in cancer immunotherapy. Chlorogenic acid (CHA) has been identified as a potent immunomodulator that promotes the polarization of TAMs from an M2 to an M1 phenotype. However, rapid clearance in vivo and low tumor accumulation have compromised the immunotherapeutic efficacy of CHA in clinical trials. In this study, mannosylated liposomes are developed for targeted delivery of CHA to TAMs. The immunoregulatory effects of CHA, along with the overall antitumor efficacy of CHA-encapsulated mannosylated liposomes, are investigated through in vitro and in vivo experiments. The prepared CHA-encapsulated mannosylated liposomes exhibit an ideal particle size, favorable stability, and preferential accumulation in tumors via the mannose receptor-mediated TAMs-targeting effects. Further, CHA-encapsulated mannosylated liposomes inhibit G422 glioma tumor growth by efficiently promoting the polarization of the pro-tumorigenic M2 phenotype to the anti-tumorigenic M1 phenotype. Overall, these findings indicate that CHA-encapsulated mannosylated liposomes have great potential to enhance the immunotherapeutic efficacy of CHA by inducing a shift from the M2 to the M1 phenotype.
Collapse
Affiliation(s)
- Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Jing Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Ming Ji
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Yue Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Yu Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| |
Collapse
|
88
|
Zhukova OV, Kovaleva TF, Arkhipova EV, Ryabov SA, Mukhina IV. Tumor-associated macrophages: Role in the pathological process of tumorigenesis and prospective therapeutic use (Review). Biomed Rep 2020; 13:47. [PMID: 32934819 DOI: 10.3892/br.2020.1354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 07/10/2020] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to evaluate the current body of knowledge regarding tumor-associated macrophages (TAMs) and their potential use in antitumor therapy, based on their role in the pathological process of tumorigenesis. For this purpose, a critical analysis of published data and summarization of the findings available from original studies, focusing on the role of TAMs in the pathological process, and their potential therapeutic application was performed. Promising key avenues of research were identified in this field. The following issues seem the most promising and thus worth further investigation: i) The process of M1/M2 macrophage polarization, macrophage characteristics at intermediate polarization steps and their role in the tumor process; ii) determining the conditions necessary for transitions between the M1 and M2 macrophage phenotypes and the role of signals from the microenvironment in this process; iii) cause-and-effect associations between the quantity and quality of macrophages, and the prognosis and outcome of the pathological process; iv) modulation of macrophages and stimulation of their phagocytic activity with drugs; v) targeted vector-based systems for drug delivery to macrophages; and vi) targeted drug delivery systems with macrophages as carriers, thus potentially combining chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Olga V Zhukova
- Department of Pharmaceutical Technology, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Tatiana F Kovaleva
- Department of Molecular and Cellular Technologies, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Evgenia V Arkhipova
- Pre-Clinical Research Center, Central Research Laboratory, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Sergey A Ryabov
- Department of High-Molecular and Colloid Chemistry, National Research Lobachevsky State University, Nizhny Novgorod 603950, Russia
| | - Irina V Mukhina
- Fundamental Medicine Institute and Physiology Department, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| |
Collapse
|
89
|
Wu K, Lin K, Li X, Yuan X, Xu P, Ni P, Xu D. Redefining Tumor-Associated Macrophage Subpopulations and Functions in the Tumor Microenvironment. Front Immunol 2020; 11:1731. [PMID: 32849616 PMCID: PMC7417513 DOI: 10.3389/fimmu.2020.01731] [Citation(s) in RCA: 377] [Impact Index Per Article: 94.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/29/2020] [Indexed: 12/25/2022] Open
Abstract
The immunosuppressive status of the tumor microenvironment (TME) remains poorly defined due to a lack of understanding regarding the function of tumor-associated macrophages (TAMs), which are abundant in the TME. TAMs are crucial drivers of tumor progression, metastasis, and resistance to therapy. Intra- and inter-tumoral spatial heterogeneities are potential keys to understanding the relationships between subpopulations of TAMs and their functions. Antitumor M1-like and pro-tumor M2-like TAMs coexist within tumors, and the opposing effects of these M1/M2 subpopulations on tumors directly impact current strategies to improve antitumor immune responses. Recent studies have found significant differences among monocytes or macrophages from distinct tumors, and other investigations have explored the existence of diverse TAM subsets at the molecular level. In this review, we discuss emerging evidence highlighting the redefinition of TAM subpopulations and functions in the TME and the possibility of separating macrophage subsets with distinct functions into antitumor M1-like and pro-tumor M2-like TAMs during the development of tumors. Such redefinition may relate to the differential cellular origin and monocyte and macrophage plasticity or heterogeneity of TAMs, which all potentially impact macrophage biomarkers and our understanding of how the phenotypes of TAMs are dictated by their ontogeny, activation status, and localization. Therefore, the detailed landscape of TAMs must be deciphered with the integration of new technologies, such as multiplexed immunohistochemistry (mIHC), mass cytometry by time-of-flight (CyTOF), single-cell RNA-seq (scRNA-seq), spatial transcriptomics, and systems biology approaches, for analyses of the TME.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
90
|
Nadella V, Garg M, Kapoor S, Barwal TS, Jain A, Prakash H. Emerging neo adjuvants for harnessing therapeutic potential of M1 tumor associated macrophages (TAM) against solid tumors: Enusage of plasticity. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1029. [PMID: 32953829 PMCID: PMC7475467 DOI: 10.21037/atm-20-695] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Macrophages are a major component of the tumor microenvironment (TME) of most tumors. They are characterized by a high degree of functional plasticity which enable these cells to both promote and eliminate established tumors. Under the influence of immunosuppressive TME, tumor infiltrating iNOS+ and CD11b+ M-1 effector macrophages get polarized towards tumor associated macrophages (TAM) which are tropic to variety of tumors. Increased infiltration and density of TAM is associated with tumor progression and poor prognosis in the plethora of tumors due to their angiogenetic and tissue re-modelling nature. Importantly, TAMs are also responsible for developing endothelium anergy, a major physical barrier for majority of cancer directed immune/chemotherapies. Therefore, functional retuning/re-educating TAM to M-1 phenotypic macrophages is paramount for effective immunotherapy against established tumors. In this review, we discuss and provide comprehensive update on TAM-targeted approaches for enhancing immunity against various solid tumors.
Collapse
Affiliation(s)
- Vinod Nadella
- Laboratory of Translational Medicine, School of Life Sciences, University of Hyderabad, Telangana, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem cell Research, Amity University Uttar Pradesh, Sector 125, Noida, India
| | - Sonia Kapoor
- Amity Institute of Molecular Medicine and Stem cell Research, Amity University Uttar Pradesh, Sector 125, Noida, India
| | | | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bhatinda, India
| | - Hridayesh Prakash
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector 125, Noida, India
| |
Collapse
|
91
|
Shen HY, Zhou Y, Zhou QJ, Li MY, Chen J. Mudskipper interleukin-34 modulates the functions of monocytes/macrophages via the colony-stimulating factor-1 receptor 1. Zool Res 2020; 41:123-137. [PMID: 32150792 PMCID: PMC7109011 DOI: 10.24272/j.issn.2095-8137.2020.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Interleukin-34 (IL-34) is a novel cytokine that plays an important role in innate immunity and inflammatory processes by binding to the colony-stimulating factor-1 receptor (CSF-1R). However, information on the function of IL-34 in fish remains limited. In the present study, we identified an IL-34 homolog from mudskippers (Boleophthalmus pectinirostris). In silico analysis showed that the mudskipper IL-34 (BpIL-34) was similar to other known IL-34 variants in sequence and structure and was most closely related to an orange-spotted grouper (Epinephelus coioides) homolog. BpIL-34 transcripts were constitutively expressed in various tissues, with the highest level of expression found in the brain. Edwardsiella tarda infection significantly up-regulated the mRNA expression of BpIL-34 in the mudskipper tissues. The recombinant mature BpIL-34 peptide (rBpIL-34) was purified and used to produce anti-rBpIL-34 IgG. Western blot analysis combined with PNGase F digestion revealed that native BpIL-34 in monocytes/macrophages (MOs/MФs) was N-glycosylated. In vitro, rBpIL-34 treatment enhanced the phagocytotic and bactericidal activity of mudskipper MOs/MФs, as well as the mRNA expression of pro-inflammatory cytokines like tumor necrosis factor α (BpTNF-α) and BpIL-1β in these cells. Furthermore, the knockdown of mudskipper CSF-1R1 (BpCSF-1R1), but not mudskipper BpCSF-1R2, significantly inhibited the rBpIL-34-mediated enhanced effect on MO/MФ function. In conclusion, our results indicate that mudskipper BpIL-34 modulates the functions of MOs/MФs via BpCSF-1R1.
Collapse
Affiliation(s)
- Hai-Yu Shen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Yan Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Qian-Jin Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China. E-mail: .,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315832, China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Ming-Yun Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang 315832, China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, Zhejiang 315832, China E-mail: jchen1975@ 163.com
| |
Collapse
|
92
|
Zhang X, Li W, Sun J, Yang Z, Guan Q, Wang R, Li X, Li Y, Feng Y, Wang Y. How to use macrophages to realise the treatment of tumour. J Drug Target 2020; 28:1034-1045. [PMID: 32603199 DOI: 10.1080/1061186x.2020.1775236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Macrophages (Mø) are immune cells with natural phagocytic ability and play an important role in tumorigenesis, development and metastasis. Mø play a dual role of tumour inhibition and tumour promotion in tumour development due to their two different phenotypes. Mø in the tumour microenvironment have long been referred to as tumour-associated Mø (TAMs). Mø are mainly involved in tumour resistance, cancer metastasis and mediating immunosuppression. Nowadays, Mø and Mø membranes have been widely used in drug delivery systems (DDSs) because of their good biocompatibility, natural phagocytosis and their important role in tumour development. In this review, from the perspective of Mø's role in tumour development, we present strategies and drugs of Mø targeting and focusing on the several types of biomimetic nanoparticles constructed by Mø and Mø membranes in tumour therapy, and discuss the problem of this delivery system in present research and future directions.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Jialin Sun
- Biological Science and Technology Department, Heilongjiang Vocational College for Nationalities, Harbin, P.R. China
| | - Zhixin Yang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Qingxia Guan
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Rui Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Xiuyan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Yongji Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Yufei Feng
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, People's Republic of China
| |
Collapse
|
93
|
Banu N, Panikar SS, Leal LR, Leal AR. Protective role of ACE2 and its downregulation in SARS-CoV-2 infection leading to Macrophage Activation Syndrome: Therapeutic implications. Life Sci 2020; 256:117905. [PMID: 32504757 PMCID: PMC7832382 DOI: 10.1016/j.lfs.2020.117905] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 02/06/2023]
Abstract
In light of the outbreak of the 2019 novel coronavirus disease (COVID-19), the international scientific community has joined forces to develop effective treatment strategies. The Angiotensin-Converting Enzyme (ACE) 2, is an essential receptor for cell fusion and engulfs the SARS coronavirus infections. ACE2 plays an important physiological role, practically in all the organs and systems. Also, ACE2 exerts protective functions in various models of pathologies with acute and chronic inflammation. While ACE2 downregulation by SARS-CoV-2 spike protein leads to an overactivation of Angiotensin (Ang) II/AT1R axis and the deleterious effects of Ang II may explain the multiorgan dysfunction seen in patients. Specifically, the role of Ang II leading to the appearance of Macrophage Activation Syndrome (MAS) and the cytokine storm in COVID-19 is discussed below. In this review, we summarized the latest research progress in the strategies of treatments that mainly focus on reducing the Ang II-induced deleterious effects rather than attenuating the virus replication. Protective role of ACE2 in the organs and system Downregulation of ACE2 expression by SARS-CoV-2 leads to Ang II-induced organ damage. The appearance of MAS in COVID-19 patient Suggested treatment to diminish the deleterious effect of Ang II or appearance of MAS
Collapse
Affiliation(s)
- Nehla Banu
- Instituto de Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - Sandeep Surendra Panikar
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autonoma de México (UNAM), Apartado Postal 1-1010, Queretaro, Queretaro 76000, Mexico
| | - Lizbeth Riera Leal
- Hospital General Regional número 45, Instituto Mexicano del Seguro Social (IMSS), Guadalajara, Jalisco, Mexico
| | - Annie Riera Leal
- UC DAVIS Institute for Regenerative Cure, Department of Dermatology, University of California, 2921 Stockton Blvd, Rm 1630, 95817 Sacramento, CA, USA.
| |
Collapse
|
94
|
Oronsky B, Carter C, Reid T, Brinkhaus F, Knox SJ. Just eat it: A review of CD47 and SIRP-α antagonism. Semin Oncol 2020; 47:117-124. [PMID: 32517874 DOI: 10.1053/j.seminoncol.2020.05.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/04/2020] [Accepted: 05/04/2020] [Indexed: 01/08/2023]
Abstract
The mammalian immune system consists of two distinct arms, nonspecific innate and more specific adaptive, with the innate immune response as the first line of defense and protection, which primes and amplifies subsequent adaptive responses. On the basis of this binary immune interplay, stimulation of T cells through checkpoint inhibitors (CIs), which bypasses innate involvement, seems likely to engender suboptimal or incomplete anticancer immunity, given that the successful induction of effect or responses depends on two-way innate/adaptive coordination. Indeed, the majority of patients-70%-80%, do not respond to CIs, which is potentially problematic if access to more optimal standard therapies is withheld or delayed in favor of ineffective or only marginally effective anti-PD-1/PD-L1 treatment. Therefore, stimulation of the innate immune response in combination with CIs (or other inducers of T cell cytotoxicity) has the potential to make the immune system "whole" and thereby to enhance and broaden the anti-tumor activity of PD-1/PD-L1 inhibitors for example, in relatively nonimmunogenic or "cold" tumor types. A critical innate macrophage immune checkpoint and druggable target is the antiphagocytic and "marker of self" CD47-SIRPα pathway, which is co-opted by cancer cells to mediate escape from immune-mediated clearance and checkpoint inhibition. This review summarizes the status of key CD47 antagonists in clinical trials, including the biologics, Hu5F9-G4 (5F9), TTI-621, and ALX148, as well as the small molecule, RRx-001, now in a Phase 3 clinical trial, which has not been previously included in CD47-SIRPα reviews focused on biologics. Hu5F9-G4 (5F9), TTI-621, ALX148, and RRx-001 are chosen as compounds with potentially promising data that have advanced the farthest in clinical development.
Collapse
Affiliation(s)
| | | | - Tony Reid
- Department of Medical Oncology, UC San Diego School of Medicine, San Diego, California
| | | | - Susan J Knox
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
95
|
Xie Y, Hu C, Feng Y, Li D, Ai T, Huang Y, Chen X, Huang L, Tan J. Osteoimmunomodulatory effects of biomaterial modification strategies on macrophage polarization and bone regeneration. Regen Biomater 2020; 7:233-245. [PMID: 32523726 PMCID: PMC7266668 DOI: 10.1093/rb/rbaa006] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/02/2020] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Biomaterials as bone substitutes are always considered as foreign bodies that can trigger host immune responses. Traditional designing principles have been always aimed at minimizing the immune reactions by fabricating inert biomaterials. However, clinical evidence revealed that those methods still have limitations and many of which were only feasible in the laboratory. Currently, osteoimmunology, the very pioneering concept is drawing more and more attention-it does not simply regard the immune response as an obstacle during bone healing but emphasizes the intimate relationship of the immune and skeletal system, which includes diverse cells, cytokines, and signaling pathways. Properties of biomaterials like topography, wettability, surface charge, the release of cytokines, mediators, ions and other bioactive molecules can impose effects on immune responses to interfere with the skeletal system. Based on the bone formation mechanisms, the designing methods of the biomaterials change from immune evasive to immune reprogramming. Here, we discuss the osteoimmunomodulatory effects of the new modification strategies-adjusting properties of bone biomaterials to induce a favorable osteoimmune environment. Such strategies showed potential to benefit the development of bone materials and lay a solid foundation for the future clinical application.
Collapse
Affiliation(s)
- Yajuan Xie
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Cheng Hu
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Yi Feng
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Danfeng Li
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Tingting Ai
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Yulei Huang
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Xiaodan Chen
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Lijia Huang
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| | - Jiali Tan
- Guangdong Provincial Key Laboratory of Stomatology, Department of Orthodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, P. R. China
| |
Collapse
|
96
|
Grégoire H, Roncali L, Rousseau A, Chérel M, Delneste Y, Jeannin P, Hindré F, Garcion E. Targeting Tumor Associated Macrophages to Overcome Conventional Treatment Resistance in Glioblastoma. Front Pharmacol 2020; 11:368. [PMID: 32322199 PMCID: PMC7158850 DOI: 10.3389/fphar.2020.00368] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/10/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GB) is the most common and devastating form of brain cancer. Despite conventional treatments, progression or recurrences are systematic. In recent years, immunotherapies have emerged as an effective treatment in a number of cancers, leaving the question of their usefulness also faced with the particular case of brain tumors. The challenge here is major not only because the brain is the seat of our consciousness but also because of its isolation by the blood-brain barrier and the presence of a unique microenvironment that constitutes the central nervous system (CNS) with very specific constituent or patrolling cells. Much of the microenvironment is made up of immune cells or inflammation. Among these, tumor-associated macrophages (TAMs) are of significant interest as they are often involved in facilitating tumor progression as well as the development of resistance to standard therapies. In this review, the ubiquity of TAMs in GB will be discussed while the specific case of microglia resident in the brain will be also emphasized. In addition, the roles of TAMs as accomplices in the progression of GB and resistance to treatment will be presented. Finally, clinical trials targeting TAMs as a means of treating cancer will be discussed.
Collapse
Affiliation(s)
- Hélène Grégoire
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Loris Roncali
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| | - Audrey Rousseau
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Département de Pathologie Cellulaire et Tissulaire, CHU Angers, Angers, France
| | - Michel Chérel
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Yves Delneste
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Laboratoire d'Immunologie et Allergologie, CHU d'Angers, Angers, France
| | - Pascale Jeannin
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Laboratoire d'Immunologie et Allergologie, CHU d'Angers, Angers, France
| | - François Hindré
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,PRIMEX, Plateforme de radiobiologie et d'imagerie expérimentale, SFR ICAT, Université d'Angers, Angers, France
| | - Emmanuel Garcion
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,PACeM, Plateforme d'analyses cellulaires et moléculaires, SFR ICAT, Université d'Angers, Angers, France
| |
Collapse
|
97
|
Paolini L, Adam C, Beauvillain C, Preisser L, Blanchard S, Pignon P, Seegers V, Chevalier LM, Campone M, Wernert R, Verrielle V, Raro P, Ifrah N, Lavoué V, Descamps P, Morel A, Catros V, Tcherkez G, Lenaers G, Bocca C, Kouassi Nzoughet J, Procaccio V, Delneste Y, Jeannin P. Lactic Acidosis Together with GM-CSF and M-CSF Induces Human Macrophages toward an Inflammatory Protumor Phenotype. Cancer Immunol Res 2020; 8:383-395. [PMID: 31924656 DOI: 10.1158/2326-6066.cir-18-0749] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 07/12/2019] [Accepted: 01/03/2020] [Indexed: 11/16/2022]
Abstract
In established tumors, tumor-associated macrophages (TAM) orchestrate nonresolving cancer-related inflammation and produce mediators favoring tumor growth, metastasis, and angiogenesis. However, the factors conferring inflammatory and protumor properties on human macrophages remain largely unknown. Most solid tumors have high lactate content. We therefore analyzed the impact of lactate on human monocyte differentiation. We report that prolonged lactic acidosis induces the differentiation of monocytes into macrophages with a phenotype including protumor and inflammatory characteristics. These cells produce tumor growth factors, inflammatory cytokines, and chemokines as well as low amounts of IL10. These effects of lactate require its metabolism and are associated with hypoxia-inducible factor-1α stabilization. The expression of some lactate-induced genes is dependent on autocrine M-CSF consumption. Finally, TAMs with protumor and inflammatory characteristics (VEGFhigh CXCL8+ IL1β+) are found in solid ovarian tumors. These results show that tumor-derived lactate links the protumor features of TAMs with their inflammatory properties. Treatments that reduce tumor glycolysis or tumor-associated acidosis may help combat cancer.
Collapse
Affiliation(s)
- Léa Paolini
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France
| | - Clément Adam
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France
| | - Céline Beauvillain
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France.,Laboratoire d'Immunologie et Allergologie, CHU d'Angers, Angers, France
| | - Laurence Preisser
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France
| | - Simon Blanchard
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France.,Laboratoire d'Immunologie et Allergologie, CHU d'Angers, Angers, France
| | - Pascale Pignon
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France
| | - Valérie Seegers
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France.,Institut de Cancérologie de l'Ouest, Angers, France
| | - Louise-Marie Chevalier
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France.,Institut de Cancérologie de l'Ouest, Angers, France
| | - Mario Campone
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France.,Institut de Cancérologie de l'Ouest, Angers, France
| | | | | | - Pedro Raro
- Institut de Cancérologie de l'Ouest, Angers, France
| | - Norbert Ifrah
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France.,Service des Maladies du Sang, CHU d'Angers, Angers, France
| | - Vincent Lavoué
- Service de Gynécologie-obstétrique, CHU de Rennes, Rennes, France.,UMR INSERM 1242, Université de Rennes, Rennes, France
| | | | - Alain Morel
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France.,Institut de Cancérologie de l'Ouest, Angers, France
| | - Véronique Catros
- CHU de Rennes, Rennes, France; UMR INSERM 991, Rennes, France; CRB Santé de Rennes, Rennes, France
| | - Guillaume Tcherkez
- Research School of Biology, ANU Joint College of Sciences, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Guy Lenaers
- Université d'Angers, Inserm U1083, CNRS U6015, Institut MitoVasc, Angers, France
| | - Cinzia Bocca
- Université d'Angers, Inserm U1083, CNRS U6015, Institut MitoVasc, Angers, France
| | | | - Vincent Procaccio
- Université d'Angers, Inserm U1083, CNRS U6015, Institut MitoVasc, Angers, France.,Département de Biochimie et Génétique, CHU d'Angers, Angers, France
| | - Yves Delneste
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France.,Laboratoire d'Immunologie et Allergologie, CHU d'Angers, Angers, France
| | - Pascale Jeannin
- Université d'Angers, CHU d'Angers, Inserm U1232, CRCINA, Angers, France. .,Laboratoire d'Immunologie et Allergologie, CHU d'Angers, Angers, France
| |
Collapse
|
98
|
Mdlalose S, Moodley J, Naicker T. The role of follistatin and granulocyte-colony stimulating factor in HIV-associated pre-eclampsia. Pregnancy Hypertens 2019; 19:81-86. [PMID: 31926380 DOI: 10.1016/j.preghy.2019.12.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/23/2019] [Accepted: 12/27/2019] [Indexed: 01/10/2023]
Abstract
KwaZulu-Natal has a high burden of HIV infection and high blood pressure, specifically pre-eclampsia (PE) in pregnancy. Follistatin (FS) and granulocyte-colony stimulating factor (G-CSF) are two glycoproteins involved in PE pathogenesis. In light of the high maternal mortality and morbidity in South Africa (SA), we investigated the expression of FS and G-CSF in the duality of HIV-associated PE. Serum samples of normotensive and pre-eclamptic women were analysed using the Bio-Plex Multiplex Immunoassay. FS expression was significantly reduced in pre-eclamptic (median = 372.0, IQR = 719.2) compared to normotensive (median = 1569.0, IQR = 2043.0) (p < 0.0001). Furthermore, we detected significant FS expression across all study groups. There was a significant difference between HIV -ve normotensive (median = 9.0, IQR = 7.0) vs HIV +ve normotensive (median = 12.0, IQR = 5.0) groups. Additionally, G-CSF expression was notably higher in HIV +ve normotensive when compared to all study groups. This study demonstrated a downregulation of FS and G-CSF expression in PE, compared to normotensive pregnancies. This finding may be attributed to oxidative stress and its immunoregulatory role in the hyperinflammatory milieu of PE. HIV status had no effect on both analytes, albeit upregulated due to immune reconstitution emanating from highly active antiretroviral therapy. Our novel findings suggest that FS and G-CSF may have a potential predictor test value in early pregnancy, hence work on this is ongoing.
Collapse
Affiliation(s)
- Siphesihle Mdlalose
- Optics and Imaging Centre, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, Department of Obstetrics and Gynaecology, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa.
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa.
| |
Collapse
|
99
|
Zhou G, Li Y, Ni J, Jiang P, Bao Z. Role and mechanism of miR-144-5p in LPS-induced macrophages. Exp Ther Med 2019; 19:241-247. [PMID: 31853295 PMCID: PMC6909656 DOI: 10.3892/etm.2019.8218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 10/07/2019] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to explore the possible role of microRNA-144-5p (miR-144-5p) in rheumatoid arthritis (RA) and the associated mechanism. Following the induction of THP-1 cell differentiation into macrophages by phorbol ester (100 ng/ml) treatment, an in vitro inflammatory model of RA was established by treating the THP-1 macrophages with 1 µg/ml lipopolysaccharide (LPS). The level of miR-144-5p was subsequently measured using reverse transcription-quantitative PCR, which found that the expression of miR-144-5p was significantly reduced in LPS-treated THP-1 macrophages. Bioinformatics analysis and a dual-luciferase reporter assay were used to predict and confirm TLR2 as a direct target of miR-144-5p, respectively. Toll-like receptor 2 (TLR2) was then validated as a target gene of miR-144-5p. The effects of miR-144-5p upregulation and TLR2 silencing on LPS-treated THP-1 macrophages were then determined by transfection with miR-144-5p mimic and TLR2-siRNA, respectively. Cell viability was subsequently measured using a Cell Counting Kit-8 assay, whilst the expression of tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-8 secreted by THP-1 macrophages was measured using ELISA. Western blotting was performed to measure p65 phosphorylation (p-p65) in the NK-κB signaling pathway. It was found that miR-144-5p overexpression reduced macrophage cell viability, reduced the expression of TNF-α, IL-6 and IL-8, and reduced the expression of TLR2 and p-p65 compared with the control group. Likewise, TLR2 silencing also reduced macrophage cell viability and reduced the expression of TNF-α, IL-6 and IL-8 in THP-1 macrophages. In conclusion, the data from the present study suggested that miR-144-5p overexpression reduced THP-1 macrophage cell viability and inhibited the expression of TNF-α, IL-6 and IL-8 in cells, possibly by inhibiting the expression of TLR2 and suppressing the activation of NK-κB signaling. Therefore, miR-144-5p may serve as a novel therapeutic target for the treatment of RA.
Collapse
Affiliation(s)
- Guozhu Zhou
- Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210000, P.R. China.,Department of Orthopedics, Jingjiang Chinese Medicine Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Yuwei Li
- Department of Orthopedics, Suzhou Hospital of Traditional Chinese Medicine, Suzhou, Jiangsu 215000, P.R. China
| | - Jianping Ni
- Department of Orthopedics, Jingjiang Chinese Medicine Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Ping Jiang
- Department of Orthopedics, Jingjiang Chinese Medicine Hospital, Taizhou, Jiangsu 214500, P.R. China
| | - Zili Bao
- Department of Orthopedics, Jingjiang Chinese Medicine Hospital, Taizhou, Jiangsu 214500, P.R. China
| |
Collapse
|
100
|
Catalan-Dibene J, McIntyre LL, Zlotnik A. Interleukin 30 to Interleukin 40. J Interferon Cytokine Res 2019; 38:423-439. [PMID: 30328794 DOI: 10.1089/jir.2018.0089] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cytokines are important molecules that regulate the ontogeny and function of the immune system. They are small secreted proteins usually produced upon activation of cells of the immune system, including lymphocytes and myeloid cells. Many cytokines have been described, and several have been recognized as pivotal players in immune responses and in human disease. In fact, several anticytokine antibodies have proven effective therapeutics, especially in various autoimmune diseases. In the last 15 years, new cytokines have been described, and many remain poorly understood. Among the most recent cytokines discovered are interleukins-30 (IL-30) to IL-40. Several of these are members of other cytokine superfamilies, including several IL-1 superfamily members (IL-33, IL-36, IL-37, and IL-38) as well as several new members of the IL-12 family (IL-30, IL-35, and IL-39). The rest (IL-31, IL-32, IL-34, and IL-40) are encoded by genes that do not belong to any cytokine superfamily. Our aim of this review was to present a concise version of the information available on these novel cytokines to facilitate their understanding by members of the immunological community.
Collapse
Affiliation(s)
- Jovani Catalan-Dibene
- 1 Department of Physiology and Biophysics and University of California , Irvine, Irvine, California.,2 Institute for Immunology, University of California, Irvine, Irvine, California
| | - Laura L McIntyre
- 3 Department of Molecular Biology and Biochemistry, University of California , Irvine, Irvine, California.,2 Institute for Immunology, University of California, Irvine, Irvine, California
| | - Albert Zlotnik
- 1 Department of Physiology and Biophysics and University of California , Irvine, Irvine, California.,2 Institute for Immunology, University of California, Irvine, Irvine, California
| |
Collapse
|