1
|
Garemilla SSS, Gampa SC, Garimella S. Role of the tumor microenvironment in cancer therapy: unveiling new targets to overcome drug resistance. Med Oncol 2025; 42:202. [PMID: 40332723 DOI: 10.1007/s12032-025-02754-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Abstract
Cancer is a leading cause of death globally, with resistance to therapy representing a major obstacle to effective treatment. The tumor microenvironment (TME), comprising a complex network to cellular and non-cellular components including cancer-associated fibroblasts, immune cells, the extracellular matrix and region of hypoxia, is integral to cancer progression and therapeutic resistance. This review delves into the multifaceted interactions within the TME that contribute to tumor growth, survival and immune evasion. Key elements such as the role of cancer- associated fibroblasts in remodeling the extracellular matrix and promoting angiogenesis, the influence of immune cells such as tumor-associated macrophages in creating an immunosuppressive milieu and the impact of hypoxia conditions on metabolic adaptation and therapy resistance are thoroughly examined. This review evaluates current and emerging TME-targeted therapeutic strategies, including inhibitors of extracellular matrix components, modulators of immune cell activity and approached to alleviate hypoxia. Combination therapies that integrate TME-targeted agents with conventional treatments such as chemotherapy and immunotherapy are also discussed for their potential to enhance treatment efficacy and circumvent resistance mechanisms. By synthesising recent advances in TME research and therapeutic innovation, this paper aims to underscore the importance of TME in cancer therapy and highlight promising avenues for improving patient outcomes through targeted intervention.
Collapse
Affiliation(s)
| | - Siri Chandana Gampa
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India
| | - Sireesha Garimella
- Department of Life Sciences, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India.
| |
Collapse
|
2
|
Alshahrani S, Ashafaq M, Jali AM, Almoshari Y, Alam MI, Al Shahi H, Alshamrani AA, Hussain S. Nephrotoxic effect of cypermethrin ameliorated by nanocurcumin through antioxidative mechanism. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03825-5. [PMID: 39878820 DOI: 10.1007/s00210-025-03825-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025]
Abstract
Cypermethrin is a pyrethroid showing nephrotoxicity by generating ROS-impaired oxidative stress and changes in inflammatory and apoptotic markers. The harmful consequences are intended to be mitigated by the imbalance between oxidants and antioxidants. The anti-inflammatory and antioxidant possessions of nanocurcumin (NC) with improved bioavailability ameliorate Cyp toxicity in rat kidneys. In our study, Group I was the control while Group II was treated alone with NC (5 mg). Group III was given 50 mg/kg of Cyp for two weeks. Groups IV, V, and VI received Cyp as in group III and co-treatment with varying NC doses after 5 days of Cyp dosing, respectively. All treatments were given orally for two weeks. After the termination of the study, LPO, 4-HNE, GSH, antioxidant catalase, and SOD were evaluated as markers of inflammation and apoptosis along with ELISA, qRT-PCR, and histopathology were used to assess morphological changes. Our work has shown that Cyp causes nephrotoxicity by altering all parameters. The Cyp-treated group was shown to have higher expression of the oxidative stress marker LPO and inflammatory interleukins as well as Bax, NF-kB, caspase-3, and caspase-9. Although LPO, inflammation, and apoptosis are reduced, antioxidant status is improved by NC.
Collapse
Affiliation(s)
- Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Mohammad Ashafaq
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Abdulmajeed M Jali
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Yosif Almoshari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Mohammad Intakhab Alam
- Department of Pharmaceutics, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Hamad Al Shahi
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Ayed A Alshamrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia
| | - Sohail Hussain
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, 45142, Jazan, Saudi Arabia.
- Gandaki Medical College, Lekhnath-27, Pokhara, Nepal.
| |
Collapse
|
3
|
Topham B, Hock B, Phillips E, Wiggins G, Currie M. The Role of Innate Priming in Modifying Tumor-associated Macrophage Phenotype. FRONT BIOSCI-LANDMRK 2024; 29:418. [PMID: 39735978 DOI: 10.31083/j.fbl2912418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 12/31/2024]
Abstract
Tumor-associated macrophages (TAMs) are innate immune cells that exert far reaching influence over the tumor microenvironment (TME). Depending on cues within the local environment, TAMs may promote tumor angiogenesis, cancer cell invasion and immunosuppression, or, alternatively, inhibit tumor progression via neoantigen presentation, tumoricidal reactive oxygen species generation and pro-inflammatory cytokine secretion. Therefore, TAMs have a pivotal role in determining tumor progression and response to therapy. TAM phenotypes are driven by cytokines and physical cues produced by tumor cells, adipocytes, fibroblasts, pericytes, immune cells, and other cells within the TME. Research has shown that TAMs can be primed by environmental stimuli, adding another layer of complexity to the environmental context that determines TAM phenotype. Innate priming is a functional consequence of metabolic and epigenetic reprogramming of innate cells by a primary stimulant, resulting in altered cellular response to future secondary stimulation. Innate priming offers a novel target for development of cancer immunotherapy and improved prognosis of disease, but also raises the risk of exacerbating existing inflammatory pathologies. This review will discuss the mechanisms underlying innate priming including metabolic and epigenetic modification, its relevance to TAMs and tumor progression, and possible clinical implications for cancer treatment.
Collapse
Affiliation(s)
- Ben Topham
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| | - Barry Hock
- Haematology Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| | - George Wiggins
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| | - Margaret Currie
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch, 8011 Christchurch, Aotearoa New Zealand
| |
Collapse
|
4
|
Liu Z, Xue X, Geng S, Jiang Z, Ge Z, Zhao C, Xu Y, Wang X, Zhang W, Lin L, Chen Z. The differences in cytokine signatures between severe fever with thrombocytopenia syndrome (SFTS) and hemorrhagic fever with renal syndrome (HFRS). J Virol 2024; 98:e0078624. [PMID: 38916398 PMCID: PMC11265425 DOI: 10.1128/jvi.00786-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) virus and hantavirus are categorized under the Bunyavirales order. The severe disease progression in both SFTS and hemorrhagic fever with renal syndrome (HFRS) is associated with cytokine storms. This study aimed to explore the differences in cytokine profiles and immune responses between the two diseases. A cross-sectional, single-center study involved 100 participants, comprising 46 SFTS patients, 48 HFRS patients, and 6 healthy controls. The study employed the Luminex cytokine detection platform to measure 48 cytokines. The differences in cytokine profiles and immune characteristics between the two diseases were further analyzed using multiple linear regression, principal component analysis, and random forest method. Among the 48 cytokines tested, 30 showed elevated levels in SFTS and/or HFRS compared to the healthy control group. Furthermore, there were 19 cytokines that exhibited significant differences between SFTS and HFRS. Random forest analysis suggested that TRAIL and CTACK were predictive of SFTS, while IL2Ralpha, MIG, IL-8, IFNalpha2, HGF, SCF, MCP-3, and PDGFBB were more common with HFRS. It was further verified by the receiver operating characteristic with area under the curve >0.8 and P-values <0.05, except for TRAIL. Significant differences were observed in the cytokine profiles of SFTS and HFRS, with TRAIL, IL2Ralpha, MIG, and IL-8 being the top 4 cytokines that most clearly distinguished the two diseases. IMPORTANCE SFTS and HFRS differ in terms of cytokine immune characteristics. TRAIL, IL-2Ralpha, MIG, and IL-8 were the top 4 that differed markedly between SFTS and HFRS.
Collapse
Affiliation(s)
- Zishuai Liu
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyu Xue
- Department of Infectious Disease, Beijing Ditan Hospital, Peking University, Beijing, China
| | - Shuying Geng
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, China
| | - Zhouling Jiang
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ziruo Ge
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Chenxi Zhao
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yanli Xu
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, China
| | - Xiaolei Wang
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ling Lin
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, China
| | - Zhihai Chen
- Department of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Department of Infectious Disease, Beijing Ditan Hospital, Peking University, Beijing, China
| |
Collapse
|
5
|
Luo C, He S, Shi F, Zhou J, Shang L. The Role of TRAIL Signaling in Cancer: Searching for New Therapeutic Strategies. BIOLOGY 2024; 13:521. [PMID: 39056714 PMCID: PMC11274015 DOI: 10.3390/biology13070521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
Cancer continues to pose a significant threat to global health, with its status as a leading cause of death remaining unchallenged. Within the realm of cancer research, the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) stands out as a critical player, having been identified in the 1990s as the tenth member of the TNF family. This review examines the pivotal role of TRAIL in cancer biology, focusing on its ability to induce apoptosis in malignant cells through both endogenous and exogenous pathways. We provide an in-depth analysis of TRAIL's intracellular signaling and intercellular communication, underscoring its potential as a selective anticancer agent. Additionally, the review explores TRAIL's capacity to reshape the tumor microenvironment, thereby influencing cancer progression and response to therapy. With an eye towards future developments, we discuss the prospects of harnessing TRAIL's capabilities for the creation of tailored, precision-based cancer treatments, aiming to enhance efficacy and improve patient survival rates.
Collapse
Affiliation(s)
- Cheng Luo
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Shan He
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Jianhua Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
| | - Li Shang
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
| |
Collapse
|
6
|
Cornice J, Verzella D, Arboretto P, Vecchiotti D, Capece D, Zazzeroni F, Franzoso G. NF-κB: Governing Macrophages in Cancer. Genes (Basel) 2024; 15:197. [PMID: 38397187 PMCID: PMC10888451 DOI: 10.3390/genes15020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/26/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are the major component of the tumor microenvironment (TME), where they sustain tumor progression and or-tumor immunity. Due to their plasticity, macrophages can exhibit anti- or pro-tumor functions through the expression of different gene sets leading to distinct macrophage phenotypes: M1-like or pro-inflammatory and M2-like or anti-inflammatory. NF-κB transcription factors are central regulators of TAMs in cancers, where they often drive macrophage polarization toward an M2-like phenotype. Therefore, the NF-κB pathway is an attractive therapeutic target for cancer immunotherapy in a wide range of human tumors. Hence, targeting NF-κB pathway in the myeloid compartment is a potential clinical strategy to overcome microenvironment-induced immunosuppression and increase anti-tumor immunity. In this review, we discuss the role of NF-κB as a key driver of macrophage functions in tumors as well as the principal strategies to overcome tumor immunosuppression by targeting the NF-κB pathway.
Collapse
Affiliation(s)
- Jessica Cornice
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Paola Arboretto
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| | - Davide Vecchiotti
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, 67100 L’Aquila, Italy; (D.V.); (D.C.); (F.Z.)
| | - Guido Franzoso
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK; (J.C.); (P.A.)
| |
Collapse
|
7
|
Kiran GS, Kumar PK, Mitra P, Kumar K, Goyal T, Sharma S, Rana S, Sharma P. Unravelling blood-based epigenetic mechanisms: the impact of hsa-miR-146a and histone H3 acetylation in lead-induced inflammation among occupational workers. Int Arch Occup Environ Health 2023; 96:1257-1266. [PMID: 37572134 DOI: 10.1007/s00420-023-02004-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/26/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Occupational and environmental exposure to lead (Pb) is a persistent health problem majorly in developing countries and has been implied to cause epigenetic alterations. Its effect on histone post-translational modifications is not explored in human population. MicroRNAs are epigenetic modulators reported to be differentially expressed under Pb exposure. The present study was targeted to find plausible association between the role of hsa-miR-146a and global histone (H3) acetylation in Pb-induced inflammation in occupationally exposed workers. MATERIALS AND METHODS A total of 100 occupationally exposed individuals working in different industries were recruited for the study and divided into 2 groups based on the median Pb levels [low Pb group (Pb < 5 μg/dL) and High Pb group (Pb > 5 μg/dL)]. The Pb levels were measured in whole blood using atomic absorption spectrometry to confirm Pb exposure. Histone H3 acetylation and serum interleukin-6 (IL-6) levels were measured using colorimetric methods and enzyme-linked immunosorbent assay (ELISA), respectively. MicroRNA-146a expression was quantified using TaqMan assay. RESULTS The median BLL of the study population was 5 μg/dL. BLL, IL-6, and Histone (H3) acetylation increased significantly with the duration of exposure. BLL level showed a significant positive correlation with IL-6 and histone H3 acetylation level. We also found that hsa-miR-146a exhibited significantly increased expression in the high Pb group compared to the low Pb group (Fold change: 2.56; P = 0.014). The linear regression model suggested that BLL has significantly predicted histone H3 acetylation, hsa-miR-146a, and IL-6 in the study subjects. CONCLUSION The finding that hsa-miR146a was significantly upregulated in individuals with high BLL and had a significant negative correlation with serum IL-6 suggests that Pb-induced oxidative stress likely activates H3 acetylation, which then releases inflammatory cytokines like IL-6.
Collapse
Affiliation(s)
- Gangam Sai Kiran
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Rajasthan, India
| | - Pvsn Kiran Kumar
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Rajasthan, India
- Department of Biochemistry, Andhra Medical College, Visakhapatnam, India
| | - Prasenjit Mitra
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kanishka Kumar
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Rajasthan, India
| | - Taru Goyal
- Department of Paediatric Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shailja Sharma
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Rajasthan, India.
| | - Shweta Rana
- Department of Physical and Natural Sciences, FLAME University, Pune, India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Rajasthan, India
| |
Collapse
|
8
|
Pimentel JM, Zhou JY, Kim S, Gurdziel K, Wu GS. The role of the immune response and inflammatory pathways in TNF-related apoptosis-inducing ligand (TRAIL) resistance in triple-negative breast cancer cells. Am J Cancer Res 2023; 13:4678-4692. [PMID: 37970367 PMCID: PMC10636687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 07/31/2023] [Indexed: 11/17/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer, and the majority of TNBC lacks targeted therapies. Previous studies have shown that TNBC cells are highly sensitive to TNF-related apoptosis-inducing ligand (TRAIL), making it a potentially viable treatment option for TNBC. However, the development of TRAIL resistance limits its potential for clinical use, and the underlying mechanisms are not fully understood. To better understand the mechanism of resistance to TRAIL, we performed RNA sequencing to identify the candidates that are responsible for resistance to TRAIL in two previously established TRAIL-resistant MDA231 and SUM159 cells. This approach led us to identify differentially expressed genes (DEGs) and pathways in TRAIL-resistant MDA231 and SUM159 cells compared to their TRAIL-sensitive counterparts. We showed that several DEGs and pathways were associated with inflammation in TRAIL-resistant cells, including IL-1α and IL6. By downregulating IL-1α and IL6 expression, we showed that TRAIL sensitivity can be significantly restored in TRAIL-resistant cells. Therefore, this study identifies a mechanism by which the inflammation pathway promotes TRAIL resistance, which could be targeted for enhancing TRAIL-based therapies in TNBC cells.
Collapse
Affiliation(s)
- Julio M Pimentel
- Cancer Biology Program, Wayne State University School of MedicineDetroit, Michigan 48201, USA
- Department of Oncology, Wayne State UniversityDetroit, Michigan 48201, USA
| | - Jun-Ying Zhou
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan 48201, USA
- Department of Oncology, Wayne State UniversityDetroit, Michigan 48201, USA
| | - Seongho Kim
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan 48201, USA
- Department of Oncology, Wayne State UniversityDetroit, Michigan 48201, USA
| | - Katherine Gurdziel
- Department of Pharmacology, Wayne State UniversityDetroit, Michigan 48201, USA
- Institute of Environmental Health Sciences, Wayne State UniversityDetroit, Michigan 48201, USA
| | - Gen Sheng Wu
- Cancer Biology Program, Wayne State University School of MedicineDetroit, Michigan 48201, USA
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of MedicineDetroit, Michigan 48201, USA
- Department of Oncology, Wayne State UniversityDetroit, Michigan 48201, USA
- Department of Pathology, Wayne State University School of MedicineDetroit, Michigan 48201, USA
| |
Collapse
|
9
|
Gunalp S, Helvaci DG, Oner A, Bursalı A, Conforte A, Güner H, Karakülah G, Szegezdi E, Sag D. TRAIL promotes the polarization of human macrophages toward a proinflammatory M1 phenotype and is associated with increased survival in cancer patients with high tumor macrophage content. Front Immunol 2023; 14:1209249. [PMID: 37809073 PMCID: PMC10551148 DOI: 10.3389/fimmu.2023.1209249] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
Background TNF-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can either induce cell death or activate survival pathways after binding to death receptors (DRs) DR4 or DR5. TRAIL is investigated as a therapeutic agent in clinical trials due to its selective toxicity to transformed cells. Macrophages can be polarized into pro-inflammatory/tumor-fighting M1 macrophages or anti-inflammatory/tumor-supportive M2 macrophages and an imbalance between M1 and M2 macrophages can promote diseases. Therefore, identifying modulators that regulate macrophage polarization is important to design effective macrophage-targeted immunotherapies. The impact of TRAIL on macrophage polarization is not known. Methods Primary human monocyte-derived macrophages were pre-treated with either TRAIL or with DR4 or DR5-specific ligands and then polarized into M1, M2a, or M2c phenotypes in vitro. The expression of M1 and M2 markers in macrophage subtypes was analyzed by RNA sequencing, qPCR, ELISA, and flow cytometry. Furthermore, the cytotoxicity of the macrophages against U937 AML tumor targets was assessed by flow cytometry. TCGA datasets were also analyzed to correlate TRAIL with M1/M2 markers, and the overall survival of cancer patients. Results TRAIL increased the expression of M1 markers at both mRNA and protein levels while decreasing the expression of M2 markers at the mRNA level in human macrophages. TRAIL also shifted M2 macrophages towards an M1 phenotype. Our data showed that both DR4 and DR5 death receptors play a role in macrophage polarization. Furthermore, TRAIL enhanced the cytotoxicity of macrophages against the AML cancer cells in vitro. Finally, TRAIL expression was positively correlated with increased expression of M1 markers in the tumors from ovarian and sarcoma cancer patients and longer overall survival in cases with high, but not low, tumor macrophage content. Conclusions TRAIL promotes the polarization of human macrophages toward a proinflammatory M1 phenotype via both DR4 and DR5. Our study defines TRAIL as a new regulator of macrophage polarization and suggests that targeting DRs can enhance the anti-tumorigenic response of macrophages in the tumor microenvironment by increasing M1 polarization.
Collapse
Affiliation(s)
- Sinem Gunalp
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Derya Goksu Helvaci
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Aysenur Oner
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | | | - Alessandra Conforte
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Hüseyin Güner
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Molecular Biology and Genetics, Faculty of Life and Natural Science, Abdullah Gül University, Kayseri, Türkiye
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Eva Szegezdi
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Duygu Sag
- Izmir Biomedicine and Genome Center, Izmir, Türkiye
- Department of Genomic Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| |
Collapse
|
10
|
Qiu N, Zhang Z, Wei X, Xu C, Jia X, Wang K, Chen Y, Wang S, Su R, Cen B, Shen Y, Chen C, Liu Y, Xu X. Peritoneal Gene Transfection of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand for Tumor Surveillance and Prophylaxis. NANO LETTERS 2023; 23:7859-7868. [PMID: 37433066 DOI: 10.1021/acs.nanolett.3c01568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Abstract
Peritoneal metastasis is very common in gastrointestinal, reproductive, and genitourinary tract cancers in late stages or postsurgery, causing poor prognosis, so effective and nontoxic prophylactic strategies against peritoneal metastasis are highly imperative. Herein, we demonstrate the first gene transfection as a nontoxic prophylaxis preventing peritoneal metastasis or operative metastatic dissemination. Lipopolyplexes of TNF-related-apoptosis-inducing-ligand (TRAIL) transfected peritonea and macrophages to express TRAIL for over 15 days. The expressed TRAIL selectively induced tumor cell apoptosis while exempting normal tissue, providing long-term tumor surveillance. Therefore, tumor cells inoculated in the pretransfected peritoneal cavity quickly underwent apoptosis and, thus, barely formed tumor nodules, significantly prolonging the mouse survival time compared with chemotherapy prophylaxis. Furthermore, lipopolyplex transfection showed no sign of toxicity. Therefore, this peritoneal TRAIL-transfection is an effective and safe prophylaxis, preventing peritoneal metastasis.
Collapse
Affiliation(s)
- Nasha Qiu
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Zhejiang Key Laboratory of Smart Biomaterials and College of Chemical and Biological Engineering, Zhejiang Univeristy, Hangzhou 310027, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100090, China
| | - Zhen Zhang
- Zhejiang Longcharm Bio-tech Pharma Co., Ltd. Hangzhou 310027, China
| | - Xuyong Wei
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chang Xu
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaolong Jia
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Kai Wang
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yunqi Chen
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Shuai Wang
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Renyi Su
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Beini Cen
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and College of Chemical and Biological Engineering, Zhejiang Univeristy, Hangzhou 310027, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100090, China
| | - Yanpeng Liu
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiao Xu
- The Center for Integrated Oncology and Precision Medicine, Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
11
|
Immune Phenotypic Characterization of a TRAIL-Knockout Mouse. Cancers (Basel) 2023; 15:cancers15051475. [PMID: 36900266 PMCID: PMC10000729 DOI: 10.3390/cancers15051475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
The TNF-superfamily member TRAIL is known to mediate selective apoptosis in tumor cells suggesting this protein as a potential antitumor drug target. However, initial successful pr-clinical results could not be translated into the clinic. Reasons for the ineffectiveness of TRAIL-targeting in tumor therapies could include acquired TRAIL resistance. A tumor cell acquires TRAIL resistance, for example, by upregulation of antiapoptotic proteins. In addition, TRAIL can also influence the immune system and thus, tumor growth. We were able to show in our previous work that TRAIL-/- mice show improved survival in a mouse model of pancreatic carcinoma. Therefore, in this study we aimed to immunologically characterize the TRAIL-/- mice. We observed no significant differences in the distribution of CD3+, CD4+, CD8+ T-cells, Tregs, and central memory CD4+ and CD8+ cells. However, we provide evidence for relevant differences in the distribution of effector memory T-cells and CD8+CD122+ cells but also in dendritic cells. Our findings suggest that T-lymphocytes of TRAIL-/- mice proliferate at a lower rate, and that the administration of recombinant TRAIL significantly increases their proliferation, while regulatory T-cells (Tregs) from TRAIL-/- mice are less suppressive. Regarding the dendritic cells, we found more type-2 conventional dendritic cells (DC2s) in the TRAIL-/- mice. For the first time (to the best of our knowledge), we provide a comprehensive characterization of the immunological landscape of TRAIL-deficient mice. This will establish an experimental basis for future investigations of TRAIL-mediated immunology.
Collapse
|
12
|
Chakraborty A, Diwan A. Biomarkers and molecular mechanisms of Amyotrophic Lateral Sclerosis. AIMS Neurosci 2022; 9:423-443. [PMID: 36660079 PMCID: PMC9826749 DOI: 10.3934/neuroscience.2022023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease in adults involving non-demyelinating motor disorders. About 90% of ALS cases are sporadic, while 10-12% of cases are due to some genetic reasons. Mutations in superoxide dismutase 1 (SOD1), TAR, c9orf72 (chromosome 9 open reading frame 72) and VAPB genes are commonly found in ALS patients. Therefore, the mechanism of ALS development involves oxidative stress, endoplasmic reticulum stress, glutamate excitotoxicity and aggregation of proteins, neuro-inflammation and defective RNA function. Cholesterol and LDL/HDL levels are also associated with ALS development. As a result, sterols could be a suitable biomarker for this ailment. The main mechanisms of ALS development are reticulum stress, neuroinflammation and RNA metabolism. The multi-nature development of ALS makes it more challenging to pinpoint a treatment.
Collapse
|
13
|
Immunophenotypic Analysis of T Lymphocytes and Cytokine Production in Elderly Practicing Physical Activities and Its Relationship with Quality of Life and Depression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7985596. [PMID: 36193083 PMCID: PMC9526638 DOI: 10.1155/2022/7985596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/12/2022] [Indexed: 11/18/2022]
Abstract
Aging is a complex process often associated with a chronic inflammatory profile that alters several biological functions, including the immune system and cognitive and physical capacity. The practice of physical activity is increasingly gaining popularity as a method of preventing infections, depression, and other disorders that affect the quality of life of the elderly. Thus, this work analyzes the profile of cytokines and molecular markers expressed in immune cells of elderly people who practice physical activities or not, evaluating their impacts on the immune system and quality of life. For this, 48 individuals were recruited, and peripheral blood samples were collected for hemogram analysis, cytokine determination, and immunophenotyping. Elderly people were separated into two groups: practitioners with low-intensity physical activity and non-practitioners. Quality of life was assessed using the Whoqol-Old instrument, and depression was assessed using the Beck II Depression Inventory. When comparing the scores of the Whoqol-Old and Beck questionnaires, we observed a significant negative correlation between these two factors. The perception of a higher quality of life was present in the elderly who exercised and was related to greater autonomy and sensory abilities, whereas the presence of depression was lower. In the hemogram, we observed higher basophil and segmented counts in the sedentary elderly, whereas lymphocytes and monocytes had lower counts. Elderly practitioners of physical activities had higher levels of IFN-γ, IL-4, and IL-10; increased expression of CD69, PD1, and TIM-3 in CD4+ T lymphocytes and increased CD14+CD80+ and CD14+CD86+ monocytes. Elderly people with an increased perception of quality of life had higher levels of IFN-γ, higher expression of CD14+CD80+CD86+, and decreased levels of TRAIL. An increase in TRAIL was observed in individuals with depression, in addition to an increased expression of CD14+CD86+. These results show a clear correlation between the quality of life, level of depression, physical activity, and immune system function. Although some cytokines with a typical proinflammatory profile (IFN-γ) were observed, the results point to a protective state with benefits reflected in the general well-being of the elderly who exercise.
Collapse
|
14
|
Jang B, Song HK, Hwang J, Lee S, Park E, Oh A, Hwang ES, Sung JY, Kim YN, Park K, Lee YM, Oh ES. Shed syndecan-2 enhances colon cancer progression by increasing cooperative angiogenesis in the tumor microenvironment. Matrix Biol 2022; 107:40-58. [DOI: 10.1016/j.matbio.2022.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/12/2022] [Accepted: 02/02/2022] [Indexed: 12/24/2022]
|
15
|
Wang X, Wu Y, Gu J, Xu J. Tumor-associated macrophages in lung carcinoma: From mechanism to therapy. Pathol Res Pract 2021; 229:153747. [PMID: 34952424 DOI: 10.1016/j.prp.2021.153747] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/09/2022]
Abstract
Tumor-associated macrophages (TAMs), which could be classified into the classical (M1-like) and alternatively activated (M2-like) phenotype, were considered to be important tumor-promoting components in lung cancer microenvironment. Several studies reported that TAMs in lung tumor islet or stroma are usually correlated with poor prognosis. Further studies showed that TAMs could promote the initiation of tumor cells, inhibit antitumor immune responses, and stimulate tumor angiogenesis and subsequently tumor metastasis of lung carcinoma. Currently, TAMs have been considered as penitential targets of lung cancer. This review summarizes from the fundamental information of TAMs to the its role in metastasis and present evidence for TAMs as a potential target of cancer therapy.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yining Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiahui Gu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jian Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; National Key Clinical Department of Laboratory Medicine, Nanjing 210029, China.
| |
Collapse
|
16
|
Lu J, He X, Zhang L, Zhang R, Li W. Acetylation in Tumor Immune Evasion Regulation. Front Pharmacol 2021; 12:771588. [PMID: 34880761 PMCID: PMC8645962 DOI: 10.3389/fphar.2021.771588] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
Acetylation is considered as one of the most common types of epigenetic modifications, and aberrant histone acetylation modifications are associated with the pathological process of cancer through the regulation of oncogenes and tumor suppressors. Recent studies have shown that immune system function and tumor immunity can also be affected by acetylation modifications. A comprehensive understanding of the role of acetylation function in cancer is essential, which may help to develop new therapies to improve the prognosis of cancer patients. In this review, we mainly discussed the functions of acetylase and deacetylase in tumor, immune system and tumor immunity, and listed the information of drugs targeting these enzymes in tumor immunotherapy.
Collapse
Affiliation(s)
- Jun Lu
- Hunan Normal University School of Medicine, Changsha, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
| | - Xiang He
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China.,Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
| | - Lijuan Zhang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Ran Zhang
- Hunan Normal University School of Medicine, Changsha, China
| | - Wenzheng Li
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Liu H, Wang H, Ma J, Qiao Z, Zhang L, Ge G. MicroRNA-146a-3p/HDAC1/KLF5/IKBα signal axis modulates plaque formation of atherosclerosis mice. Life Sci 2021; 284:119615. [PMID: 34004248 DOI: 10.1016/j.lfs.2021.119615] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Atherosclerosis (AS) is a multifocal, smoldering immune inflammatory disease of medium and large arteries driven by lipids. The aim of this study is to discuss the mechanism of microRNA-146a-3p (miR-146a-3p)/histone deacetylase 1 (HDAC1)/Krüppel-like factor 5 (KLF5)/inhibitors of kappa B α (IKBα) signal axis in plaque formation of AS mice. METHODS ApoE-/- mice were fed with high-fat feed for 12 weeks to establish AS mice model. The expression of miR-146a-3p, KLF5, HDAC1 and IKBα in aortic wall tissues of AS mice was tested. The targeting relationship between miR-146a-3p and HDAC1 was verified. AS mice were injected with miR-146a-3p antagomir or HDAC1 overexpression to verify the impacts of miR-146a-3p and HDAC1 on blood lipids and inflammatory factors in serum, aortic wall apoptotic cells, antioxidant stress capacity and the plaque area in AS mice. VECs proliferation and apoptosis were also measured in vitro. RESULTS miR-146a-3p and KLF5 were increased while HDAC1 and IKBα were reduced in aortic wall tissues of AS mice. miR-146a-3p directly targeted to HDAC1. Depletion of miR-146a-3p or restoration of HDAC1 was correlated to lower plasma lipid level, reduced inflammatory factors in serum, attenuated aortic wall apoptosis, increased antioxidant stress capacity and improved the stability of pathological plaque of AS mice. miR-146a-3p down-regulation or HDAC1 up-regulation promoted VECs proliferation and inhibited apoptosis. CONCLUSION Functional studies show that depleted miR-146a-3p advances HDAC1 and IKBα expression as well as inhibits KLF5 expression to facilitate the stability of pathological plaques in AS mice.
Collapse
Affiliation(s)
- Huajin Liu
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China.
| | - Hongwei Wang
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| | - Jiangwei Ma
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| | - Zengyong Qiao
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| | - Li Zhang
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| | - Guanghao Ge
- Department of Cardiology, Fengxian District Central Hospital, Shanghai, China
| |
Collapse
|
18
|
Reis-Sobreiro M, Teixeira da Mota A, Jardim C, Serre K. Bringing Macrophages to the Frontline against Cancer: Current Immunotherapies Targeting Macrophages. Cells 2021; 10:2364. [PMID: 34572013 PMCID: PMC8464913 DOI: 10.3390/cells10092364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/07/2021] [Accepted: 08/29/2021] [Indexed: 12/21/2022] Open
Abstract
Macrophages are found in all tissues and display outstanding functional diversity. From embryo to birth and throughout adult life, they play critical roles in development, homeostasis, tissue repair, immunity, and, importantly, in the control of cancer growth. In this review, we will briefly detail the multi-functional, protumoral, and antitumoral roles of macrophages in the tumor microenvironment. Our objective is to focus on the ever-growing therapeutic opportunities, with promising preclinical and clinical results developed in recent years, to modulate the contribution of macrophages in oncologic diseases. While the majority of cancer immunotherapies target T cells, we believe that macrophages have a promising therapeutic potential as tumoricidal effectors and in mobilizing their surroundings towards antitumor immunity to efficiently limit cancer progression.
Collapse
Affiliation(s)
| | | | | | - Karine Serre
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal; (M.R.-S.); (A.T.d.M.); (C.J.)
| |
Collapse
|
19
|
Makled MN, Said E. Tranilast abrogates cisplatin-induced testicular and epididymal injuries: An insight into its modulatory impact on apoptosis/proliferation. J Biochem Mol Toxicol 2021; 35:e22817. [PMID: 34047436 DOI: 10.1002/jbt.22817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/17/2021] [Accepted: 05/18/2021] [Indexed: 12/20/2022]
Abstract
Cisplatin is a chemotherapeutic agent whose therapeutic use is greatly limited by the associated organs' toxicity and particularly, testicular toxicity. Cisplatin-induced testicular damage reported being mediated through mitochondria-mediated apoptosis, inflammation, and oxidative stress. Evidence showed that tranilast (TRN) has the ability to restore the oxidative status and modulate TRAIL/caspase-8 signaling. This led us to hypothesize that TRN could abrogate cisplatin-induced testicular and epididymal injuries via inhibiting oxidative stress and modulating proliferation and TRAIL/caspase-8/cJNK signaling. Cisplatin injection induced oligospermia and abnormalities in testicular and epididymal structure along with impaired oxidative status. TRN administration (100 or 300 mg/kg) for 7 days post-cisplatin injection preserved spermatogenesis and restored testicular and epididymal architecture, but restoration was more so in TRN300 than TRN100. This was in line with the restoration of balanced oxidative status as indicated by the increased total antioxidant capacity, glutathione and superoxide dismutase activity, and the decreased malondialdehyde content in testes (p < 0.05 vs. cisplatin). TRN increased the cell proliferation revealed by the increased expression of proliferating cell nuclear antigen in a dose-dependent manner (p < 0.05 vs. cisplatin) whereas only TRN300 decreased testicular cJNK, TRAIL, and caspase-8 expression (p < 0.05 vs. cisplatin). Moreover, TRN dose-dependently inhibited the pro-inflammatory transcription factor NF-kB and the cytokine TNF-α expressions in testes. In conclusion, TRN300 was more effective than TRN100 in alleviating cisplatin-induced testicular and epididymal injuries and in enhancing spermatogenesis. This curative effect of TRN might be mediated through its antioxidant and anti-inflammatory impacts along with its modulatory impact on cJNK/TRAIL/caspase-8 signaling favoring proliferation rather than apoptosis.
Collapse
Affiliation(s)
- Mirhan N Makled
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Eman Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
20
|
Xu L, Xie X, Luo Y. The role of macrophage in regulating tumour microenvironment and the strategies for reprogramming tumour-associated macrophages in antitumour therapy. Eur J Cell Biol 2021; 100:151153. [PMID: 33476912 DOI: 10.1016/j.ejcb.2021.151153] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 01/07/2023] Open
Abstract
Tumour-associated macrophages (TAMs) that present abundantly in the tumour microenvironment (TME) exhibit a protumour property, such as promoting genetic instability, tumour metastasis and immunosuppression. Macrophage-targeted therapeutic approaches hence have been applied and shown their significances in the process of tumour immune treatment, including blocking TAM recruitment, depleting or transforming TAMs that already exist in the tumour site. Here, we summarized the functional regulation of TAMs in the respects of hypoxia environment, metabolism in the tumour microenvironment and the transcription factors involved. We reviewed the strategies for transforming TAMs, including immune stimuli targeting TAMs, inhibitors against TAMs, pathogen or irradiation stimulation on TAMs, and the application of natural compounds in TAMs. Furthermore, we also discussed the macrophage-targeted therapies in the clinical studies. Taken together, this review tries to shed light on the TAM regulation and the main strategies of TAM reprogramming for an enhanced immune surveillance.
Collapse
Affiliation(s)
- Liping Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, PR China; Medical School, Kunming University of Science and Technology, Kunming, 650500, PR China
| | - Xiaoli Xie
- Medical School, Kunming University of Science and Technology, Kunming, 650500, PR China
| | - Ying Luo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, PR China; Guizhou Provincial Key Laboratory & Drug Development on Common Disease, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
21
|
Hu W, Zhang L, Dong Y, Tian Z, Chen Y, Dong S. Tumour dormancy in inflammatory microenvironment: A promising therapeutic strategy for cancer-related bone metastasis. Cell Mol Life Sci 2020; 77:5149-5169. [PMID: 32556373 PMCID: PMC11104789 DOI: 10.1007/s00018-020-03572-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/22/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023]
Abstract
Cancer metastasis is a unique feature of malignant tumours. Even bone can become a common colonization site due to the tendency of solid tumours, including breast cancer (BCa) and prostate cancer (PCa), to metastasize to bone. Currently, a previous concept in tumour metabolism called tumour dormancy may be a promising target for antitumour treatment. When disseminated tumour cells (DTCs) metastasize to the bone microenvironment, they form a flexible regulatory network called the "bone-tumour-inflammation network". In this network, bone turnover as well as metabolism, tumour progression, angiogenesis and inflammatory responses are highly unified and coordinated, and a slight shift in this balance can result in the disruption of the microenvironment, uncontrolled inflammatory responses and excessive tumour growth. The purpose of this review is to highlight the regulatory effect of the "bone-tumour-inflammation network" in tumour dormancy. Osteoblast-secreted factors, bone turnover and macrophages are emphasized and occupy in the main part of the review. In addition, the prospective clinical application of tumour dormancy is also discussed, which shows the direction of future research.
Collapse
Affiliation(s)
- Wenhui Hu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Lincheng Zhang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yutong Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhansong Tian
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yueqi Chen
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
22
|
Cardoso Alves L, Corazza N, Micheau O, Krebs P. The multifaceted role of TRAIL signaling in cancer and immunity. FEBS J 2020; 288:5530-5554. [PMID: 33215853 DOI: 10.1111/febs.15637] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/29/2022]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can lead to the induction of apoptosis in tumor or infected cells. However, activation of TRAIL signaling may also trigger nonapoptotic pathways in cancer and in nontransformed cells, that is, immune cells. Here, we review the current knowledge on noncanonical TRAIL signaling. The biological outcomes of TRAIL signaling in immune and malignant cells are presented and explained, with a focus on the role of TRAIL for natural killer (NK) cell function. Furthermore, we highlight the technical difficulties in dissecting the precise molecular mechanisms involved in the switch between apoptotic and nonapoptotic TRAIL signaling. Finally, we discuss the consequences thereof for a therapeutic manipulation of TRAIL in cancer and possible approaches to bypass these difficulties.
Collapse
Affiliation(s)
| | - Nadia Corazza
- Institute of Pathology, University of Bern, Switzerland
| | - Olivier Micheau
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
| | | |
Collapse
|
23
|
Maixner N, Pecht T, Haim Y, Chalifa-Caspi V, Goldstein N, Tarnovscki T, Liberty IF, Kirshtein B, Golan R, Berner O, Monsonego A, Bashan N, Blüher M, Rudich A. A TRAIL-TL1A Paracrine Network Involving Adipocytes, Macrophages, and Lymphocytes Induces Adipose Tissue Dysfunction Downstream of E2F1 in Human Obesity. Diabetes 2020; 69:2310-2323. [PMID: 32732304 PMCID: PMC7576574 DOI: 10.2337/db19-1231] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 07/24/2020] [Indexed: 12/19/2022]
Abstract
Elevated expression of E2F1 in adipocyte fraction of human visceral adipose tissue (hVAT) associates with a poor cardiometabolic profile. We hypothesized that beyond directly activating autophagy and MAP3K5 (ASK)-MAP kinase signaling, E2F1 governs a distinct transcriptome that contributes to adipose tissue and metabolic dysfunction in obesity. We performed RNA sequencing of hVAT samples from age-, sex-, and BMI-matched patients, all obese, whose visceral E2F1 protein expression was either high (E2F1high) or low (E2F1low). Tumor necrosis factor superfamily (TNFSF) members, including TRAIL (TNFSF10), TL1A (TNFSF15), and their receptors, were enriched in E2F1high While TRAIL was equally expressed in adipocytes and stromal vascular fraction (SVF), TL1A was mainly expressed in SVF, and TRAIL-induced TL1A was attributed to CD4+ and CD8+ subclasses of hVAT T cells. In human adipocytes, TL1A enhanced basal and impaired insulin-inhibitable lipolysis and altered adipokine secretion, and in human macrophages it induced foam cell biogenesis and M1 polarization. Two independent human cohorts confirmed associations between TL1A and TRAIL expression in hVAT and higher leptin and IL6 serum concentrations, diabetes status, and hVAT-macrophage lipid content. Jointly, we propose an intra-adipose tissue E2F1-associated TNFSF paracrine loop engaging lymphocytes, macrophages, and adipocytes, ultimately contributing to adipose tissue dysfunction in obesity.
Collapse
Affiliation(s)
- Nitzan Maixner
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The Joyce & Irving Goldman Medical School, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tal Pecht
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yulia Haim
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vered Chalifa-Caspi
- The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nir Goldstein
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tania Tarnovscki
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Idit F Liberty
- The Joyce & Irving Goldman Medical School, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Soroka Academic Medical Center, Beer-Sheva, Israel
| | - Boris Kirshtein
- The Joyce & Irving Goldman Medical School, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Soroka Academic Medical Center, Beer-Sheva, Israel
| | - Rachel Golan
- Department of Epidemiology and Preventive Medicine, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Omer Berner
- Department of Microbiology and Immunology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Monsonego
- Department of Microbiology and Immunology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nava Bashan
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The Joyce & Irving Goldman Medical School, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Assaf Rudich
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The National Institute of Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The Joyce & Irving Goldman Medical School, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
24
|
Lambrecht J, van Grunsven LA, Tacke F. Current and emerging pharmacotherapeutic interventions for the treatment of liver fibrosis. Expert Opin Pharmacother 2020; 21:1637-1650. [PMID: 32543284 DOI: 10.1080/14656566.2020.1774553] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Chronic liver disease is due to various causes of persistent liver damage and will eventually lead to the development of liver fibrosis. If no treatment is initiated, this condition may progress to cirrhosis and hepatocellular carcinoma. Current treatments comprise the elimination of the cause of injury, such as by lifestyle changes, alcohol abstinence, and antiviral agents. However, such etiology-driven therapy is often insufficient in patients with late-stage fibrosis/cirrhosis, therefore maintaining the need for efficient antifibrotic pharmacotherapeutic interventions. AREAS COVERED The authors discuss the recent advances in the development of antifibrotic drugs, which target various pathways of the fibrogenesis process, including cell death, inflammation, gut-liver axis, and myofibroblast activation. Due to the significant burden of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH), various agents which specifically target metabolic pathways and their related receptors/ligands have been developed. For some of them, e.g., obeticholic acid, advanced stage clinical trials indicate antifibrotic efficacy in NAFLD and NASH. EXPERT OPINION Significant advances have been made in the development of novel antifibrotic pharmacotherapeutics. The authors expect that the development of combinatorial therapies, which combine compounds that target various pathways of fibrosis progression, will have a major impact as future etiology-independent therapies.
Collapse
Affiliation(s)
- Joeri Lambrecht
- Liver Cell Biology Research Group, Vrije Universiteit Brussel , Brussels, Belgium
| | - Leo A van Grunsven
- Liver Cell Biology Research Group, Vrije Universiteit Brussel , Brussels, Belgium
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité University Medical Center , Berlin, Germany
| |
Collapse
|
25
|
Sarode P, Schaefer MB, Grimminger F, Seeger W, Savai R. Macrophage and Tumor Cell Cross-Talk Is Fundamental for Lung Tumor Progression: We Need to Talk. Front Oncol 2020; 10:324. [PMID: 32219066 PMCID: PMC7078651 DOI: 10.3389/fonc.2020.00324] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
Regardless of the promising results of certain immune checkpoint blockers, current immunotherapeutics have met a bottleneck concerning response rate, toxicity, and resistance in lung cancer patients. Accumulating evidence forecasts that the crosstalk between tumor and immune cells takes center stage in cancer development by modulating tumor malignancy, immune cell infiltration, and immune evasion in the tumor microenvironment (TME). Cytokines and chemokines secreted by this crosstalk play a major role in cancer development, progression, and therapeutic management. An increased infiltration of Tumor-associated macrophages (TAMs) was observed in most of the human cancers, including lung cancer. In this review, we emphasize the role of cytokines and chemokines in TAM-tumor cell crosstalk in the lung TME. Given the role of cytokines and chemokines in immunomodulation, we propose that TAM-derived cytokines and chemokines govern the cancer-promoting immune responses in the TME and offer a new immunotherapeutic option for lung cancer treatment.
Collapse
Affiliation(s)
- Poonam Sarode
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Martina Barbara Schaefer
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
26
|
Liang C, Cao H, Cao X. Tetrandrine can alleviate inflammation and delay the growth of lung cancer during low-dose radiotherapy of non-small cell lung cancer. BIOTECHNOL BIOTEC EQ 2020. [DOI: 10.1080/13102818.2020.1736951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Affiliation(s)
- Chenglei Liang
- Department of Pharmacy, Xuyi County Hospital of Traditional Chinese Medicine, Xuyi, Jiangsu, P.R. China
| | - Hu Cao
- Department of Pharmacy, Xuyi County Hospital of Traditional Chinese Medicine, Xuyi, Jiangsu, P.R. China
| | - Xiaopin Cao
- Department of Pharmacy, Xuyi County Hospital of Traditional Chinese Medicine, Xuyi, Jiangsu, P.R. China
| |
Collapse
|
27
|
LPS Inhibits Fatty Acid Absorption in Enterocytes through TNF-α Secreted by Macrophages. Cells 2019; 8:cells8121626. [PMID: 31842409 PMCID: PMC6953048 DOI: 10.3390/cells8121626] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/28/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023] Open
Abstract
Diarrhea, such as steatorrhea, could result from fat absorption disorders, which could be caused by many factors, including Escherichia coli infection. However, it is not clear how E. coli affects fatty acid absorption in animals. Lipopolysaccharide (LPS), as one of the main pathogenic components of E. coli, is the main cause of the virulence of E. coli. Therefore, we used LPS to explore the underlying mechanism of E. coli that causes the inhibition of fatty acid absorption in the intestine. In this study, we found that LPS caused apoptosis of intestinal epithelial cells in mice. Further, caspase-3 activation caused the inhibition of fatty acid absorption in the intestinal porcine enterocyte cell line (IPEC-J2). However, direct treatment of LPS did not induce any significant change in fatty acid absorption in IPEC-J2. We then prepared conditioned medium of LPS-treated porcine macrophage cell line (3D4/2) for incubating IPEC-J2, as LPS initiates inflammation by activating immune cells. The conditioned medium decreased fatty acid absorption and caspase-3 activation in IPEC-J2. While inhibiting the activation of caspase-3 in IPEC-J2, conditioned medium no longer caused serious deficiency of fatty acid absorption. As IL-1β, IL-6, and TNF-α in conditioned medium increase significantly, IPEC-J2 was treated with IL-1β, IL-6, and TNF-α, respectively. Only TNF-α induced caspase-3 activation in IPEC-J2. Reducing the secretion of TNF-α in 3D4/2, there was no obvious activation of caspase-3 in IPEC-J2, and fatty acid absorption recovered effectively. Based on the above results, we hold the opinion that LPS does not suppress fatty acid absorption directly in the intestine, but may work on macrophages that secrete cytokines, such as TNF-α, inducing caspase-3 activation and finally leading to the inhibition of fatty acid absorption in intestine.
Collapse
|
28
|
Yin M, Shen J, Yu S, Fei J, Zhu X, Zhao J, Zhai L, Sadhukhan A, Zhou J. Tumor-Associated Macrophages (TAMs): A Critical Activator In Ovarian Cancer Metastasis. Onco Targets Ther 2019; 12:8687-8699. [PMID: 31695427 PMCID: PMC6814357 DOI: 10.2147/ott.s216355] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAMs) that appear in every stage of cancer progression are usually tumor-promoting cells and are present abundantly in the tumor-associated microenvironment. In ovarian cancer, the overall and intratumoral M1/M2 ratio is a relatively efficient TAM parameter for predicting the prognosis of patients, especially for serous tissue type cancer. TAMs exhibit immunological checkpoint modulators, such as the B7 family and programmed death-ligand 1 (PD-L1), and play a key role in the development, metastasis and invasion of ovarian cancer, but the underlying mechanism is barely understood. Ovarian cancer is a severe gynecological malignancy with high mortality. Ovarian cancer-associated death can primarily be attributed to cancer metastasis. The majority of patients are diagnosed with wide dissemination in the peritoneum and omentum, limiting the effectiveness of surgery and chemotherapy. In addition, unlike other well-documented cancers, metastasis through vasculature is not a usual dissemination pathway in ovarian cancer. This review sheds light on TAMs and the main process and mechanism of ovarian cancer metastasis.
Collapse
Affiliation(s)
- Meichen Yin
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiayu Shen
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Shuqian Yu
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jing Fei
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaoqing Zhu
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiayao Zhao
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Lingyun Zhai
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Annapurna Sadhukhan
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
29
|
Sag D, Ayyildiz ZO, Gunalp S, Wingender G. The Role of TRAIL/DRs in the Modulation of Immune Cells and Responses. Cancers (Basel) 2019; 11:cancers11101469. [PMID: 31574961 PMCID: PMC6826877 DOI: 10.3390/cancers11101469] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/09/2019] [Accepted: 09/20/2019] [Indexed: 12/26/2022] Open
Abstract
Expression of TRAIL (tumor necrosis factor–related apoptosis–inducing ligand) by immune cells can lead to the induction of apoptosis in tumor cells. However, it becomes increasingly clear that the interaction of TRAIL and its death receptors (DRs) can also directly impact immune cells and influence immune responses. Here, we review what is known about the role of TRAIL/DRs in immune cells and immune responses in general and in the tumor microenvironment in particular.
Collapse
Affiliation(s)
- Duygu Sag
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Zeynep Ozge Ayyildiz
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Sinem Gunalp
- Department of Genome Sciences and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| | - Gerhard Wingender
- Izmir Biomedicine and Genome Center (IBG), 35340 Balcova/Izmir, Turkey.
- Department of Biomedicine and Health Technologies, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, 35340 Balcova/Izmir, Turkey.
| |
Collapse
|
30
|
de Looff M, de Jong S, Kruyt FAE. Multiple Interactions Between Cancer Cells and the Tumor Microenvironment Modulate TRAIL Signaling: Implications for TRAIL Receptor Targeted Therapy. Front Immunol 2019; 10:1530. [PMID: 31333662 PMCID: PMC6617985 DOI: 10.3389/fimmu.2019.01530] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/19/2019] [Indexed: 01/09/2023] Open
Abstract
Tumor necrosis factor (TNF) related apoptosis-inducing ligand (TRAIL) signaling is far more complex than initially anticipated and can lead to either anti- or protumorigenic effects, hampering the successful clinical use of therapeutic TRAIL receptor agonists. Cell autonomous resistance mechanisms have been identified in addition to paracrine factors that can modulate apoptosis sensitivity. The tumor microenvironment (TME), consisting of cellular and non-cellular components, is a source for multiple signals that are able to modulate TRAIL signaling in tumor and stromal cells. Particularly immune effector cells, also part of the TME, employ the TRAIL/TRAIL-R system whereby cell surface expressed TRAIL can activate apoptosis via TRAIL receptors on tumor cells, which is part of tumor immune surveillance. In this review we aim to dissect the impact of the TME on signaling induced by endogenous and exogenous/therapeutic TRAIL, thereby distinguishing different components of the TME such as immune effector cells, neutrophils, macrophages, and non-hematopoietic stromal cells. In addition, also non-cellular biochemical and biophysical properties of the TME are considered including mechanical stress, acidity, hypoxia, and glucose deprivation. Available literature thus far indicates that tumor-TME interactions are complex and often bidirectional leading to tumor-enhancing or tumor-reducing effects in a tumor model- and tumor type-dependent fashion. Multiple signals originating from different components of the TME simultaneously affect TRAIL receptor signaling. We conclude that in order to unleash the full clinical potential of TRAIL receptor agonists it will be necessary to increase our understanding of the contribution of different TME components on outcome of therapeutic TRAIL receptor activation in order to identify the most critical mechanism responsible for resistance, allowing the design of effective combination treatments.
Collapse
Affiliation(s)
- Margot de Looff
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Steven de Jong
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Frank A E Kruyt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
31
|
Iacona JR, Lutz CS. miR-146a-5p: Expression, regulation, and functions in cancer. WILEY INTERDISCIPLINARY REVIEWS-RNA 2019; 10:e1533. [PMID: 30895717 DOI: 10.1002/wrna.1533] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/14/2022]
Abstract
Cancer as we know it is actually an umbrella term for over 100 very unique malignancies in various tissues throughout the human body. Each type, and even subtype of cancer, has different genetic, epigenetic, and other cellular events responsible for malignant development and metastasis. Recent work has indicated that microRNAs (miRNAs) play a major role in these processes, sometimes by promoting cancer growth and other times by suppressing tumorigenesis. miRNAs are small, noncoding RNAs that negatively regulate expression of specific target genes. This review goes into an in-depth look at the most recent finding regarding the significance of one particular miRNA, miR-146a-5p, and its involvement in cancer. Target gene validation and pathway analysis have provided mechanistic insight into this miRNA's purpose in assorted tissues. Additionally, this review outlines novel findings that suggest miR-146a-5p may be useful as a noninvasive biomarker and as a targeted therapeutic in several cancers. This article is categorized under: RNA in Disease and Development > RNA in Disease Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs.
Collapse
Affiliation(s)
- Joseph R Iacona
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School and the School of Graduate Studies, Health Sciences Campus - Newark, Newark, New Jersey
| | - Carol S Lutz
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School and the School of Graduate Studies, Health Sciences Campus - Newark, Newark, New Jersey
| |
Collapse
|
32
|
The Interplay between MicroRNAs and Cellular Components of Tumour Microenvironment (TME) on Non-Small-Cell Lung Cancer (NSCLC) Progression. J Immunol Res 2019; 2019:3046379. [PMID: 30944831 PMCID: PMC6421779 DOI: 10.1155/2019/3046379] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/26/2018] [Accepted: 01/06/2019] [Indexed: 12/30/2022] Open
Abstract
Cellular components of the tumour microenvironment (TME) are recognized to regulate the hallmarks of cancers including tumour proliferation, angiogenesis, invasion, and metastasis, as well as chemotherapeutic resistance. The linkage between miRNA, TME, and the development of the hallmarks of cancer makes miRNA-mediated regulation of TME a potential therapeutic strategy to complement current cancer therapies. Despite significant advances in cancer therapy, lung cancer remains the deadliest form of cancer among males in the world and has overtaken breast cancer as the most fatal cancer among females in more developed countries. Therefore, there is an urgent need to develop more effective treatments for NSCLC, which is the most common type of lung cancer. Hence, this review will focus on current literature pertaining to antitumour or protumourigenic effects elicited by nonmalignant stromal cells of TME in NSCLC through miRNA regulation as well as current status and future prospects of miRNAs as therapeutic agents or targets to regulate TME in NSCLC.
Collapse
|
33
|
Collison AM, Li J, de Siqueira AP, Lv X, Toop HD, Morris JC, Starkey MR, Hansbro PM, Zhang J, Mattes J. TRAIL signals through the ubiquitin ligase MID1 to promote pulmonary fibrosis. BMC Pulm Med 2019; 19:31. [PMID: 30732588 PMCID: PMC6367767 DOI: 10.1186/s12890-019-0786-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/10/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) has previously been demonstrated to play a pro-inflammatory role in allergic airways disease and COPD through the upregulation of the E3 ubiquitin ligase MID1 and the subsequent deactivation of protein phosphatase 2A (PP2A). METHODS Biopsies were taken from eight IPF patients presenting to the Second Affiliated Hospital of Jilin University, China between January 2013 and February 2014 with control samples obtained from resected lung cancers. Serum TRAIL, MID1 protein and PP2A activity in biopsies, and patients' lung function were measured. Wild type and TRAIL deficient Tnfsf10-/- BALB/c mice were administered bleomycin to induce fibrosis and some groups were treated with the FTY720 analogue AAL(s) to activate PP2A. Mouse fibroblasts were treated with recombinant TRAIL and fibrotic responses were assessed. RESULTS TRAIL in serum and MID1 protein levels in biopsies from IPF patients were increased compared to controls. MID1 levels were inversely associated while PP2A activity levels correlated with DLco. Tnfsf10-/- and mice treated with the PP2A activator AAL(s) were largely protected against bleomycin-induced reductions in lung function and fibrotic changes. Addition of recombinant TRAIL to mouse fibroblasts in-vitro increased collagen production which was reversed by PP2A activation with AAL(s). CONCLUSION TRAIL signalling through MID1 deactivates PP2A and promotes fibrosis with corresponding lung function decline. This may provide novel therapeutic targets for IPF.
Collapse
Affiliation(s)
- Adam M. Collison
- Experimental and Translational Respiratory Medicine Group, Level 2 East, Hunter Medical Research Institute, School of Medicine and Public Health, Faculty of Health, University of Newcastle, Callaghan, NSW 2308 Australia
- Priority Research Centre GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Junyao Li
- Experimental and Translational Respiratory Medicine Group, Level 2 East, Hunter Medical Research Institute, School of Medicine and Public Health, Faculty of Health, University of Newcastle, Callaghan, NSW 2308 Australia
- Priority Research Centre GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Jilin University, Changchun, Jilin, 130041 People’s Republic of China
| | - Ana Pereira de Siqueira
- Experimental and Translational Respiratory Medicine Group, Level 2 East, Hunter Medical Research Institute, School of Medicine and Public Health, Faculty of Health, University of Newcastle, Callaghan, NSW 2308 Australia
- Priority Research Centre GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Xuejiao Lv
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Jilin University, Changchun, Jilin, 130041 People’s Republic of China
| | - Hamish D. Toop
- School of Chemistry, University of New South Wales, Sydney, New South Wales Australia
| | - Jonathan C. Morris
- School of Chemistry, University of New South Wales, Sydney, New South Wales Australia
| | - Malcolm R. Starkey
- Priority Research Centre GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
- Priority Research Centre for Healthy Lungs, The University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Philip M. Hansbro
- Priority Research Centre for Healthy Lungs, The University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Jie Zhang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Jilin University, Changchun, Jilin, 130041 People’s Republic of China
| | - Joerg Mattes
- Experimental and Translational Respiratory Medicine Group, Level 2 East, Hunter Medical Research Institute, School of Medicine and Public Health, Faculty of Health, University of Newcastle, Callaghan, NSW 2308 Australia
- Priority Research Centre GrowUpWell, The University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
- Paediatric Respiratory & Sleep Medicine Department, Newcastle Children’s Hospital, Kaleidoscope, Newcastle, Australia
| |
Collapse
|
34
|
Toka FN, Dunaway K, Smaltz F, Szulc-Dąbrowska L, Drnevich J, Mielcarska MB, Bossowska-Nowicka M, Schweizer M. Bacterial and viral pathogen-associated molecular patterns induce divergent early transcriptomic landscapes in a bovine macrophage cell line. BMC Genomics 2019; 20:15. [PMID: 30621583 PMCID: PMC6323673 DOI: 10.1186/s12864-018-5411-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/26/2018] [Indexed: 02/08/2023] Open
Abstract
Background Pathogens stimulate immune functions of macrophages. Macrophages are a key sentinel cell regulating the response to pathogenic ligands and orchestrating the direction of the immune response. Our study aimed at investigating the early transcriptomic changes of bovine macrophages (Bomacs) in response to stimulation with CpG DNA or polyI:C, representing bacterial and viral ligands respectively, and performed transcriptomics by RNA sequencing (RNASeq). KEGG, GO and IPA analytical tools were used to reconstruct pathways, networks and to map out molecular and cellular functions of differentially expressed genes (DE) in stimulated cells. Results A one-way ANOVA analysis of RNASeq data revealed significant differences between the CpG DNA and polyI:C-stimulated Bomac. Of the 13,740 genes mapped to the bovine genome, 2245 had p-value ≤0.05, deemed as DE. At 6 h post stimulation of Bomac, poly(I:C) induced a very different transcriptomic profile from that induced by CpG DNA. Whereas, 347 genes were upregulated and 210 downregulated in response to CpG DNA, poly(I:C) upregulated 761 genes and downregulated 414 genes. The topmost DE genes in poly(I:C)-stimulated cells had thousand-fold changes with highly significant p-values, whereas in CpG DNA stimulated cells had 2–5-fold changes with less stringent p-values. The highest DE genes in both stimulations belonged to the TNF superfamily, TNFSF18 (CpG) and TNFSF10 (poly(I:C)) and in both cases the lowest downregulated gene was CYP1A1. CpG DNA highly induced canonical pathways that are unrelated to immune response in Bomac. CpG DNA influenced expression of genes involved in molecular and cellular functions in free radical scavenging. By contrast, poly(I:C) highly induced exclusively canonical pathways directly related to antiviral immune functions mediated by interferon signalling genes. The transcriptomic profile after poly(I:C)-stimulation was consistent with induction of TLR3 signalling. Conclusion CpG DNA and poly(I:C) induce different early transcriptional landscapes in Bomac, but each is suited to a specific function of macrophages during interaction with pathogens. Poly(I:C) influenced antiviral response genes, whereas CpG DNA influenced genes important for phagocytic processes. Poly(I:C) was more potent in setting the inflammatory landscape desirable for an efficient immune response against virus infection. Electronic supplementary material The online version of this article (10.1186/s12864-018-5411-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Felix N Toka
- Department of Biomedical Sciences, Center for Integrative Mammalian Research, Ross University School of Veterinary Medicine, 00-334, Basseterre, Saint Kitts and Nevis. .,Department of Preclinical Sciences, Faculty of Veterinary Medicine, SGGW, Warsaw, Poland.
| | - Kiera Dunaway
- Department of Biomedical Sciences, Center for Integrative Mammalian Research, Ross University School of Veterinary Medicine, 00-334, Basseterre, Saint Kitts and Nevis
| | - Felicia Smaltz
- Department of Biomedical Sciences, Center for Integrative Mammalian Research, Ross University School of Veterinary Medicine, 00-334, Basseterre, Saint Kitts and Nevis
| | - Lidia Szulc-Dąbrowska
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, SGGW, Warsaw, Poland
| | - Jenny Drnevich
- HPCBio and the Carver Biotechnology Center, University of Illinois, Champaign, IL, USA
| | | | | | - Matthias Schweizer
- Institute of Virology and Immunology, Federal Food Safety and Veterinary Office FSVO, Bern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
35
|
van Dalen FJ, van Stevendaal MHME, Fennemann FL, Verdoes M, Ilina O. Molecular Repolarisation of Tumour-Associated Macrophages. Molecules 2018; 24:molecules24010009. [PMID: 30577495 PMCID: PMC6337345 DOI: 10.3390/molecules24010009] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 12/21/2022] Open
Abstract
The tumour microenvironment (TME) is composed of extracellular matrix and non-mutated cells supporting tumour growth and development. Tumour-associated macrophages (TAMs) are among the most abundant immune cells in the TME and are responsible for the onset of a smouldering inflammation. TAMs play a pivotal role in oncogenic processes as tumour proliferation, angiogenesis and metastasis, and they provide a barrier against the cytotoxic effector function of T lymphocytes and natural killer (NK) cells. However, TAMs are highly plastic cells that can adopt either pro- or anti-inflammatory roles in response to environmental cues. Consequently, TAMs represent an attractive target to recalibrate immune responses in the TME. Initial TAM-targeted strategies, such as macrophage depletion or disruption of TAM recruitment, have shown beneficial effects in preclinical models and clinical trials. Alternatively, reprogramming TAMs towards a proinflammatory and tumouricidal phenotype has become an attractive strategy in immunotherapy. This work summarises the molecular wheelwork of macrophage biology and presents an overview of molecular strategies to repolarise TAMs in immunotherapy.
Collapse
Affiliation(s)
- Floris J van Dalen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.
| | - Marleen H M E van Stevendaal
- Department of Bio-Organic Chemistry, Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands.
| | - Felix L Fennemann
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.
| | - Olga Ilina
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands.
| |
Collapse
|
36
|
Tan X, Zhang Z, Yao H, Shen L. RETRACTED: Tim-4 promotes the growth of colorectal cancer by activating angiogenesis and recruiting tumor-associated macrophages via the PI3K/AKT/mTOR signaling pathway. Cancer Lett 2018; 436:119-128. [PMID: 30118845 DOI: 10.1016/j.canlet.2018.08.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/14/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the authors. After publication, the authors noted unreliable data in the experimental validations. Specifically, representative images of IHC staining for CD31 and α-SMA in CT26-Vector and CT26-Tim-4 tumors in Fig. 2C and the expression of MMP9, VEGF and LOX in CT26-Tim-4 and CT26-Vector tumors was assessed by immunohistochemistry in Fig. 2E; incorrect photos were provided due to file placement confusion. All authors “agree with this retraction and deeply regret these errors and apologize to the editorial board and readers for any inconvenience caused.” Additionally, after publication, the journal was made aware of comments in relation to this article (https://pubpeer.com/publications/D191AE022DDD7B810564039CB16395). The authors have not responded to the journal request to respond to these comments.
Collapse
Affiliation(s)
- Xiao Tan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Zhongqiang Zhang
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Hongliang Yao
- Department of General Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
37
|
Dorn S, Schoergenhofer C, Krainer M, Müller M, Jilma B. LUBAC and ABIN-1 Modulate TRAIL-Based NF-κB Induction in Human Embryonic Kidney 293 Cells. Biores Open Access 2018; 7:81-89. [PMID: 29862142 PMCID: PMC5982153 DOI: 10.1089/biores.2018.0006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is known to activate the canonical NF-κB pathway similar to TNF. The exact mechanism of the entire signaling cascade is still under investigation. The involvement of linear ubiquitylation as upregulating component has already been shown recently in some cell lines, but not in human embryonic kidney 293 (HEK293) cells. The downregulating function of the ABIN-1 (A20 binding and inhibitor of NF-κB) as linear ubiquitylation antagonist has been shown in combination with some NF-κB-inducing pathways, but not with TRAIL. We performed luciferase and western blot assays using HEK293 cells stimulated with either TRAIL (or TNF as a control) to analyze the involvement of linear ubiquitin chain assembly complex (LUBAC) components and the impact of ABIN-1 and ABIN-1-MAD (truncated form without A20 binding site) on NF-κB signaling. For overexpression experiments, we added plasmids of ABIN-1 and ABIN-1-MAD or LUBAC components HOIP, HOIL-1, or SHARPIN (single and combinations). For downregulation experiments five pairs of either SHARPIN, HOIL-1, or HOIP targeting miRNAs or one miRNA for ABIN-1 were designed and added. ABIN-1 and its truncated form ABIN-1-MAD reduced the NF-κB induction significantly indicating its involvement as antagonist (independent of deubiquitinase A20) of linear ubiquitylation in TRAIL-induced NF-κB signaling. In opposition, knockdown of ABIN-1 using a specific ABIN-1 miRNA led a clear increase of NF-κB signaling. Addition of single LUBAC components or combinations (except for SHARPIN with HOIL-1) resulted in clearly stronger NF-κB inductions. MiRNAs targeting LUBAC components significantly reduced NF-κB activation. Thus, in HEK293 cells linear ubiquitylation by LUBAC critically upregulates and ABIN-1 downregulates TRAIL-induced NF-κB signaling and may be interesting targets for future pathological therapies.
Collapse
Affiliation(s)
- Sebastian Dorn
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Michael Krainer
- Clinical Division of Oncology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Markus Müller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
38
|
Redirecting tumor-associated macrophages to become tumoricidal effectors as a novel strategy for cancer therapy. Oncotarget 2018; 8:48436-48452. [PMID: 28467800 PMCID: PMC5564660 DOI: 10.18632/oncotarget.17061] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 03/22/2017] [Indexed: 12/25/2022] Open
Abstract
Cancer research in recent decades has highlighted the potential influence of the tumor microenvironment on the progression and metastasis of most known cancer types. Within the established microenvironment, tumor-associated macrophages (TAMs) are one of the most abundant and crucial non-neoplastic cell types. The polarization of macrophages into tumor-suppressive M1 or tumor-promoting M2 types is a fundamental event in the establishment of the tumor microenvironment. Although ample evidence indicates that TAMs are primarily M2 polarized, the mechanisms responsible for the regulation and maintenance of M1 and M2 polarization imbalance remain unclear. The manipulation of this critical axis through three main approaches may provide new strategies for cancer therapy - (I) specific interference with M2-like TAM survival or inhibiting their signaling cascades, (II) repression of macrophage recruitment to tumors, and (III) repolarization of tumor-promoting M2-like TAMs to a tumoricidal M1-like phenotype. This review summarizes current strategies for cancer intervention via manipulation of macrophage polarization, with particular focus on composition of the tumor microenvironment and its influence on cancer progression and metastasis. It is clear that additional fundamental and preclinical research is required to confirm the efficacy and practicality of this novel and promising strategy for treating cancer.
Collapse
|
39
|
Bardi GT, Smith MA, Hood JL. Melanoma exosomes promote mixed M1 and M2 macrophage polarization. Cytokine 2018; 105:63-72. [PMID: 29459345 DOI: 10.1016/j.cyto.2018.02.002] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/18/2022]
Abstract
Macrophages are key participants in melanoma growth and survival. In general, macrophages can be classified as M1 or M2 activation phenotypes. Increasing evidence demonstrates that melanoma exosomes also facilitate tumor survival and metastasis. However, the role of melanoma exosomes in directly influencing macrophage function is poorly understood. Herein, we investigated the hypothesis that natural melanoma exosomes might directly influence macrophage polarization. To explore this hypothesis, ELISA, RT-qPCR, and macrophage functional studies were performed in vitro using an established source of melanoma exosomes (B16-F10). ELISA results for melanoma exosome induction of common M1 and M2 cytokines in RAW 264.7 macrophages, revealed that melanoma exosomes do not polarize macrophages exclusively in the M1 or M2 direction. Melanoma exosomes induced the M1 and M2 representative cytokines TNF-α and IL-10 respectively. Further assessment, using an RT-qPCR array with RAW 264.7 and primary macrophages, confirmed and extended the ELISA findings. Upregulation of markers common to both M1 and M2 polarization phenotypes included CCL22, IL-12B, IL-1β, IL-6, i-NOS, and TNF-α. The M2 cytokine TGF-β was upregulated in primary but not RAW 264.7 macrophages. Pro-tumor functions have been attributed to each of these markers. Macrophage functional assays demonstrated a trend toward increased i-NOS (M1) to arginase (M2) activity. Collectively, the results provide the first evidence that melanoma exosomes can induce a mixed M1 and M2 pro-tumor macrophage activation phenotype.
Collapse
Affiliation(s)
- Gina T Bardi
- University of Louisville, Department of Pharmacology and Toxicology, & James Graham Brown Cancer Center, Clinical and Translational Research Building, 505 South Hancock Street, Louisville, KY 40202, United States.
| | - Mary Ann Smith
- University of Louisville, Department of Pharmacology and Toxicology, & James Graham Brown Cancer Center, Clinical and Translational Research Building, 505 South Hancock Street, Louisville, KY 40202, United States.
| | - Joshua L Hood
- University of Louisville, Department of Pharmacology and Toxicology, & James Graham Brown Cancer Center, Clinical and Translational Research Building, 505 South Hancock Street, Louisville, KY 40202, United States.
| |
Collapse
|
40
|
Hirsova P, Weng P, Salim W, Bronk SF, Griffith TS, Ibrahim SH, Gores GJ. TRAIL Deletion Prevents Liver, but Not Adipose Tissue, Inflammation during Murine Diet-Induced Obesity. Hepatol Commun 2017; 1:648-662. [PMID: 29124251 PMCID: PMC5673124 DOI: 10.1002/hep4.1069] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tumor necrosis factor‐related apoptosis‐inducing ligand (TRAIL) and its cognate receptor(s) are up‐regulated in human and murine nonalcoholic steatohepatitis (NASH); however, the consequence of this enhanced expression on NASH pathogenesis remains unclear. TRAIL may either accentuate liver injury by promoting hepatic steatosis and inflammation or it may mitigate the disease process by improving systemic insulin resistance and averting hepatic fibrosis. Herein, we investigated the role of TRAIL in an obesity‐induced murine model of NASH. C57BL/6 wild‐type mice and Trail–/– mice were placed on a 20‐week standard chow or a high‐fat, high‐fructose, and high‐cholesterol (FFC) diet, which induces obesity, insulin resistance, and NASH. Metabolic phenotype, liver injury, inflammation and fibrosis, and adipose tissue homeostasis were examined. FFC diet‐fed Trail–/– mice displayed no difference in weight gain and metabolic profile when compared to wild‐type mice on the same diet. All FFC‐fed mice developed significant hepatic steatosis, which was attenuated in Trail–/– mice. TRAIL deficiency also significantly decreased FFC diet‐induced liver injury as manifested by reduced serum alanine aminotransferase values, hepatic terminal deoxynucleotidyl transferase‐mediated deoxyuridine triphosphate nick‐end labeling‐positive cells, and macrophage‐associated inflammation. FFC diet‐associated hepatic stellate cell activation and hepatic collagen deposition were also abrogated in Trail–/– mice. In contrast to the liver, TRAIL deletion did not improve FFC diet‐induced adipose tissue injury and inflammation and actually aggravated insulin resistance. Conclusion: NASH pathogenesis may be dissociated from other features of the metabolic syndrome, and liver‐targeted inhibition of TRAIL signaling may be salutary. (Hepatology Communications 2017;1:648–662)
Collapse
Affiliation(s)
- Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Peggy Weng
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Warda Salim
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Steven F Bronk
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Thomas S Griffith
- Department of Urology, University of Minnesota, Minneapolis, MN 55455, USA.,Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA.,Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Samar H Ibrahim
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA.,Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
41
|
Guicciardi ME, Krishnan A, Bronk SF, Hirsova P, Griffith TS, Gores GJ. Biliary tract instillation of a SMAC mimetic induces TRAIL-dependent acute sclerosing cholangitis-like injury in mice. Cell Death Dis 2017; 8:e2535. [PMID: 28055006 PMCID: PMC5386369 DOI: 10.1038/cddis.2016.459] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 12/19/2022]
Abstract
Primary sclerosing cholangitis (PSC) is a cholestatic liver disease of unknown etiopathogenesis characterized by fibrous cholangiopathy of large and small bile ducts. Systemic administration of a murine TNF-related apoptosis-inducing ligand (TRAIL) receptor agonist induces a sclerosing cholangitis injury in C57BL/6 mice, suggesting endogenous TRAIL may contribute to sclerosing cholangitis syndromes. Cellular inhibitor of apoptosis proteins (cIAP-1 and cIAP-2) are negative regulators of inflammation and TRAIL receptor signaling. We hypothesized that if endogenous TRAIL promotes sclerosing cholangitis, then cIAP depletion should also induce this biliary tract injury. Herein, we show that cIAP protein levels are reduced in the interlobular bile ducts of human PSC livers. Downregulation of cIAPs in normal human cholangiocytes in vitro by use of a SMAC mimetic (SM) induces moderate, ripoptosome-mediated apoptosis and RIP1-independent upregulation of proinflammatory cytokines and chemokines. Cytokine and chemokine expression was mediated by the non-canonical activation of NF-κB. To investigate whether downregulation of cIAPs is linked to generation of a PSC-like phenotype, an SM was directly instilled into the mouse biliary tree. Twelve hours after biliary instillation, TUNEL-positive cholangiocytes were identified; 5 days later, PSC-like changes were observed in the SM-treated mice, including a fibrous cholangiopathy of the interlobular bile ducts, portal inflammation, significant elevation of serum markers of cholestasis and cholangiographic evidence of intrahepatic biliary tract injury. In contrast, TRAIL and TRAIL-receptor deficient mice showed no sign of cholangiopathy following SM intrabiliary injection. We conclude that in vivo antagonism of cIAPs in mouse biliary epithelial cells is sufficient to trigger cholangiocytes apoptosis and a proinflammatory response resulting in a fibrous cholangiopathy resembling human sclerosing cholangitis. Therefore, downregulation of cIAPs in PSC cholangiocytes may contribute to the development of the disease. Our results also indicate that inhibition of TRAIL signaling pathways may be beneficial in the treatment of PSC.
Collapse
Affiliation(s)
- Maria Eugenia Guicciardi
- Division of Gastroenterology and Hepatology, Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA
| | - Anuradha Krishnan
- Division of Gastroenterology and Hepatology, Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA
| | - Steven F Bronk
- Division of Gastroenterology and Hepatology, Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA
| | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA
| | - Thomas S Griffith
- Department of Urology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
42
|
Marks DL, Olson RL, Fernandez-Zapico ME. Epigenetic control of the tumor microenvironment. Epigenomics 2016; 8:1671-1687. [PMID: 27700179 DOI: 10.2217/epi-2016-0110] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stromal cells of the tumor microenvironment have been shown to play important roles in both supporting and limiting cancer growth. The altered phenotype of tumor-associated stromal cells (fibroblasts, immune cells, endothelial cells etc.) is proposed to be mainly due to epigenetic dysregulation of gene expression; however, only limited studies have probed the roles of epigenetic mechanisms in the regulation of stromal cell function. We review recent studies demonstrating how specific epigenetic mechanisms (DNA methylation and histone post-translational modification-based gene expression regulation, and miRNA-mediated translational regulation) drive aspects of stromal cell phenotype, and discuss the implications of these findings for treatment of malignancies. We also summarize the effects of epigenetic mechanism-targeted drugs on stromal cells and discuss the consideration of the microenvironment response in attempts to use these drugs for cancer treatment.
Collapse
Affiliation(s)
- David L Marks
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Rachel Lo Olson
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.,University of Minnesota Rochester, Rochester, MN 55904, USA
| | | |
Collapse
|
43
|
Hirsova P, Ibrahim SH, Krishnan A, Verma VK, Bronk SF, Werneburg NW, Charlton MR, Shah VH, Malhi H, Gores GJ. Lipid-Induced Signaling Causes Release of Inflammatory Extracellular Vesicles From Hepatocytes. Gastroenterology 2016; 150:956-67. [PMID: 26764184 PMCID: PMC4808464 DOI: 10.1053/j.gastro.2015.12.037] [Citation(s) in RCA: 385] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 12/24/2015] [Accepted: 12/29/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Hepatocyte cellular dysfunction and death induced by lipids and macrophage-associated inflammation are characteristics of nonalcoholic steatohepatitis (NASH). The fatty acid palmitate can activate death receptor 5 (DR5) on hepatocytes, leading to their death, but little is known about how this process contributes to macrophage-associated inflammation. We investigated whether lipid-induced DR5 signaling results in the release of extracellular vesicles (EVs) from hepatocytes, and whether these can induce an inflammatory macrophage phenotype. METHODS Primary mouse and human hepatocytes and Huh7 cells were incubated with palmitate, its metabolite lysophosphatidylcholine, or diluent (control). The released EV were isolated, characterized, quantified, and applied to macrophages. C57BL/6 mice were placed on chow or a diet high in fat, fructose, and cholesterol to induce NASH. Some mice also were given the ROCK1 inhibitor fasudil; 2 weeks later, serum EVs were isolated and characterized by immunoblot and nanoparticle-tracking analyses. Livers were collected and analyzed by histology, immunohistochemistry, and quantitative polymerase chain reaction. RESULTS Incubation of primary hepatocytes and Huh7 cells with palmitate or lysophosphatidylcholine increased their release of EVs, compared with control cells. This release was reduced by inactivating mediators of the DR5 signaling pathway or rho-associated, coiled-coil-containing protein kinase 1 (ROCK1) inhibition. Hepatocyte-derived EVs contained tumor necrosis factor-related apoptosis-inducing ligand and induced expression of interleukin 1β and interleukin 6 messenger RNAs in mouse bone marrow-derived macrophages. Activation of macrophages required DR5 and receptor-interacting protein kinase 1. Administration of the ROCK1 inhibitor fasudil to mice with NASH reduced serum levels of EVs; this reduction was associated with decreased liver injury, inflammation, and fibrosis. CONCLUSIONS Lipids, which stimulate DR5, induce release of hepatocyte EVs, which activate an inflammatory phenotype in macrophages. Strategies to inhibit ROCK1-dependent release of EVs by hepatocytes might be developed for the treatment of patients with NASH.
Collapse
|