1
|
Ganguly N, Das T, Bhuniya A, Guha I, Chakravarti M, Dhar S, Sarkar A, Bera S, Dhar J, Dasgupta S, Saha A, Ghosh T, Das J, Sk UH, Banerjee S, Laskar S, Bose A, Baral R. Neem leaf glycoprotein binding to Dectin-1 receptors on dendritic cell induces type-1 immunity through CARD9 mediated intracellular signal to NFκB. Cell Commun Signal 2024; 22:237. [PMID: 38649988 PMCID: PMC11036628 DOI: 10.1186/s12964-024-01576-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/16/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND A water-soluble ingredient of mature leaves of the tropical mahogany 'Neem' (Azadirachta indica), was identified as glycoprotein, thus being named as 'Neem Leaf Glycoprotein' (NLGP). This non-toxic leaf-component regressed cancerous murine tumors (melanoma, carcinoma, sarcoma) recurrently in different experimental circumstances by boosting prime antitumor immune attributes. Such antitumor immunomodulation, aid cytotoxic T cell (Tc)-based annihilation of tumor cells. This study focused on identifying and characterizing the signaling gateway that initiate this systemic immunomodulation. In search of this gateway, antigen-presenting cells (APCs) were explored, which activate and induce the cytotoxic thrust in Tc cells. METHODS Six glycoprotein-binding C-type lectins found on APCs, namely, MBR, Dectin-1, Dectin-2, DC-SIGN, DEC205 and DNGR-1 were screened on bone marrow-derived dendritic cells from C57BL/6 J mice. Fluorescence microscopy, RT-PCR, flow cytometry and ELISA revealed Dectin-1 as the NLGP-binding receptor, followed by verifications through RNAi. Following detection of β-Glucans in NLGP, their interactions with Dectin-1 were explored in silico. Roles of second messengers and transcription factors in the downstream signal were studied by co-immunoprecipitation, western blotting, and chromatin-immunoprecipitation. Intracellularization of FITC-coupled NLGP was observed by processing confocal micrographs of DCs. RESULTS Considering extents of hindrance in NLGP-driven transcription rates of the cytokines IL-10 and IL-12p35 by receptor-neutralization, Dectin-1 receptors on dendritic cells were found to bind NLGP through the ligand's peripheral β-Glucan chains. The resulting signal phosphorylates PKCδ, forming a trimolecular complex of CARD9, Bcl10 and MALT1, which in turn activates the canonical NFκB-pathway of transcription-regulation. Consequently, the NFκB-heterodimer p65:p50 enhances Il12a transcription and the p50:p50 homodimer represses Il10 transcription, bringing about a cytokine-based systemic-bias towards type-1 immune environment. Further, NLGP gets engulfed within dendritic cells, possibly through endocytic activities of Dectin-1. CONCLUSION NLGP's binding to Dectin-1 receptors on murine dendritic cells, followed by the intracellular signal, lead to NFκB-mediated contrasting regulation of cytokine-transcriptions, initiating a pro-inflammatory immunopolarization, which amplifies further by the responding immune cells including Tc cells, alongside their enhanced cytotoxicity. These insights into the initiation of mammalian systemic immunomodulation by NLGP at cellular and molecular levels, may help uncovering its mode of action as a novel immunomodulator against human cancers, following clinical trials.
Collapse
Affiliation(s)
- Nilanjan Ganguly
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Tapasi Das
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Avishek Bhuniya
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Ipsita Guha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Mohona Chakravarti
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Sukanya Dhar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Anirban Sarkar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Saurav Bera
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Jesmita Dhar
- Jubilant Biosys Limited, 96, Digital Park Rd, Yesvantpur Industrial Suburb, Bengaluru, Karnataka, 560022, India
| | - Shayani Dasgupta
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Akata Saha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Tithi Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Juhina Das
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Ugir Hossain Sk
- Department of Clinical and Translational Research, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Saptak Banerjee
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Subrata Laskar
- Department of Chemistry, University of Burdwan, Burdwan, West Bengal, 713104, India
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India.
- Department of Pharmaceutical Technology-Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER),-S.A.S. Nagar, Mohali, Punjab, 160062, India.
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India.
| |
Collapse
|
2
|
Tang L, Xu H, Wu T, Wu W, Lu Y, Gu J, Wang X, Zhou M, Chen Q, Sun X, Cai H. Advances in tumor microenvironment and underlying molecular mechanisms of bladder cancer: a systematic review. Discov Oncol 2024; 15:111. [PMID: 38602556 PMCID: PMC11009183 DOI: 10.1007/s12672-024-00902-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/21/2024] [Indexed: 04/12/2024] Open
Abstract
Bladder cancer is one of the most frequent malignant tumors of the urinary system. The prevalence of bladder cancer among men and women is roughly 5:2, and both its incidence and death have been rising steadily over the past few years. At the moment, metastasis and recurrence of advanced bladder cancer-which are believed to be connected to the malfunction of multigene and multilevel cell signaling network-remain the leading causes of bladder cancer-related death. The therapeutic treatment of bladder cancer will be greatly aided by the elucidation of these mechanisms. New concepts for the treatment of bladder cancer have been made possible by the advancement of research technologies and a number of new treatment options, including immunotherapy and targeted therapy. In this paper, we will extensively review the development of the tumor microenvironment and the possible molecular mechanisms of bladder cancer.
Collapse
Affiliation(s)
- Liu Tang
- Department of Nursing, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Haifei Xu
- Department of Urology, Nantong Tumor Hospital and Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Tong Wu
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China
| | - Wenhao Wu
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China
| | - Yuhao Lu
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China
| | - Jijia Gu
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China
| | - Xiaoling Wang
- Department of Urology, Nantong Tumor Hospital and Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Mei Zhou
- Department of Nursing, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China.
| | - Qiuyang Chen
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China.
| | - Xuan Sun
- Department of Radiology, Nanjing Medical University The Fourth School of Clinical Medicine, Nanjing, Jiangsu, China.
| | - Hongzhou Cai
- Department of Urology, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China.
| |
Collapse
|
3
|
Soliman AH, Youness RA, Sebak AA, Handoussa H. Phytochemical-derived tumor-associated macrophage remodeling strategy using Phoenix dactylifera L. boosted photodynamic therapy in melanoma via H19/iNOS/PD-L1 axis. Photodiagnosis Photodyn Ther 2023; 44:103792. [PMID: 37689125 DOI: 10.1016/j.pdpdt.2023.103792] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND The tumor microenvironment (TME) represents a barrier to PDT efficacy among melanoma patients. The aim of this study is to employ a novel muti-tactic TME-remodeling strategy via repolarization of tumor-associated macrophages (TAMs), the main TME immune cells in melanoma, from the pro-tumor M2 into the antitumor M1 phenotype using Phoenix dactylifera L. (date palm) in combination with PDT. METHODS Screening of different date cultivars was employed to choose extracts of selective toxicity to melanoma and TAMs, not normal macrophages. Potential extracts were then fractionated and characterized by gas chromatography-mass spectrometry (GC-MS). Finally, the efficacy and the potential molecular mechanism of the co-treatment were portrayed via quantitative real-time polymerase chain reaction (qRT-PCR) analysis. RESULTS Initial screening resulted in the selection of the two Phoenix dactylifera L. cultivars Safawi and Sukkari methanolic extracts. Sukkari showed superior capacity to revert TAM phenotype into M1 as well as more prominent upregulation of M1 markers and repression of melanoma immunosuppressive markers relative to positive control (resiquimod). Molecularly, it was shown that PDT of melanoma cells in the presence of the secretome of repolarized TAMs surpassed the monotherapy via the modulation of the H19/iNOS/PD-L1immune-regulatory axis. CONCLUSION This study highlights the potential utilization of nutraceuticals in combination with PDT in the treatment of melanoma to provide a dual activity through alleviating the immune suppressive TME and potentiating the anti-tumor responses.
Collapse
Affiliation(s)
- Aya H Soliman
- Department of Pharmaceutical Biology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, Main Entrance El Tagamoa El Khames, New Cairo 11511, Egypt.
| | - Rana A Youness
- Department of Pharmaceutical Biology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, Main Entrance El Tagamoa El Khames, New Cairo 11511, Egypt; Department of Biology and Biochemistry, Faculty of Biotechnology, German International University, New Administrative Capital, New Cairo 11835, Egypt
| | - Aya A Sebak
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11511, Egypt.
| | - Heba Handoussa
- Department of Pharmaceutical Biology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, Main Entrance El Tagamoa El Khames, New Cairo 11511, Egypt
| |
Collapse
|
4
|
Isolation and characterization of glycoprotein (CNP) isolated from Cocos nucifera L. nutshell and its immunomodulatory role on macrophage activation. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
5
|
Bose A, Baral R. COVID-19 imparted immune manifestation can be combated by NLGP: Lessons from cancer research. Cytokine 2022; 158:155980. [PMID: 35921791 PMCID: PMC9339246 DOI: 10.1016/j.cyto.2022.155980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 01/08/2023]
Abstract
SARS-CoV-2 easily infects human monocytes, macrophages and possibly dendritic cells (DCs), causing dysfunctions of these important antigen presenting cells (APCs). Observed DC dysfunctions facilitate improper antigen presentation, which obviously results T cell anergy, exhaustion and apoptosis, thus, may be contributing significantly in SARS-CoV-2 infection associated lymphopenia. Neem Leaf Glycoprotein or NLGP has enormous role in altered DC functions, thereby, offering optimum T cell mediated cytotoxicity, as experienced from cancer system. Such NLGP guided correction of altered DCs might also be effective to generate proper SARS-CoV-2-specific effector and central memory T cells.
Collapse
Affiliation(s)
- Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India.
| |
Collapse
|
6
|
Singh A, Chatterjee A, Rakshit S, Shanmugam G, Mohanty LM, Sarkar K. Neem Leaf Glycoprotein in immunoregulation of cancer. Hum Immunol 2022; 83:768-777. [DOI: 10.1016/j.humimm.2022.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/04/2022]
|
7
|
Dasgupta S, Saha A, Ganguly N, Bhuniya A, Dhar S, Guha I, Ghosh T, Sarkar A, Ghosh S, Roy K, Das T, Banerjee S, Pal C, Baral R, Bose A. NLGP regulates RGS5-TGFβ axis to promote pericyte-dependent vascular normalization during restricted tumor growth. FASEB J 2022; 36:e22268. [PMID: 35363396 DOI: 10.1096/fj.202101093r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/05/2022] [Accepted: 03/09/2022] [Indexed: 12/25/2022]
Abstract
Altered RGS5-associated intracellular pericyte signaling and its abnormal crosstalk with endothelial cells (ECs) result chaotic tumor-vasculature, prevent effective drug delivery, promote immune-evasion and many more to ensure ultimate tumor progression. Moreover, the frequency of lethal-RGS5high pericytes within tumor was found to increase with disease progression, which signifies the presence of altered cell death pathway within tumor microenvironment (TME). In this study, we checked whether and how neem leaf glycoprotein (NLGP)-immunotherapy-mediated tumor growth restriction is associated with modification of pericytes' signaling, functions and its interaction with ECs. Analysis of pericytes isolated from tumors of NLGP treated mice suggested that NLGP treatment promotes apoptosis of NG2+ RGS5high -fuctionally altered pericytes by downregulating intra-tumoral TGFβ, along with maintenance of more matured RGS5neg pericytes. NLGP-mediated inhibition of TGFβ within TME rescues binding of RGS5 with Gαi and thereby termination of PI3K-AKT mediated survival signaling by downregulating Bcl2 and initiating pJNK mediated apoptosis. Limited availability of TGFβ also prevents complex-formation between RGS5 and Smad2 and rapid RGS5 nuclear translocation to mitigate alternate immunoregulatory functions of RGS5high tumor-pericytes. We also observed binding of Ang1 from pericytes with Tie2 on ECs in NLGP-treated tumor, which support re-association of pericytes with endothelium and subsequent vessel stabilization. Furthermore, NLGP-therapy- associated RGS5 deficiency relieved CD4+ and CD8+ T cells from anergy by regulating 'alternate-APC-like' immunomodulatory characters of tumor-pericytes. Taken together, present study described the mechanisms of NLGP's effectiveness in normalizing tumor-vasculature by chiefly modulating pericyte-biology and EC-pericyte interactions in tumor-host to further strengthen its translational potential as single modality treatment.
Collapse
Affiliation(s)
- Shayani Dasgupta
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Akata Saha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Nilanjan Ganguly
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Avishek Bhuniya
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Sukanya Dhar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Ipsita Guha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Tithi Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anirban Sarkar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Sarbari Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Kamalika Roy
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | - Tapasi Das
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Saptak Banerjee
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Chiranjib Pal
- Cellular Immunology and Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
8
|
Goswami KK, Bose A, Baral R. Macrophages in tumor: An inflammatory perspective. Clin Immunol 2021; 232:108875. [PMID: 34740843 DOI: 10.1016/j.clim.2021.108875] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/07/2021] [Accepted: 10/27/2021] [Indexed: 01/08/2023]
Abstract
Inflammation is a part of carefully co-ordinated healing immune exercise to eliminate injurious stimuli. However, in substantial number of cancer types, it contributes in shaping up of robust tumor microenvironment (TME). Solid TME promotes infiltration of tumor associated macrophages (TAMs) that contributes to cancer promotion. TAMs are functionally heterogeneous and display an extraordinary degree of plasticity, which allow 'Switching' of macrophages into an 'M2', phenotype, linked with immunosuppression, advancement of tumor angiogenesis with metastatic consequences. In contrary to the classical M1 macrophages, these M2 TAMs are high-IL-10, TGF-β secreting-'anti-inflammatory'. In this review, we will discuss the modes of infiltration and switching of TAMs into M2 anti-inflammatory state in the TME to promote immunosuppression and inflammation-driven cancer.
Collapse
Affiliation(s)
- Kuntal Kanti Goswami
- Department of Microbiology, Asutosh College, 92, S. P. Mukherjee Road, Kolkata 700026, India.
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| |
Collapse
|
9
|
Sarkar S, Singh RP, Bhattacharya G. Exploring the role of Azadirachta indica (neem) and its active compounds in the regulation of biological pathways: an update on molecular approach. 3 Biotech 2021; 11:178. [PMID: 33927969 PMCID: PMC7981372 DOI: 10.1007/s13205-021-02745-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 03/13/2021] [Indexed: 01/26/2023] Open
Abstract
In ethnomedicine, plant parts and compounds are used traditionally to treat different diseases. Neem (Azadirachta indica A. Juss) is the most versatile and useful medicinal plant ever found. Its every part is rich in bioactive compounds, which have traditionally been used to treat different ailments including infectious diseases. Bioactive compounds such as nimbolide, azarirachtin, and gedunin of neem are reported to have a tremendous ability to regulate numerous biological processes in vitro and in vivo. The present review article aims to explore the importance of neem extracts and bioactive compounds in the regulation of different biological pathways. We have reviewed research articles up to March 2020 on the role of neem in antioxidant, anti-inflammatory, antiangiogenic, immunomodulatory, and apoptotic activities. Studies on the concerned fields demonstrate that the bioactive compounds and extracts of neem have a regulatory effect on several biological mechanisms. It has been unveiled that extensive research is carried out on limonoids such as nimbolide and azarirachtin. It is evidenced by different studies that neem extracts are the potential to scavenge free radicals and reduce ROS-mediated damage to cells. Neem can be used to normalize lipid peroxidation and minimize ROS-mediated cell death. Besides, neem extracts can significantly reduce the release of proinflammatory cytokines and elevate the count of CD4 + and CD8 + T-cells. This review indicates the pivotal roles of A. indica in the regulation of different biological pathways. However, future investigations on other bioactive compounds of neem may reveal different therapeutic potentials.
Collapse
Affiliation(s)
- Subendu Sarkar
- Department of Surgery, University School of Medicine, Indiana University, Indianapolis, IN 46202 USA
| | - Rajender Pal Singh
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012 India
| | - Gorachand Bhattacharya
- Jagannath Gupta Institute of Medical Sciences & Hospital, KP Mondal Road, Buita, Nishchintapur, Budge Budge, Kolkata 700137 India
| |
Collapse
|
10
|
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
|
11
|
Saha A, Nandi P, Dasgupta S, Bhuniya A, Ganguly N, Ghosh T, Guha I, Banerjee S, Baral R, Bose A. Neem Leaf Glycoprotein Restrains VEGF Production by Direct Modulation of HIF1α-Linked Upstream and Downstream Cascades. Front Oncol 2020; 10:260. [PMID: 32211322 PMCID: PMC7067891 DOI: 10.3389/fonc.2020.00260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/14/2020] [Indexed: 12/11/2022] Open
Abstract
Neem Leaf Glycoprotein (NLGP) is a natural immunomodulator, have shown sustained tumor growth restriction as well as angiogenic normalization chiefly by activating CD8+ T cells. Here, we have investigated the direct role of NLGP as a regulator of tumor microenvironmental hypoxia and associated vascular endothelial growth factor (VEGF) production. We observed a significant reduction in VEGF level in both in vivo murine tumor and in vitro cancer cells (B16Mel, LLC) and macrophages after NLGP treatment. Interestingly, NLGP mediated VEGF downregulation in tumor cells or macrophages within hypoxic chamber was found at an early 4 h and again at late 24 h in mRNA level. Our data suggested that NLGP prevented hypoxia-induced strong binding of HIF1α with its co-factors, CBP/p300 and Sp3, but not with Sp1, which eventually limit the binding of HIF1α-transcriptional complex to hypoxia responsive element of VEGF promoter and results in restricted early VEGF transcription. On the otherhand, suppressed phosphorylation of Stat3 by NLGP results reduction of HIF1α at 24 h of hypoxia that further support sustained VEGF down-regulation. However, NLGP fails to regulate VHL activity as observed by both in vivo and in vitro studies. Therefore, this study for the first time reveals a mechanistic insight of NLGP mediated inhibition of angiogenesis by suppressing VEGF, which might help in vascular normalization to influence better drug delivery.
Collapse
Affiliation(s)
- Akata Saha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| | - Partha Nandi
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| | - Shayani Dasgupta
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| | - Avishek Bhuniya
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| | - Nilanjan Ganguly
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| | - Tithi Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| | - Ipsita Guha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| | - Saptak Banerjee
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), Kolkata, India
| |
Collapse
|
12
|
Huang SP, Guan X, Kai GY, Xu YZ, Xu Y, Wang HJ, Pang T, Zhang LY, Liu Y. Broussonin E suppresses LPS-induced inflammatory response in macrophages via inhibiting MAPK pathway and enhancing JAK2-STAT3 pathway. Chin J Nat Med 2020; 17:372-380. [PMID: 31171272 DOI: 10.1016/s1875-5364(19)30043-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Indexed: 02/07/2023]
Abstract
Macrophages play an important role in inflammation, and excessive and chronic activation of macrophages leads to systemic inflammatory diseases, such as atherosclerosis and rheumatoid arthritis. In this paper, we explored the anti-inflammatory effect of broussonin E, a novel phenolic compound isolated from the barks ofBroussonetia kanzinoki, and its underlying molecular mechanisms. We discovered that Broussonin E could suppress the LPS-induced pro-inflammatory production in RAW264.7 cells, involving TNF-α, IL-1β, IL-6, COX-2 and iNOS. And broussonin E enhanced the expressions of anti-inflammatory mediators such as IL-10, CD206 and arginase-1 (Arg-1) in LPS-stimulated RAW264.7 cells. Further, we demonstrated that broussonin E inhibited the LPS-stimulated phosphorylation of ERK and p38 MAPK. Moreover, we found that broussonin E could activate janus kinase (JAK) 2, signal transducer and activator of transcription (STAT) 3. Downregulated pro-inflammatory cytokines and upregulated anti-inflammatory factors by broussonin E were abolished by using the inhibitor of JAK2-STAT3 pathway, WP1066. Taken together, our results showed that broussonin E could suppress inflammation by modulating macrophages activation statevia inhibiting the ERK and p38 MAPK and enhancing JAK2-STAT3 signaling pathway, and can be further developed as a promising drug for the treatment of inflammation-related diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Shao-Peng Huang
- School of Basic Medicine, Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006,China
| | - Xin Guan
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 311402, China; Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Guo-Yin Kai
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Ya-Zhou Xu
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan Xu
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hao-Jie Wang
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Pang
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Lu-Yong Zhang
- School of Basic Medicine, Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006,China; Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Ying Liu
- School of Basic Medicine, Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006,China.
| |
Collapse
|
13
|
Ghosh T, Nandi P, Ganguly N, Guha I, Bhuniya A, Ghosh S, Sarkar A, Saha A, Dasgupta S, Baral R, Bose A. NLGP counterbalances the immunosuppressive effect of tumor-associated mesenchymal stem cells to restore effector T cell functions. Stem Cell Res Ther 2019; 10:296. [PMID: 31547863 PMCID: PMC6757425 DOI: 10.1186/s13287-019-1349-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 12/13/2022] Open
Abstract
Background A dynamic interaction between tumor cells and its surrounding stroma promotes the initiation, progression, metastasis, and chemoresistance of solid tumors. Emerging evidences suggest that targeting the stromal events could improve the efficacies of current therapeutics. Within tumor microenvironment (TME), stromal progenitor cells, i.e., MSCs, interact and eventually modulate the biology and functions of cancer and immune cells. Our recent finding disclosed a novel mechanism stating that tumor-associated MSCs inhibit the T cell proliferation and effector functions by blocking cysteine transport to T cells by dendritic cells (DCs), which makes MSCs as a compelling candidate as a therapeutic target. Immunomodulation by nontoxic neem leaf glycoprotein (NLGP) on dysfunctional cancer immunity offers significant therapeutic benefits to murine tumor host; however, its modulation on MSCs and its impact on T cell functions need to be elucidated. Methods Bone marrow-derived primary MSCs or murine 10 T1/2 MSCs were tumor-conditioned (TC-MSCs) and co-cultured with B16 melanoma antigen-specific DCs and MACS purified CD4+ and CD8+ T cells. T cell proliferation of T cells was checked by Ki67-based flow-cytometric and thymidine-incorporation assays. Cytokine secretion was measured by ELISA. The expression of cystathionase in DCs was assessed by RT-PCR. The STAT3/pSTAT3 levels in DCs were assessed by western blot, and STAT3 function was confirmed using specific SiRNA. Solid B16 melanoma tumor growth was monitored following adoptive transfer of conditioned CD8+ T cells. Results NLGP possesses an ability to restore anti-tumor T cell functions by modulating TC-MSCs. Supplementation of NLGP in DC-T cell co-culture significantly restored the inhibition in T cell proliferation and IFNγ secretion almost towards normal in the presence of TC-MSCs. Adoptive transfer of NLGP-treated TC-MSC supernatant educated CD8+ T cells in solid B16 melanoma bearing mice resulted in better tumor growth restriction than TC-MSC conditioned CD8+ T cells. NLGP downregulates IL-10 secretion by TC-MSCs, and concomitantly, pSTAT3 expression was downregulated in DCs in the presence of NLGP-treated TC-MSC supernatant. As pSTAT3 negatively regulates cystathionase expression in DCs, NLGP indirectly helps to maintain an almost normal level of cystathionase gene expression in DCs making them able to export sufficient amount of cysteine required for optimum T cell proliferation and effector functions within TME. Conclusions NLGP could be a prospective immunotherapeutic agent to control the functions and behavior of highly immunosuppressive TC-MSCs providing optimum CD8+ T cell functions to showcase an important new approach that might be effective in overall cancer treatment. Electronic supplementary material The online version of this article (10.1186/s13287-019-1349-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tithi Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Partha Nandi
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Nilanjan Ganguly
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Ipsita Guha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Avishek Bhuniya
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Sarbari Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Anirban Sarkar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Akata Saha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Shayani Dasgupta
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute (CNCI), 37, S. P. Mukherjee Road, Kolkata, 700026, India.
| |
Collapse
|
14
|
Wang FJ, Wang P, Chen LY, Geng YW, Chen H, Meng ZQ, Liu LM, Chen Z. TAM Infiltration Differences in "Tumor-First" and " ZHENG-First" Models and the Underlying Inflammatory Molecular Mechanism in Pancreatic Cancer. Integr Cancer Ther 2018; 17:707-716. [PMID: 29681184 PMCID: PMC6142096 DOI: 10.1177/1534735418771193] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Syndrome (ZHENG in Chinese) in
traditional Chinese medicine (TCM) refers to the intrinsic characteristics of a
pathological process at a certain stage; these characteristics are influenced by
internal and external environments and reveal the nature of a disease. Proper
syndrome differentiation is the basic principle that guides clinical treatment.
Objective: To have a good understanding of tumor progression
and the different mechanisms related to ZHENG that have
occurred before and after tumor development and to explore the valid evaluation
criteria of different pancreatic cancer syndromes to improve the guiding role of
TCM syndrome differentiation in pancreatic cancer treatment.
Methods: In this study, we established mouse subcutaneous
pancreatic cancer models, namely, Con (control), Pi-Xu (Spleen-Deficiency),
Shi-Re (Dampness-Heat), and Xue-Yu (Blood-Stasis). Then, for the first time, we
compared the different effects of “ZHENG-first” (referring to a
different disease status that occurred before tumor occurrence) and
“Tumor-first” (referring to the change in the tumor microenvironment and the
resulting changes in the state of the body) conditions on tumor progression and
evaluated the associated molecular mechanisms. Results: We found
that tumor growth in the “ZHENG-first” and “Tumor-first”
conditions was different. In the “Tumor-first” model, the tumor growth in the
Pi-Xu group was faster than that in the other groups. However, in the
“ZHENG-first” model, the tumor growth trend was most
obvious in the Shi-Re group. There was a difference in tumor-associated
macrophage infiltration between the 2 models. The expression levels of the
inflammatory cytokines IL-6, IL-10, and P-STAT3 were also
differentially altered. Conclusion: The emergence of
ZHENG conditions before or after tumor occurrence had
different impacts on pancreatic cancer development, and these impacts may be
related to differences in tumor-associated macrophage infiltration and the
involved inflammatory cytokines IL-6, IL-10, and P-STAT3. The
study results uncovered the molecular basis of syndrome differentiation in
pancreatic cancer progression, which might provide more specific guidance for
TCM treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Feng-Jiao Wang
- 1 Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China.,2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Wang
- 1 Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China.,2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lian-Yu Chen
- 1 Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China.,2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Wen Geng
- 1 Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China.,2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Chen
- 1 Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China.,2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Qiang Meng
- 1 Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China.,2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lu-Ming Liu
- 1 Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China.,2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Chen
- 1 Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China.,2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
15
|
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: 184] [Impact Index Per Article: 26.3] [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
|
16
|
(+)-Borneol improves the efficacy of edaravone against DSS-induced colitis by promoting M2 macrophages polarization via JAK2-STAT3 signaling pathway. Int Immunopharmacol 2017; 53:1-10. [DOI: 10.1016/j.intimp.2017.10.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/27/2017] [Accepted: 10/01/2017] [Indexed: 12/15/2022]
|
17
|
Goswami KK, Ghosh T, Ghosh S, Sarkar M, Bose A, Baral R. Tumor promoting role of anti-tumor macrophages in tumor microenvironment. Cell Immunol 2017; 316:1-10. [PMID: 28433198 DOI: 10.1016/j.cellimm.2017.04.005] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/09/2017] [Accepted: 04/12/2017] [Indexed: 01/02/2023]
Abstract
Recent advances in tumor biology demand detailed analysis of the complex interaction of tumor cells with their adjacent microenvironment (tumor stroma) to understand the various mechanisms involved in tumor growth and metastasis. Mononuclear phagocytes or macrophages, a type of innate immune cells, defend the organism against infection and injury. On the otherhand, tumor associated macrophages (TAMs) constitute a significant part of the tumor-infiltrating immune cells, have been linked to the growth, angiogenesis, and metastasis of a variety of cancers, most likely through polarization of TAMs to the M2 (alternative) phenotype. Clinical and experimental evidences have shown that cancer tissues with high infiltration of TAMs are associated with poor patient prognosis and resistance to therapies, thus, targeting of TAMs in tumors is considered as a promising immunotherapeutic strategy. Depletion of M2 TAMs or 're-education' of them as anti-tumor effectors might contribute significantly to the search of new modalities in anti-cancer treatments. Basic questions on the factors responsible for homing of macrophages in tumors, mechanism of conversion of M1 to M2 TAMs, their functionality and, finally, the possible ways to target M2 TAMs are discussed.
Collapse
Affiliation(s)
- Kuntal Kanti Goswami
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Tithi Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Sarbari Ghosh
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Madhurima Sarkar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Anamika Bose
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata 700026, India.
| |
Collapse
|
18
|
Chaudhary S, Kanwar RK, Sehgal A, Cahill DM, Barrow CJ, Sehgal R, Kanwar JR. Progress on Azadirachta indica Based Biopesticides in Replacing Synthetic Toxic Pesticides. FRONTIERS IN PLANT SCIENCE 2017; 8:610. [PMID: 28533783 PMCID: PMC5420583 DOI: 10.3389/fpls.2017.00610] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 04/04/2017] [Indexed: 05/13/2023]
Abstract
Over the years, extensive use of commercially available synthetic pesticides against phytophagous insects has led to their bioaccumulation in the environment causing increased resistance and reduction in soil biodiversity. Further, 90% of the applied pesticides enter the various environmental resources as a result of run-off, exposing the farmers as well as consumers of the agricultural produce to severe health issues. Therefore, growing attention has been given toward the development of alternate environmentally friendly pesticides/insecticides that would aid an efficient pest management system and also prevent chronic exposures leading to diseases. One such strategy is, the use of neem plant's (Binomial name: Azadirachta indica) active ingredients which exhibit agro-medicinal properties conferring insecticidal as well as immunomodulatory and anti-cancer properties. The most prominent constituent of neem is azadirachtin, which has been established as a pivotal insecticidal ingredient. It acts as an antifeedant, repellent, and repugnant agent and induces sterility in insects by preventing oviposition and interrupting sperm production in males. This review discusses, key neem pesticidal components, their active functional ingredients along with recent strategies on employing nanocarriers, to provide controlled release of the active ingredients and to improve their stability and sustainability.
Collapse
Affiliation(s)
- Suman Chaudhary
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research, Faculty of Health, Centre for Molecular and Medical Research, Strategic Research Centre, School of Medicine, Deakin UniversityGeelong, VIC, Australia
| | - Rupinder K. Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research, Faculty of Health, Centre for Molecular and Medical Research, Strategic Research Centre, School of Medicine, Deakin UniversityGeelong, VIC, Australia
| | - Alka Sehgal
- Department of Gynecology, Government Medical College and HospitalChandigarh, India
| | - David M. Cahill
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences, Deakin UniversityGeelong, VIC, Australia
| | - Colin J. Barrow
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences, Deakin UniversityGeelong, VIC, Australia
| | - Rakesh Sehgal
- Department of Medical Parasitology, Jawaharlal Institute of Postgraduate Medical Education and ResearchChandigarh, India
| | - Jagat R. Kanwar
- Nanomedicine-Laboratory of Immunology and Molecular Biomedical Research, Faculty of Health, Centre for Molecular and Medical Research, Strategic Research Centre, School of Medicine, Deakin UniversityGeelong, VIC, Australia
- *Correspondence: Jagat R. Kanwar
| |
Collapse
|