51
|
Noncoding RNAs: Regulating the crosstalk between tumor-associated macrophages and gastrointestinal cancer. Biomed Pharmacother 2022; 153:113370. [DOI: 10.1016/j.biopha.2022.113370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 01/19/2023] Open
|
52
|
Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov 2022; 21:799-820. [PMID: 35974096 PMCID: PMC9380983 DOI: 10.1038/s41573-022-00520-5] [Citation(s) in RCA: 659] [Impact Index Per Article: 329.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2022] [Indexed: 12/11/2022]
Abstract
Tumour-associated macrophages are an essential component of the tumour microenvironment and have a role in the orchestration of angiogenesis, extracellular matrix remodelling, cancer cell proliferation, metastasis and immunosuppression, as well as in resistance to chemotherapeutic agents and checkpoint blockade immunotherapy. Conversely, when appropriately activated, macrophages can mediate phagocytosis of cancer cells and cytotoxic tumour killing, and engage in effective bidirectional interactions with components of the innate and adaptive immune system. Therefore, they have emerged as therapeutic targets in cancer therapy. Macrophage-targeting strategies include inhibitors of cytokines and chemokines involved in the recruitment and polarization of tumour-promoting myeloid cells as well as activators of their antitumorigenic and immunostimulating functions. Early clinical trials suggest that targeting negative regulators (checkpoints) of myeloid cell function indeed has antitumor potential. Finally, given the continuous recruitment of myelomonocytic cells into tumour tissues, macrophages are candidates for cell therapy with the development of chimeric antigen receptor effector cells. Macrophage-centred therapeutic strategies have the potential to complement, and synergize with, currently available tools in the oncology armamentarium. Macrophages can promote tumorigenesis and enhance the antitumour response. This Review discusses the molecular mechanisms underlying the reprogramming of macrophages in the tumour microenvironment and provides an overview of macrophage-targeted therapies for the treatment of cancer.
Collapse
Affiliation(s)
- Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy. .,IRCCS- Humanitas Research Hospital, Milan, Italy. .,The William Harvey Research Institute, Queen Mary University of London, London, UK.
| | - Paola Allavena
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS- Humanitas Research Hospital, Milan, Italy
| | - Federica Marchesi
- IRCCS- Humanitas Research Hospital, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS- Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
53
|
Jafarzadeh A, Nemati M, Aminizadeh N, Bodhale N, Sarkar A, Jafarzadeh S, Sharifi I, Saha B. Bidirectional cytokine-microRNA control: A novel immunoregulatory framework in leishmaniasis. PLoS Pathog 2022; 18:e1010696. [PMID: 35925884 PMCID: PMC9351994 DOI: 10.1371/journal.ppat.1010696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
As effector innate immune cells and as a host to the protozoan parasite Leishmania, macrophages play a dual role in antileishmanial immunoregulation. The 2 key players in this immunoregulation are the macrophage-expressed microRNAs (miRNAs) and the macrophage-secreted cytokines. miRNAs, as small noncoding RNAs, play vital roles in macrophage functions including cytokines and chemokines production. In the reverse direction, Leishmania-regulated cytokines alter miRNAs expression to regulate the antileishmanial functions of macrophages. The miRNA patterns vary with the time and stage of infection. The cytokine-regulated macrophage miRNAs not only help parasite elimination or persistence but also regulate cytokine production from macrophages. Based on these observations, we propose a novel immunoregulatory framework as a scientific rationale for antileishmanial therapy.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- * E-mail: (AJ); (BS)
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Najmeh Aminizadeh
- Department of Histology, School of Medicine, Islamic Azad University Branch of Kerman, Kerman
| | | | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, Odisha, India
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Iran
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Bhaskar Saha
- National Centre For Cell Science, Pune, India
- Trident Academy of Creative Technology, Bhubaneswar, Odisha, India
- * E-mail: (AJ); (BS)
| |
Collapse
|
54
|
Anti-Inflammatory microRNAs for Treating Inflammatory Skin Diseases. Biomolecules 2022; 12:biom12081072. [PMID: 36008966 PMCID: PMC9405611 DOI: 10.3390/biom12081072] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 02/07/2023] Open
Abstract
Skin inflammation occurs due to immune dysregulation because of internal disorders, infections, and allergic reactions. The inflammation of the skin is a major sign of chronic autoimmune inflammatory diseases, such as psoriasis, atopic dermatitis (AD), and lupus erythematosus. Although there are many therapies for treating these cutaneous inflammation diseases, their recurrence rates are high due to incomplete resolution. MicroRNA (miRNA) plays a critical role in skin inflammation by regulating the expression of protein-coding genes at the posttranscriptional level during pathogenesis and homeostasis maintenance. Some miRNAs possess anti-inflammatory features, which are beneficial for mitigating the inflammatory response. miRNAs that are reduced in inflammatory skin diseases can be supplied transiently using miRNA mimics and agomir. miRNA-based therapies that can target multiple genes in a given pathway are potential candidates for the treatment of skin inflammation. This review article offers an overview of the function of miRNA in skin inflammation regulation, with a focus on psoriasis, AD, and cutaneous wounds. Some bioactive molecules can target and modulate miRNAs to achieve the objective of inflammation suppression. This review also reports the anti-inflammatory efficacy of these molecules through modulating miRNA expression. The main limitations of miRNA-based therapies are rapid biodegradation and poor skin and cell penetration. Consideration was given to improving these drawbacks using the approaches of cell-penetrating peptides (CPPs), nanocarriers, exosomes, and low-frequency ultrasound. A formulation design for successful miRNA delivery into skin and target cells is also described in this review. The possible use of miRNAs as biomarkers and therapeutic modalities could open a novel opportunity for the diagnosis and treatment of inflammation-associated skin diseases.
Collapse
|
55
|
Kim H, Park HJ, Chang HW, Back JH, Lee SJ, Park YE, Kim EH, Hong Y, Kwak G, Kwon IC, Lee JE, Lee YS, Kim SY, Yang Y, Kim SH. Exosome-guided direct reprogramming of tumor-associated macrophages from protumorigenic to antitumorigenic to fight cancer. Bioact Mater 2022; 25:527-540. [PMID: 37056267 PMCID: PMC10087080 DOI: 10.1016/j.bioactmat.2022.07.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/29/2022] [Accepted: 07/19/2022] [Indexed: 11/02/2022] Open
Abstract
Highly immunosuppressive tumor microenvironment containing various protumoral immune cells accelerates malignant transformation and treatment resistance. In particular, tumor-associated macrophages (TAMs), as the predominant infiltrated immune cells in a tumor, play a pivotal role in regulating the immunosuppressive tumor microenvironment. As a potential therapeutic strategy to counteract TAMs, here we explore an exosome-guided in situ direct reprogramming of tumor-supportive M2-polarized TAMs into tumor-attacking M1-type macrophages. Exosomes derived from M1-type macrophages (M1-Exo) promote a phenotypic switch from anti-inflammatory M2-like TAMs toward pro-inflammatory M1-type macrophages with high conversion efficiency. Reprogrammed M1 macrophages possessing protein-expression profiles similar to those of classically activated M1 macrophages display significantly increased phagocytic function and robust cross-presentation ability, potentiating antitumor immunity surrounding the tumor. Strikingly, these M1-Exo also lead to the conversion of human patient-derived TAMs into M1-like macrophages that highly express MHC class II, offering the clinical potential of autologous and allogeneic exosome-guided direct TAM reprogramming for arming macrophages to join the fight against cancer.
Collapse
|
56
|
Zatterale F, Raciti GA, Prevenzano I, Leone A, Campitelli M, De Rosa V, Beguinot F, Parrillo L. Epigenetic Reprogramming of the Inflammatory Response in Obesity and Type 2 Diabetes. Biomolecules 2022; 12:biom12070982. [PMID: 35883538 PMCID: PMC9313117 DOI: 10.3390/biom12070982] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
For the past several decades, the prevalence of obesity and type 2 diabetes (T2D) has continued to rise on a global level. The risk contributing to this pandemic implicates both genetic and environmental factors, which are functionally integrated by epigenetic mechanisms. While these conditions are accompanied by major abnormalities in fuel metabolism, evidence indicates that altered immune cell functions also play an important role in shaping of obesity and T2D phenotypes. Interestingly, these events have been shown to be determined by epigenetic mechanisms. Consistently, recent epigenome-wide association studies have demonstrated that immune cells from obese and T2D individuals feature specific epigenetic profiles when compared to those from healthy subjects. In this work, we have reviewed recent literature reporting epigenetic changes affecting the immune cell phenotype and function in obesity and T2D. We will further discuss therapeutic strategies targeting epigenetic marks for treating obesity and T2D-associated inflammation.
Collapse
Affiliation(s)
- Federica Zatterale
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Gregory Alexander Raciti
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Immacolata Prevenzano
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Alessia Leone
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Michele Campitelli
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Veronica De Rosa
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
| | - Francesco Beguinot
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
- Correspondence: (F.B.); (L.P.); Tel.: +39-081-746-3248 (F.B.); +39-081-746-3045 (L.P.)
| | - Luca Parrillo
- Department of Translational Medical Science, Federico II University of Naples, 80131 Naples, Italy; (F.Z.); (G.A.R.); (I.P.); (A.L.); (M.C.)
- URT Genomic of Diabetes, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy;
- Correspondence: (F.B.); (L.P.); Tel.: +39-081-746-3248 (F.B.); +39-081-746-3045 (L.P.)
| |
Collapse
|
57
|
Decoding microRNA drivers in Atherosclerosis. Biosci Rep 2022; 42:231479. [PMID: 35758143 PMCID: PMC9289798 DOI: 10.1042/bsr20212355] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/17/2022] [Accepted: 06/26/2022] [Indexed: 11/17/2022] Open
Abstract
An estimated 97% of the human genome consists of non-protein-coding sequences. As our understanding of genome regulation improves, this has led to the characterization of a diverse array of non-coding RNAs (ncRNA). Among these, micro-RNAs (miRNAs) belong to the short ncRNA class (22–25 nucleotides in length), with approximately 2500 miRNA genes encoded within the human genome. From a therapeutic perspective, there is interest in exploiting miRNA as biomarkers of disease progression and response to treatments, as well as miRNA mimics/repressors as novel medicines. miRNA have emerged as an important class of RNA master regulators with important roles identified in the pathogenesis of atherosclerotic cardiovascular disease. Atherosclerosis is characterized by a chronic inflammatory build-up, driven largely by low-density lipoprotein cholesterol accumulation within the artery wall and vascular injury, including endothelial dysfunction, leukocyte recruitment and vascular remodelling. Conventional therapy focuses on lifestyle interventions, blood pressure-lowering medications, high-intensity statin therapy and antiplatelet agents. However, a significant proportion of patients remain at increased risk of cardiovascular disease. This continued cardiovascular risk is referred to as residual risk. Hence, a new drug class targeting atherosclerosis could synergise with existing therapies to optimise outcomes. Here, we review our current understanding of the role of ncRNA, with a focus on miRNA, in the development and progression of atherosclerosis, highlighting novel biological mechanisms and therapeutic avenues.
Collapse
|
58
|
Wicks EE, Ran KR, Kim JE, Xu R, Lee RP, Jackson CM. The Translational Potential of Microglia and Monocyte-Derived Macrophages in Ischemic Stroke. Front Immunol 2022; 13:897022. [PMID: 35795678 PMCID: PMC9251541 DOI: 10.3389/fimmu.2022.897022] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The immune response to ischemic stroke is an area of study that is at the forefront of stroke research and presents promising new avenues for treatment development. Upon cerebral vessel occlusion, the innate immune system is activated by danger-associated molecular signals from stressed and dying neurons. Microglia, an immune cell population within the central nervous system which phagocytose cell debris and modulate the immune response via cytokine signaling, are the first cell population to become activated. Soon after, monocytes arrive from the peripheral immune system, differentiate into macrophages, and further aid in the immune response. Upon activation, both microglia and monocyte-derived macrophages are capable of polarizing into phenotypes which can either promote or attenuate the inflammatory response. Phenotypes which promote the inflammatory response are hypothesized to increase neuronal damage and impair recovery of neuronal function during the later phases of ischemic stroke. Therefore, modulating neuroimmune cells to adopt an anti-inflammatory response post ischemic stroke is an area of current research interest and potential treatment development. In this review, we outline the biology of microglia and monocyte-derived macrophages, further explain their roles in the acute, subacute, and chronic stages of ischemic stroke, and highlight current treatment development efforts which target these cells in the context of ischemic stroke.
Collapse
|
59
|
Role of magnesium-doped calcium sulfate and β-tricalcium phosphate composite ceramics in macrophage polarization and osteo-induction. Odontology 2022; 110:735-746. [PMID: 35653001 PMCID: PMC9463206 DOI: 10.1007/s10266-022-00708-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 04/12/2022] [Indexed: 11/30/2022]
Abstract
In the current study, we explored the role of Mg2+-doped CaSO4/β-TCP composite biopolymer in regulating macrophage polarization and its relation with enhanced osteogenic differentiation of periodontal ligament stem cells. Furthermore, mechanism underling the regulation of macrophage polarization by CaSO4/β-TCP was evaluated. Mg2+-doped CaSO4/β-TCP composite was characterized by scanning electron microscopy (SEM) and X-ray diffraction (XRD). Macrophage polarization was characterized using flow cytometry analysis. Macrophage morphometric analysis was conducted by FITC phalloidin staining. Western blot and qRT-PCR assays were used to assess gene expression levels and miRNAs, respectively. SEM morphology of CaSO4/β-TCP ceramic revealed a particle size of 10–50 μm, and XRD spectrum showed that characteristic peak of samples was consistent with that of CaSO4 and β-TCP. Results from flow cytometry evidenced significant upregulation of M2 macrophage markers after adding ceramic biopolymer, indicating the induction of inactivated M0 macrophage polarization to M2 macrophage. Macrophage morphometric analysis revealed development of lamellar pseudopodia on day 7 in CaSO4/β-TCP group. Furthermore, flow cytometry revealed high positivity rate of 90.34% (CD44) and 89.36% (CD146). qRT-PCR results showed that the level of miR-21-5p was significantly decreased in M2 macrophages. Moreover, western blot analysis revealed upregulated expression levels of RUNX2, osterix (Osx), and osteopontin (OPN), and ELISA exhibited increase in cytokine levels (IL-1β, IL-10, TGF-β1, and BMP-2) in the presence of macrophages, indicating the osteogenic differentiation ability of periodontal ligament stem cells. The study evidenced the regulation of macrophage polarization by Mg2+-doped CaSO4/β-TCP composite ceramic and its mediation through lncRNA PVT1/miR-21-5p/smad2 molecular axis.
Collapse
|
60
|
Wang LL, Li ZH, Wang H, Kwak-Kim J, Liao AH. Cutting edge: the regulatory mechanisms of macrophage polarization and function during pregnancy. J Reprod Immunol 2022; 151:103627. [DOI: 10.1016/j.jri.2022.103627] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023]
|
61
|
Challagundla N, Saha B, Agrawal-Rajput R. Insights into inflammasome regulation: cellular, molecular, and pathogenic control of inflammasome activation. Immunol Res 2022; 70:578-606. [PMID: 35610534 DOI: 10.1007/s12026-022-09286-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Maintenance of immune homeostasis is an intricate process wherein inflammasomes play a pivotal role by contributing to innate and adaptive immune responses. Inflammasomes are ensembles of adaptor proteins that can trigger a signal following innate sensing of pathogens or non-pathogens eventuating in the inductions of IL-1β and IL-18. These inflammatory cytokines substantially influence the antigen-presenting cell's costimulatory functions and T helper cell differentiation, contributing to adaptive immunity. As acute and chronic disease conditions may accompany parallel tissue damage, we analyze the critical role of extracellular factors such as cytokines, amyloids, cholesterol crystals, etc., intracellular metabolites, and signaling molecules regulating inflammasome activation/inhibition. We develop an operative framework for inflammasome function and regulation by host cell factors and pathogens. While inflammasomes influence the innate and adaptive immune components' interplay modulating the anti-pathogen adaptive immune response, pathogens may target inflammasome inhibition as a survival strategy. As trapped between health and diseases, inflammasomes serve as promising therapeutic targets and their modus operandi serves as a scientific rationale for devising better therapeutic strategies.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India
| | - Bhaskar Saha
- National Centre for Cell Science, Lab-5, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Reena Agrawal-Rajput
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India.
| |
Collapse
|
62
|
Expression Profile of mRNAs and miRNAs Related to the Oxidative-Stress Phenomenon in the Ishikawa Cell Line Treated Either Cisplatin or Salinomycin. Biomedicines 2022; 10:biomedicines10051190. [PMID: 35625926 PMCID: PMC9138494 DOI: 10.3390/biomedicines10051190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/14/2022] [Accepted: 05/18/2022] [Indexed: 12/10/2022] Open
Abstract
The oxidative stress phenomenon is a result of anticancer therapy. The aim of this study was the assessment of gene expression profile changes, and to determine the miRNAs regulating genes’ transcriptional activity in an Ishikawa endometrial cancer culture exposed to cisplatin or salinomycin, compared to a control culture. The molecular analysis comprised the microarray technique (mRNAs and micro RNA (miRNA), the real-time quantitative reverse transcription reaction (RTqPCR), enzyme-linked immunosorbent assay (ELISA) reactions, and Western blot. NR4A2, MAP3K8, ICAM1, IL21, CXCL8, CCL7, and SLC7A11 were statistically significantly differentiated depending not only on time, but also on the drug used in the experiment. The conducted assessment indicated that the strongest links were between NR4A2 and hsa-miR-30a-5p and has-miR-302e, MAP3K8 and hsa-miR-144-3p, CXCL8 and hsa-miR-140-3p, and SLC7A11 and hsa-miR-144-3p. The obtained results suggest that four mRNAs—NR4A2, MAP3K8, CXCL8 and SLC7A11—and four miRNAs—hsa-miR-30a-5p, hsa-miR-302e, hsa-miR-144-3p and hsa-miR-140-3—changed their expressions regardless of the chemotherapeutic agent used, which suggests the possibility of their use in monitoring the severity of oxidative stress in endometrial cancer. However, considering the results at both the mRNA and the protein level, it is most likely that the expressions of NR4A2, MAP3K8, CXCL8 and SLC7A11 are regulated by miRNA molecules as well as other epigenetic mechanisms.
Collapse
|
63
|
Aljedaie MM. Epigenetic paradigms/exemplars of the macrophage: inflammasome axis in Leishmaniasis. Mol Cell Biochem 2022; 477:2553-2565. [PMID: 35595955 DOI: 10.1007/s11010-022-04460-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/28/2022] [Indexed: 11/26/2022]
Abstract
The infectious paradigms have recently led to the recognition interplay of complex phenomenon underpinning disease diagnosis and prognosis. Evidently, parasitic infection studies are depicting converging trends of the epigenetic, environmental, and microbiome contributions, assisting pathogen-directed modulations of host biological system. The molecular details of epigenetic variations and memory, along with the multi-omics data at the interface of the host-pathogen level becomes strong indicator of immune cell plasticity, differentiation, and pathogen survival. Despite being one of the most important aspects of the disease's etiopathology, the epigenetic regulation of host-pathogen interactions and evolutionary epigenetics have received little attention thus far. Recent evidence has focused on the growing need to link epigenetic and microbiome modulations on parasite phenotypic plasticity and pathogen-induced host phenotypic plasticity for designing futuristic therapeutic regimes. Leishmaniasis is a neglected tropical illness with varying degrees of disease severity that is linked to a trans-species and epigenetic heredity process, including the pathogen-induced host and strain-specific modulations. The review configures research findings aligning to the epigenetic epidemiology niche, involving co-evolutionary epigenetic inheritance and plasticity disease models. The epigenetic exemplars focus on the host-pathogen interactome expanse at the macrophage-inflammasome axis.
Collapse
Affiliation(s)
- Manei M Aljedaie
- Department of Biology, College of Sciences and Humanities, Prince Sattam Bin Abdulaziz University, PO Box 173, Al-Kharj, 11942, Saudi Arabia.
| |
Collapse
|
64
|
Matsuzaka Y, Yashiro R. Immune Modulation Using Extracellular Vesicles Encapsulated with MicroRNAs as Novel Drug Delivery Systems. Int J Mol Sci 2022; 23:ijms23105658. [PMID: 35628473 PMCID: PMC9146104 DOI: 10.3390/ijms23105658] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 12/13/2022] Open
Abstract
Self-tolerance involves protection from self-reactive B and T cells via negative selection during differentiation, programmed cell death, and inhibition of regulatory T cells. The breakdown of immune tolerance triggers various autoimmune diseases, owing to a lack of distinction between self-antigens and non-self-antigens. Exosomes are non-particles that are approximately 50–130 nm in diameter. Extracellular vesicles can be used for in vivo cell-free transmission to enable intracellular delivery of proteins and nucleic acids, including microRNAs (miRNAs). miRNAs encapsulated in exosomes can regulate the molecular pathways involved in the immune response through post-transcriptional regulation. Herein, we sought to summarize and review the molecular mechanisms whereby exosomal miRNAs modulate the expression of genes involved in the immune response.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, University of Tokyo, Minato-ku 108-8639, Tokyo, Japan
- Correspondence: ; Tel.: +81-3-5449-5372
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan; or
| |
Collapse
|
65
|
Abstract
MicroRNAs (miRNAs) are key players in gene regulation that target specific mRNAs for degradation or translational repression. Each miRNA is synthesized as a miRNA duplex comprising two strands (5p and 3p). However, only one of the two strands becomes active and is selectively incorporated into the RNA-induced silencing complex in a process known as miRNA strand selection. Recently, significant progress has been made in understanding the factors and processes involved in strand selection. Here, we explore the selection and functionality of the miRNA star strand (either 5p or 3p), which is generally present in the cell at low levels compared to its partner strand and, historically, has been thought to possess no biological activity. We also highlight the concepts of miRNA arm switching and miRNA isomerism. Finally, we offer insights into the impact of aberrant strand selection on immunity and cancer. Leading us through this journey is miR-155, a well-established regulator of immunity and cancer, and the increasing evidence that its 3p strand plays a role in these arenas. Interestingly, the miR-155-5p/-3p ratio appears to vary dependent on the timing of the immune response, and the 3p strand seems to play a regulatory role upon its partner 5p strand.
Collapse
Affiliation(s)
- Owen Dawson
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | | |
Collapse
|
66
|
Daood U, Ilyas MS, Ashraf M, Akbar M, Bapat RA, Khan AS, Pichika MR, Parolia A, Seow LL, Khoo SP, Yiu C. Biochemical changes and macrophage polarization of a silane-based endodontic irrigant in an animal model. Sci Rep 2022; 12:6354. [PMID: 35428859 PMCID: PMC9012771 DOI: 10.1038/s41598-022-10290-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/21/2022] [Indexed: 02/08/2023] Open
Abstract
Silane-based/fully hydrolyzed, endodontic irrigant exhibiting antimicrobial properties, is prepared, and is hypothesized to control macrophage polarization for tissue repair. Albino wistar rats were injected with 0.1 ml root canal irrigant, and bone marrow cells procured. Cellular mitochondria were stained with MitoTracker green along with Transmission Electron Microscopy (TEM) performed for macrophage extracellular vesicle. Bone marrow stromal cells (BMSCs) were induced for M1 and M2 polarization and Raman spectroscopy with scratch assay performed. Cell counting was used to measure cytotoxicity, and fluorescence microscopy performed for CD163. Scanning Electron Microscopy (SEM) was used to investigate interaction of irrigants with Enterococcus faecalis. K21 specimens exhibited reduction in epithelium thickness and more mitochondrial mass. EVs showed differences between all groups with decrease and increase in IL-6 and IL-10 respectively. 0.5%k21 enhanced wound healing with more fibroblastic growth inside scratch analysis along with increased inflammation-related genes (ICAM-1, CXCL10, CXCL11, VCAM-1, CCL2, and CXCL8; tissue remodelling-related genes, collagen 1, EGFR and TIMP-2 in q-PCR analysis. Sharp bands at 1643 cm-1 existed in all with variable intensities. 0.5%k21 had a survival rate of BMSCs comparable to control group. Bacteria treated with 0.5%k21/1%k21, displayed damage. Antimicrobial and reparative efficacy of k21 disinfectant is a proof of concept for enhanced killing of bacteria across root dentin acquiring functional type M2 polarization for ethnopharmacological effects.
Collapse
Affiliation(s)
- Umer Daood
- Restorative Dentistry, School of Dentistry, International Medical University Kuala Lumpur, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Bukit Jalil, Wilayah Persekutuan Kuala Lumpur, Malaysia.
| | - Muhammad Sharjeel Ilyas
- Department of Oral Biology, Post Graduate Medical Institute, 6 Birdwood Road, Lahore, Pakistan
| | - Mariam Ashraf
- Department of Oral Biology, Post Graduate Medical Institute, 6 Birdwood Road, Lahore, Pakistan
| | - Munazza Akbar
- Department of Oral Biology, Post Graduate Medical Institute, 6 Birdwood Road, Lahore, Pakistan
| | - Ranjeet Ajit Bapat
- Restorative Dentistry, School of Dentistry, International Medical University Kuala Lumpur, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Bukit Jalil, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Abdul Samad Khan
- Department of Restorative Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Mallikarjuna Rao Pichika
- Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Abhishek Parolia
- Restorative Dentistry, School of Dentistry, International Medical University Kuala Lumpur, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Bukit Jalil, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Liang Lin Seow
- Restorative Dentistry, School of Dentistry, International Medical University Kuala Lumpur, 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Bukit Jalil, Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Suan Phaik Khoo
- Division of Oral Diagnostic and Surgical Sciences, School of Dentistry, International Medical University, Kuala Lumpur, Malaysia
| | - Cynthia Yiu
- Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, 34 Hospital Road, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
67
|
Wu R, Fan X, Wang Y, Shen M, Zheng Y, Zhao S, Yang L. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Liver Immunity and Therapy. Front Immunol 2022; 13:833878. [PMID: 35309311 PMCID: PMC8930843 DOI: 10.3389/fimmu.2022.833878] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/15/2022] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs), as the most common cell source for stem cell therapy, play an important role in the modulation of innate and adaptive immune responses and have been widely used in clinical trials to treat autoimmune and inflammatory diseases. Recent experimental and clinical studies have shown that MSC-derived extracellular vesicles (MSC-EVs) can inhibit the activation and proliferation of a variety of proinflammatory cells, such as Th1, Th17 and M1 macrophages, reducing the secretion of proinflammatory cytokines, while promoting the proliferation of anti-inflammatory cells, such as M2 macrophages and Tregs, and increasing the secretion of anti-inflammatory cytokines, thus playing a role in immune regulation and exhibiting immunomodulatory functions. Besides MSC-EVs are more convenient and less immunogenic than MSCs. There is growing interest in the role of MSC-EVs in liver diseases owing to the intrinsic liver tropism of MSC-EVs. In this review, we focus on the immunomodulatory effects of MSC-EVs and summarize the pivotal roles of MSC-EVs as a cell-free therapy in liver diseases, including NAFLD, AIH, acute liver failure, liver fibrosis and hepatic ischemia–reperfusion injury. Moreover, we provide a concise overview of the potential use and limits of MSC-EVs in clinical application.
Collapse
|
68
|
MicroRNA-181c-5p modulates phagocytosis efficiency in bone marrow-derived macrophages. Inflamm Res 2022; 71:321-330. [PMID: 35020000 PMCID: PMC8919373 DOI: 10.1007/s00011-022-01539-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/09/2021] [Accepted: 01/03/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE AND DESIGN Phagocytosis and clearance of apoptotic cells are essential for inflammation resolution, efficient wound healing, and tissue homeostasis. MicroRNAs are critical modulators of macrophage polarization and function. The current study aimed to investigate the role of miR-181c-5p in macrophage phagocytosis. MATERIALS AND METHODS miR-181c-5p was identified as a potential candidate in microRNA screening of RAW264.7 macrophages fed with apoptotic cells. To investigate the role of miR-181c-5p in phagocytosis, the expression of miR-181c-5p was assessed in phagocyting bone marrow-derived macrophages. Phagocytosis efficiency was measured by fluorescence microscopy. Gain- and loss-of-function studies were performed using miR-181c-5p-specific mimic and inhibitor. The expression of the phagocytosis-associated genes and proteins of interest was evaluated by RT2 profiler PCR array and western blotting, respectively. RESULTS miR-181c-5p expression was significantly upregulated in the phagocyting macrophages. Furthermore, mimic-induced overexpression of miR-181c-5p resulted in the increased phagocytic ability of macrophages. Moreover, overexpression of miR-181c-5p resulted in upregulation of WAVE-2 in phagocyting macrophages, suggesting that miR-181c-5p may regulate cytoskeletal arrangement during macrophage phagocytosis. CONCLUSION Altogether, our data provide a novel function of miR-181c-5p in macrophage biology and suggest that targeting macrophage miR-181c-5p in injured tissues might improve clearance of dead cells and lead to efficient inflammation resolution.
Collapse
|
69
|
Ganguly S, Ghoshal B, Banerji I, Bhattacharjee S, Chakraborty S, Goswami A, Mukherjee K, Bhattacharyya SN. Leishmania survives by exporting miR-146a from infected to resident cells to subjugate inflammation. Life Sci Alliance 2022; 5:5/6/e202101229. [PMID: 35210329 PMCID: PMC8881743 DOI: 10.26508/lsa.202101229] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/24/2022] Open
Abstract
Leishmania donovani, the causative agent of visceral leishmaniasis, infects and resides within tissue macrophage cells. It is not clear how the parasite infected cells crosstalk with the noninfected cells to regulate the infection process. During infection, Leishmania adopts a dual strategy for its survival by regulating the intercellular transport of host miRNAs to restrict inflammation. The parasite, by preventing mitochondrial function of host cells, restricts the entry of liver cell derived miR-122-containing extracellular vesicles in infected macrophages to curtail the inflammatory response associated with miR-122 entry. On contrary, the parasite up-regulates the export of miR-146a from the infected macrophages. The miR-146a, associated with the extracellular vesicles released by infected cells, restricts miR-122 production in hepatocytes while polarizing neighbouring naïve macrophages to the M2 state by affecting the cytokine expression. On entering the recipient macrophages, miR-146a dominates the miRNA antagonist RNA-binding protein HuR to inhibit the expression of proinflammatory cytokine mRNAs having HuR-interacting AU-rich elements whereas up-regulates anti-inflammatory IL-10 by exporting the miR-21 to polarize the recipient cells to M2 stage.
Collapse
Affiliation(s)
- Satarupa Ganguly
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Bartika Ghoshal
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Ishani Banerji
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Ghaziabad, India
| | - Shreya Bhattacharjee
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Ghaziabad, India
| | - Sreemoyee Chakraborty
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Ghaziabad, India
| | - Avijit Goswami
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Kamalika Mukherjee
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Suvendra N Bhattacharyya
- RNA Biology Research Laboratory, Molecular Genetics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India .,Academy of Scientific and Innovative Research (AcSIR), CSIR-Human Resource Development Centre, (CSIR-HRDC) Campus, Ghaziabad, India
| |
Collapse
|
70
|
Maranini B, Ciancio G, Ferracin M, Cultrera R, Negrini M, Sabbioni S, Govoni M. microRNAs and Inflammatory Immune Response in SARS-CoV-2 Infection: A Narrative Review. Life (Basel) 2022; 12:life12020288. [PMID: 35207576 PMCID: PMC8879390 DOI: 10.3390/life12020288] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/15/2022] Open
Abstract
The current SARS-CoV-2 pandemic has emerged as an international challenge with strong medical and socioeconomic impact. The spectrum of clinical manifestations of SARS-CoV-2 is wide, covering asymptomatic or mild cases up to severe and life-threatening complications. Critical courses of SARS-CoV-2 infection are thought to be driven by the so-called “cytokine storm”, derived from an excessive immune response that induces the release of proinflammatory cytokines and chemokines. In recent years, non-coding RNAs (ncRNAs) emerged as potential diagnostic and therapeutic biomarkers in both inflammatory and infectious diseases. Therefore, the identification of SARS-CoV-2 miRNAs and host miRNAs is an important research topic, investigating the host–virus crosstalk in COVID-19 infection, trying to answer the pressing question of whether miRNA-based therapeutics can be employed to tackle SARS-CoV-2 complications. In this review, we aimed to directly address ncRNA role in SARS-CoV-2-immune system crosstalk upon COVID-19 infection, particularly focusing on inflammatory pathways and cytokine storm syndromes.
Collapse
Affiliation(s)
- Beatrice Maranini
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.C.); (M.G.)
- Correspondence:
| | - Giovanni Ciancio
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.C.); (M.G.)
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138 Bologna, Italy;
| | - Rosario Cultrera
- Infectious Diseases, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy;
| | - Massimo Negrini
- Laboratorio per le Tecnologie delle Terapie Avanzate (LTTA), Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy;
| | - Silvia Sabbioni
- Department of Life Sciences and Biotechnologies, University of Ferrara, 44121 Ferrara, Italy;
| | - Marcello Govoni
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.C.); (M.G.)
| |
Collapse
|
71
|
An Apple a Day Keeps the Doctor Away: Potential Role of miRNA 146 on Macrophages Treated with Exosomes Derived from Apples. Biomedicines 2022; 10:biomedicines10020415. [PMID: 35203624 PMCID: PMC8962404 DOI: 10.3390/biomedicines10020415] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/02/2022] [Accepted: 02/06/2022] [Indexed: 12/12/2022] Open
Abstract
The constant dialogue between the plant world and the animal world (including man among them) has been known since the time of Adam and Eve, where an apple was the origin of the evils of the world. Apart from Snow White—who might have something to object to when it comes to the use of apples—fruits, plants, and natural extracts have been known for millennia as remedies for human health-related ailments. In the light of such evidence, the aim of the present work was to investigate from a biological point of view the potential role of apple exosomes in inflammatory processes on human cells. To this end we isolated and characterized apple exosomes and treated human cells such as macrophages and NCTC L929 as cancer cells in order to evaluate the tumorigenic and anti-inflammatory effect of apple exomes. Microscopic and molecular biology analyses were conducted to characterize exosomes and to assess cell proliferation, death, and miRNA line, as well as gene expression and the uptake of exosomes by cells. The results confirm the absolute biological safety of exosomes and their anti-inflammatory effect, mediated mainly by miRNA146 production by M2 macrophages.
Collapse
|
72
|
Non-Coding RNAs in the Crosstalk between Breast Cancer Cells and Tumor-Associated Macrophages. Noncoding RNA 2022; 8:ncrna8010016. [PMID: 35202089 PMCID: PMC8874851 DOI: 10.3390/ncrna8010016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/19/2022] [Accepted: 02/01/2022] [Indexed: 11/17/2022] Open
Abstract
Non-coding RNAs (ncRNAs) play a pivotal role in regulating the tumor microenvironment (TME) by controlling gene expression at multiple levels. In tumors, ncRNAs can mediate the crosstalk between cancer cells and other cells in the TME, such as immune cells, stromal cells, and endothelial cells, influencing tumor development and progression. Tumor-associated macrophages (TAMs) are among the most abundant inflammatory cells infiltrating solid cancers that promote tumorigenesis, and their infiltration correlates with a poor prognosis in many tumors. Cancer cells produce different ncRNAs that orchestrate TAM recruitment and polarization toward a tumor-promoting phenotype. Tumor-reprogrammed macrophages shape the TME by promoting angiogenesis and tissue remodeling, and suppressing the anti-tumor activity of adaptive immune cells. TAMs can also produce ncRNA molecules that boost cancer cell proliferation and direct their phenotype and metabolic changes facilitating cancer progression and metastasis. This review will focus on the crosstalk between cancer cells and TAMs mediated by microRNAs and long non-coding RNAs during breast cancer (BC) initiation and progression.
Collapse
|
73
|
Baradaran A, Asadzadeh Z, Hemmat N, Baghbanzadeh A, Shadbad MA, Khosravi N, Derakhshani A, Alemohammad H, Afrashteh Nour M, Safarpour H, Silvestris N, Brunetti O, Baradaran B. The cross-talk between tumor-associated macrophages and tumor endothelium: Recent advances in macrophage-based cancer immunotherapy. Biomed Pharmacother 2022; 146:112588. [PMID: 35062062 DOI: 10.1016/j.biopha.2021.112588] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/02/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are among the abundant cell populations of the tumor microenvironment (TME), which have pivotal roles in tumor development, chemoresistance, immune evasion, and metastasis. Growing evidence indicates that TAMs and the cross-talk between TAMs and tumoral endothelial cells can substantially contribute to tumor angiogenesis, which is considered a vital process for cancer development. Besides, tumoral endothelial cells can regulate the leukocyte infiltration to the TME in solid cancers and contribute to immune evasion. Therefore, targeting the immunosuppressive TAMs and the cross-talk between them can be a promising strategy for improving anti-tumoral immune responses. This review aims to summarize the biology of TAMs, their recently identified roles in tumor development/angiogenesis, and recent advances in macrophage-based cancer immunotherapy approaches for treating cancers.
Collapse
Affiliation(s)
- Ali Baradaran
- Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia; Research & Development, BSD Robotics, Queensland, Australia
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Khosravi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Derakhshani
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Hajar Alemohammad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Nicola Silvestris
- Medical Oncology Unit-IRCCS IstitutoTumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology DIMO-University of Bari, Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit-IRCCS IstitutoTumori "Giovanni Paolo II" of Bari, Bari, Italy.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
74
|
Geiß C, Salas E, Guevara-Coto J, Régnier-Vigouroux A, Mora-Rodríguez RA. Multistability in Macrophage Activation Pathways and Metabolic Implications. Cells 2022; 11:404. [PMID: 35159214 PMCID: PMC8834178 DOI: 10.3390/cells11030404] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/22/2022] [Indexed: 12/22/2022] Open
Abstract
Macrophages are innate immune cells with a dynamic range of reversible activation states including the classical pro-inflammatory (M1) and alternative anti-inflammatory (M2) states. Deciphering how macrophages regulate their transition from one state to the other is key for a deeper understanding of inflammatory diseases and relevant therapies. Common regulatory motifs reported for macrophage transitions, such as positive or double-negative feedback loops, exhibit a switchlike behavior, suggesting the bistability of the system. In this review, we explore the evidence for multistability (including bistability) in macrophage activation pathways at four molecular levels. First, a decision-making module in signal transduction includes mutual inhibitory interactions between M1 (STAT1, NF-KB/p50-p65) and M2 (STAT3, NF-KB/p50-p50) signaling pathways. Second, a switchlike behavior at the gene expression level includes complex network motifs of transcription factors and miRNAs. Third, these changes impact metabolic gene expression, leading to switches in energy production, NADPH and ROS production, TCA cycle functionality, biosynthesis, and nitrogen metabolism. Fourth, metabolic changes are monitored by metabolic sensors coupled to AMPK and mTOR activity to provide stability by maintaining signals promoting M1 or M2 activation. In conclusion, we identify bistability hubs as promising therapeutic targets for reverting or blocking macrophage transitions through modulation of the metabolic environment.
Collapse
Affiliation(s)
- Carsten Geiß
- Institute for Developmental Biology and Neurobiology (IDN), Johannes Gutenberg University, 55128 Mainz, Germany;
| | - Elvira Salas
- Department of Biochemistry, Faculty of Medicine, Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica;
| | - Jose Guevara-Coto
- Department of Computer Sciences and Informatics (ECCI), Faculty of Engineering, Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica;
- Research Center for Information and Communication Technologies (CITIC), Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica
| | - Anne Régnier-Vigouroux
- Institute for Developmental Biology and Neurobiology (IDN), Johannes Gutenberg University, 55128 Mainz, Germany;
| | - Rodrigo A. Mora-Rodríguez
- Institute for Developmental Biology and Neurobiology (IDN), Johannes Gutenberg University, 55128 Mainz, Germany;
- Research Center on Surgery and Cancer (CICICA), Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica
- Research Center for Tropical Diseases (CIET), Lab of Tumor Chemosensitivity (LQT), Faculty of Microbiology, Campus Rodrigo Facio, University of Costa Rica, San José 11501-2060, Costa Rica
| |
Collapse
|
75
|
Christopoulos PF. Hacking macrophages to combat cancer and inflammatory diseases-Current advances and challenges. Scand J Immunol 2022; 95:e13140. [PMID: 35000232 DOI: 10.1111/sji.13140] [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: 08/24/2021] [Revised: 12/23/2021] [Accepted: 01/03/2022] [Indexed: 11/30/2022]
Abstract
Recently, immunotherapy has been served as the treatment of choice for various human pathophysiologies, including inflammatory diseases and cancer. Though most of the current approaches target the lymphoid compartment, macrophages intimately implicated in the induction or resolution of inflammation have rationally gained their place into the therapeutics arena. In this review, I discuss the past and novel groundbreaking strategies focusing on macrophages in different human diseases and highlight the current challenges and considerations underlying their translational potentials.
Collapse
Affiliation(s)
- Panagiotis F Christopoulos
- Department of Pathology, section of Research, Rikshospitalet, Oslo University Hospital and University of Oslo, 0424, Oslo, Norway
| |
Collapse
|
76
|
Shi L, Gu H. Emerging Nanoparticle Strategies for Modulating Tumor-Associated Macrophage Polarization. Biomolecules 2021; 11:biom11121912. [PMID: 34944555 PMCID: PMC8699338 DOI: 10.3390/biom11121912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 01/05/2023] Open
Abstract
Immunotherapy has made great progress in recent years, yet the efficacy of solid tumors remains far less than expected. One of the main hurdles is to overcome the immune-suppressive tumor microenvironment (TME). Among all cells in TME, tumor-associated macrophages (TAMs) play pivotal roles because of their abundance, multifaceted interactions to adaptive and host immune systems, as well as their context-dependent plasticity. Underlying the highly plastic characteristic, lots of research interests are focused on repolarizing TAMs from M2-like pro-tumor phenotype towards M1-like antitumoral ones. Nanotechnology offers great opportunities for targeting and modulating TAM polarization to mount the therapeutic efficacy in cancer immunotherapy. Here, this mini-review highlights those emerging nano-approaches for TAM repolarization in the last three years.
Collapse
|
77
|
Sun X, Gao J, Meng X, Lu X, Zhang L, Chen R. Polarized Macrophages in Periodontitis: Characteristics, Function, and Molecular Signaling. Front Immunol 2021; 12:763334. [PMID: 34950140 PMCID: PMC8688840 DOI: 10.3389/fimmu.2021.763334] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/04/2021] [Indexed: 12/23/2022] Open
Abstract
Periodontitis (PD) is a common chronic infectious disease. The local inflammatory response in the host may cause the destruction of supporting periodontal tissue. Macrophages play a variety of roles in PD, including regulatory and phagocytosis. Moreover, under the induction of different factors, macrophages polarize and form different functional phenotypes. Among them, M1-type macrophages with proinflammatory functions and M2-type macrophages with anti-inflammatory functions are the most representative, and both of them can regulate the tendency of the immune system to exert proinflammatory or anti-inflammatory functions. M1 and M2 macrophages are involved in the destructive and reparative stages of PD. Due to the complex microenvironment of PD, the dynamic development of PD, and various local mediators, increasing attention has been given to the study of macrophage polarization in PD. This review summarizes the role of macrophage polarization in the development of PD and its research progress.
Collapse
Affiliation(s)
- Xiaoyu Sun
- *Correspondence: Lei Zhang, ; Xiaoyu Sun,
| | | | | | | | - Lei Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, Department of Periodontology, Stomatologic Hospital & College, Anhui Medical University, Hefei, China
| | | |
Collapse
|
78
|
Fontanella RA, Scisciola L, Rizzo MR, Surina S, Sardu C, Marfella R, Paolisso G, Barbieri M. Adiponectin Related Vascular and Cardiac Benefits in Obesity: Is There a Role for an Epigenetically Regulated Mechanism? Front Cardiovasc Med 2021; 8:768026. [PMID: 34869683 PMCID: PMC8639875 DOI: 10.3389/fcvm.2021.768026] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/22/2021] [Indexed: 12/23/2022] Open
Abstract
In obesity, several epigenetic modifications, including histones remodeling, DNA methylation, and microRNAs, could accumulate and determine increased expression of inflammatory molecules, the adipokines, that in turn might induce or accelerate the onset and development of cardiovascular and metabolic disorders. In order to better clarify the potential epigenetic mechanisms underlying the modulation of the inflammatory response by adipokines, the DNA methylation profile in peripheral leukocytes of the promoter region of IL-6 and NF-kB genes and plasma miRNA-21 levels were evaluated in 356 healthy subjects, using quantitative pyrosequencing-based analysis, and correlated with plasma adiponectin levels, body fat content and the primary pro-inflammatory markers. In addition, correlation analysis of DNA methylation profiles and miRNA-21 plasma levels with intima-media thickness (IMT), a surrogate marker for early atherosclerosis, left ventricular mass (LVM), left ventricular ejection fraction (LVEF), and cardiac performance index (MPI) was also performed to evaluate any potential clinical implication in terms of cardiovascular outcome. Results achieved confirmed the role of epigenetics in the obesity-related cardiovascular complications and firstly supported the potential role of plasma miRNA-21 and IL-6 and NF-kB DNA methylation changes in nucleated blood cells as potential biomarkers for predicting cardiovascular risk in obesity. Furthermore, our results, showing a role of adiponectin in preventing epigenetic modification induced by increased adipose tissue content in obese subjects, provide new evidence of an additional mechanism underlying the anti-inflammatory properties and the cardiovascular benefits of adiponectin. The exact mechanisms underlying the obesity-related epigenetic modifications found in the blood cells and whether similar epigenetic changes reflect adipose and myocardial tissue modifications need to be further investigated in future experiments.
Collapse
Affiliation(s)
- Rosaria Anna Fontanella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Surina Surina
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.,Mediterrannea Cardiocentro, Naples, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.,Mediterrannea Cardiocentro, Naples, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
79
|
Role of microRNAs in the Pathophysiology of Ulcerative Colitis. IMMUNO 2021. [DOI: 10.3390/immuno1040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Ulcerative colitis (UC) is an intractable disorder characterized by a chronic inflammation of the colon. Studies have identified UC as a multifactorial disorder affected by both genetic and environmental factors; however, the precise mechanism remains unclear. Recent advances in the field of microRNA (miRNA) research have identified an association between this small non-coding RNA in the pathophysiology of UC and altered miRNA expression profiles in patients with UC. Nevertheless, the roles of individual miRNAs are uncertain due to heterogeneity in both research samples and clinical backgrounds. In this review, we focus on miRNA expression in colonic mucosa where inflammation occurs in UC and discuss the potential roles of individual miRNAs in disease development, outlining the pathophysiology of UC.
Collapse
|
80
|
Castillo-Salazar M, Sánchez-Muñoz F, Springall del Villar R, Navarrete-Vázquez G, Hernández-DiazCouder A, Mojica-Cardoso C, García-Jiménez S, Toledano-Jaimes C, Bernal-Fernández G. Nitazoxanide Exerts Immunomodulatory Effects on Peripheral Blood Mononuclear Cells from Type 2 Diabetes Patients. Biomolecules 2021; 11:1817. [PMID: 34944461 PMCID: PMC8699442 DOI: 10.3390/biom11121817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a low-grade inflammatory condition with abnormalities in the immune response mediated by T lymphocytes and macrophages. Drug repositioning for immunomodulatory molecules is an attractive proposal for treating T2D. Nitazoxanide (NTZ) is a broad-spectrum drug with promising immunomodulatory effects. Thus, we investigated the immunomodulatory effect of NTZ on peripheral blood mononuclear cells (PBMCs) from patients with T2D. METHODS Fifty patients with T2D were selected, and the proliferative response of T lymphocytes and the M1/M2 ratio of macrophages post cell culture were evaluated by flow cytometry, as well as measuring the concentration of cytokines by ELISA and the relative expression of microRNAs (miRNAs) related to the immune response by real-time PCR. RESULTS NTZ exerts an inhibitory effect on the cell proliferation of T lymphocytes stimulated with anti-CD3 and anti-CD28 antibodies without modifying cell viability, and significant decreases in the supernatant concentrations of interleukin (IL)-1β, IL-2, IL-6, IL-10, and IL-12. Furthermore, NTZ negatively regulates the relative expression of miR-155-5p without changes in miR-146a-5p. The M1/M2 ratio of monocytes/macrophages decreased the M1 and increased the M2 subpopulation by NTZ. CONCLUSIONS Our results suggest that NTZ exerts immunomodulatory effects on PBMCs from T2D patients, and shows potential alternative therapeutic benefits.
Collapse
Affiliation(s)
- Mauricio Castillo-Salazar
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| | - Fausto Sánchez-Muñoz
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, Mexico City 14080, Mexico; (F.S.-M.); (R.S.d.V.); (A.H.-D.)
| | - Rashidi Springall del Villar
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, Mexico City 14080, Mexico; (F.S.-M.); (R.S.d.V.); (A.H.-D.)
| | - Gabriel Navarrete-Vázquez
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| | - Adrián Hernández-DiazCouder
- Immunology Department, Instituto Nacional de Cardiología Ignacio Chávez, Tlalpan, Mexico City 14080, Mexico; (F.S.-M.); (R.S.d.V.); (A.H.-D.)
| | | | - Sara García-Jiménez
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| | - Cairo Toledano-Jaimes
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| | - Germán Bernal-Fernández
- Pharmacy Faculty, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico; (M.C.-S.); (G.N.-V.); (S.G.-J.); (C.T.-J.)
| |
Collapse
|
81
|
Xue S, Li X, Li S, Chen N, Zhan Q, Long L, Zhao J, Hou X, Yuan X. Bone fracture microenvironment responsive hydrogel for timing sequential release of cargoes. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
82
|
Lecoeur H, Prina E, Gutiérrez-Sanchez M, Späth GF. Going ballistic: Leishmania nuclear subversion of host cell plasticity. Trends Parasitol 2021; 38:205-216. [PMID: 34666937 DOI: 10.1016/j.pt.2021.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/16/2022]
Abstract
Intracellular parasites have evolved intricate strategies to subvert host cell functions for their own survival. These strategies are particularly damaging to the host if the infection involves immune cells, as illustrated by protozoan parasites of the genus Leishmania that thrive inside mononuclear phagocytic cells, causing devastating immunopathologies. While the impact of Leishmania infection on host cell phenotype and functions has been well documented, the regulatory mechanisms underlying host cell subversion were only recently investigated. Here we summarize the current knowledge on how Leishmania infection affects host nuclear activities and propose thought-provoking new concepts on the reciprocal relationship between epigenetic and transcriptional regulation in host cell phenotypic plasticity, its potential subversion by the intracellular parasite, and its relevance for host-directed therapy.
Collapse
Affiliation(s)
- Hervé Lecoeur
- Institut Pasteur, Université de Paris, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Eric Prina
- Institut Pasteur, Université de Paris, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Maria Gutiérrez-Sanchez
- Institut Pasteur, Université de Paris, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France; UMR 8076 CNRS BioCIS, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Gerald F Späth
- Institut Pasteur, Université de Paris, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France.
| |
Collapse
|
83
|
Chang YJ, Wang KC. Therapeutic perspectives of extracellular vesicles and extracellular microRNAs in atherosclerosis. CURRENT TOPICS IN MEMBRANES 2021; 87:255-277. [PMID: 34696887 DOI: 10.1016/bs.ctm.2021.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Extracellular signaling molecules, such as growth factors, cytokines, and hormones, regulate cell behaviors and fate through endocrine, paracrine, and autocrine actions and play essential roles in maintaining tissue homeostasis. MicroRNAs, an important class of posttranscriptional modulators, could stably present in extracellular space and body fluids and participate in intercellular communication in health and diseases. Indeed, recent studies demonstrated that microRNAs could be secreted through vesicular and non-vesicular routes, transported in body fluids, and then transmitted to recipient cells to regulate target gene expression and signaling events. Over the past decade, a great deal of effort has been made to investigate the functional roles of extracellular vesicles and extracellular microRNAs in pathological conditions. Emerging evidence suggests that altered levels of extracellular vesicles and extracellular microRNAs in body fluids, as part of the cellular responses to atherogenic factors, are associated with the development of atherosclerosis. This review article provides a brief overview of extracellular vesicles and perspectives of their applications as therapeutic tools for cardiovascular pathologies. In addition, we highlight the role of extracellular microRNAs in atherogenesis and offer a summary of circulating microRNAs in liquid biopsies associated with atherosclerosis.
Collapse
Affiliation(s)
- Ya-Ju Chang
- Department of Family Medicine and Public Health, School of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Kuei-Chun Wang
- School of Biological and Health Systems Engineering, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, United States.
| |
Collapse
|
84
|
Motta JM, Rumjanek VM, Mantovani A, Locati M. Tumor-Released Products Promote Bone Marrow-Derived Macrophage Survival and Proliferation. Biomedicines 2021; 9:biomedicines9101387. [PMID: 34680504 PMCID: PMC8533124 DOI: 10.3390/biomedicines9101387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Macrophages play a central role within the tumor microenvironment, with relevant implications for tumor progression. The modulation of their phenotype is one of the mechanisms used by tumors to escape from effective immune responses. This study was designed to analyze the influence of soluble products released by tumors, here represented by the tumor-conditioned media of two tumor cell lines (3LL from Lewis lung carcinoma and MN/MCA from fibrosarcoma), on murine macrophage differentiation and polarization in vitro. Data revealed that tumor-conditioned media stimulated macrophage differentiation but influenced the expression levels of macrophage polarization markers, cytokine production, and microRNAs of relevance for macrophage biology. Interestingly, tumor-derived soluble products supported the survival and proliferation rate of bone marrow precursor cells, an effect observed even with mature macrophages in the presence of M2 but not M1 inducers. Despite presenting low concentrations of macrophage colony-stimulating factor (M-CSF), tumor-conditioned media alone also supported the proliferation of cells to a similar extent as exogenous M-CSF. This effect was only evident in cells positive for the expression of the M-CSF receptor (CD115) and occurred preferentially within the CD16+ subset. Blocking CD115 partially reversed the effect on proliferation. These results suggest that tumors release soluble products that not only promote macrophage development from bone marrow precursors but also stimulate the proliferation of cells with specific phenotypes that could support protumoral functions.
Collapse
Affiliation(s)
- Juliana Maria Motta
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.M.M.); (V.M.R.)
- Humanitas Clinical and Research Center—IRCCS, 20089 Rozzano, Italy;
| | - Vivian Mary Rumjanek
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (J.M.M.); (V.M.R.)
| | | | - Massimo Locati
- Humanitas Clinical and Research Center—IRCCS, 20089 Rozzano, Italy;
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
- Correspondence:
| |
Collapse
|
85
|
Azpiroz MA, Orguilia L, Palacio MI, Malpartida A, Mayol S, Mor G, Gutiérrez G. Potential biomarkers of infertility associated with microbiome imbalances. Am J Reprod Immunol 2021; 86:e13438. [PMID: 33960055 PMCID: PMC8464490 DOI: 10.1111/aji.13438] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 12/13/2022] Open
Abstract
PROBLEM The aim of this study was to investigate the possible relationship between vaginal/rectal microbiome disbalances and miRNA expression with infertility. METHOD OF STUDY Observational, exploratory, preliminary study. A total of 287 multiple IVF failure infertile patients were recruited. Twenty fertile women, not IVF failure, were recruited as the control group. Swab samples were collected from the vagina and rectum. Microbial composition by NGS and miRNA expression by real-time PCR of vaginal and rectal samples was measured. Immunometabolic markers from blood (insulin, vitamin D, LDL-cholesterol, ANA, TPO, Tg, and ASCA antibodies) and saliva (sIgA) were analyzed. RESULT(S) Infertile patients showed a lower bacterial richness and increased Firmicutes/Bacteroidetes ratio at rectal level and an increased Lactobacillus brevis/Lactobacillus iners ratio in vaginal samples regarding the fertile group. In the same rectal swab samples, we found that miR-21-5p, which is associated with tight junction disruption and yeast overgrowth, is upregulated and that miR-155-5p, which is associated with inflammation, is overexpressed in the unexplained infertile group (*p < .05). These deregulated miRNAs were also upregulated in the vaginal samples from the same patients (*p < .05). CONCLUSION miRNAs could be potential biomarkers of the inflammatory impact of microbiome disbalances in unexplained infertile women.
Collapse
Affiliation(s)
| | - Lucila Orguilia
- Inmunogenesis, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | | | | | | | - Gil Mor
- Wayne State University, Detroit, MI, USA
| | | |
Collapse
|
86
|
Rashidi S, Mansouri R, Ali-Hassanzadeh M, Ghani E, Barazesh A, Karimazar M, Nguewa P, Carrera Silva EA. Highlighting the interplay of microRNAs from Leishmania parasites and infected-host cells. Parasitology 2021; 148:1434-1446. [PMID: 34218829 PMCID: PMC11010138 DOI: 10.1017/s0031182021001177] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/10/2021] [Accepted: 06/27/2021] [Indexed: 02/05/2023]
Abstract
Leishmania parasites, the causative agents of leishmaniasis, are protozoan parasites with the ability to modify the signalling pathway and cell responses of their infected host cells. These parasite strategies alter the host cell environment and conditions favouring their replication, survival and pathogenesis. Since microRNAs (miRNAs) are able to post-transcriptionally regulate gene expression processes, these biomolecules can exert critical roles in controlling Leishmania-host cell interplay. Therefore, the identification of relevant miRNAs differentially expressed in Leishmania parasites as well as in infected cells, which affect the host fitness, could be critical to understand the infection biology, pathogenicity and immune response against these parasites. Accordingly, the current review aims to address the differentially expressed miRNAs in both, the parasite and infected host cells and how these biomolecules change cell signalling and host immune responses during infection. A deep understanding of these processes could provide novel guidelines and therapeutic strategies for managing and treating leishmaniasis.
Collapse
Affiliation(s)
- Sajad Rashidi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Esmaeel Ghani
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afshin Barazesh
- Department of Microbiology and Parasitology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammadreza Karimazar
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paul Nguewa
- University of Navarra, ISTUN Instituto de Salud Tropical, Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), c/Irunlarrea 1, 31008Pamplona, Spain
| | | |
Collapse
|
87
|
Kumar P, Zadjali F, Yao Y, Bissler JJ. Renal cystic disease in tuberous sclerosis complex. Exp Biol Med (Maywood) 2021; 246:2111-2117. [PMID: 34488473 DOI: 10.1177/15353702211038378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is associated with TSC1 or TSC2 gene mutations resulting in hyperactivation of the mTORC1 pathway. This mTORC1 activation is associated with abnormal tissue development and proliferation such that in the kidney there are both solid tumors and cystic lesions. This review summarizes recent advances in tuberous sclerosis complex nephrology and focuses on the genetics and cell biology of tuberous sclerosis complex renal disease, highlighting a role of extracellular vesicles and the innate immune system in disease pathogenesis.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - Fahad Zadjali
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, PC 123, Oman
| | - Ying Yao
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - John J Bissler
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Pediatric Medicine Department, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
88
|
Chamseddine AN, Assi T, Mir O, Chouaib S. Modulating tumor-associated macrophages to enhance the efficacy of immune checkpoint inhibitors: A TAM-pting approach. Pharmacol Ther 2021; 231:107986. [PMID: 34481812 DOI: 10.1016/j.pharmthera.2021.107986] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAM) plasticity and diversity are both essential hallmarks of the monocyte-macrophage lineage and the tumor-derived inflammation. TAM exemplify the perfect adaptable cell with dynamic phenotypic modifications that reflect changes in their functional polarization status. Under several tumor microenvironment (TME)-related cues, TAM shift their polarization, hence promoting or halting cancer progression. Immune checkpoint inhibitors (ICI) displayed unprecedented clinical responses in various refractory cancers; but only approximately a third of patients experienced durable responses. It is, therefore, crucial to enhance the response rate of immunotherapy. Several mechanisms of resistance to ICI have been elucidated including TAM role with its essential immunosuppressive functions that reduce both anti-tumor immunity and the subsequent ICI efficacy. In the past few years, thorough research has led to a better understanding of TAM biology and innovative approaches can now be adapted through targeting macrophages' recruitment axis as well as TAM activation and polarization status within the TME. Some of these therapeutic strategies are currently being evaluated in several clinical trials in association with ICI agents. This combination between TAM modulation and ICI allows targeting TAM intrinsic immunosuppressive functions and tumor-promoting factors as well as overcoming ICI resistance. Hence, such strategies, with a better understanding of the mechanisms driving TAM modulation, may have the potential to optimize ICI efficacy.
Collapse
Affiliation(s)
- Ali N Chamseddine
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Biostatistics and Epidemiology, CESP INSERM U1018, OncoStat, Gustave Roussy, F-94805, Villejuif, France.
| | - Tarek Assi
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France
| | - Olivier Mir
- Department of Medical Oncology, Gustave Roussy, F-94805, Villejuif, France; Department of Pharmacology, Gustave Roussy, F-94805, Villejuif, France; Department of Ambulatory Care, Gustave Roussy, F-94805, Villejuif, France
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, F-94805, Villejuif, France
| |
Collapse
|
89
|
Theobald SJ, Simonis A, Georgomanolis T, Kreer C, Zehner M, Eisfeld HS, Albert M, Chhen J, Motameny S, Erger F, Fischer J, Malin JJ, Gräb J, Winter S, Pouikli A, David F, Böll B, Koehler P, Vanshylla K, Gruell H, Suárez I, Hallek M, Fätkenheuer G, Jung N, Cornely OA, Lehmann C, Tessarz P, Altmüller J, Nürnberg P, Kashkar H, Klein F, Koch M, Rybniker J. Long-lived macrophage reprogramming drives spike protein-mediated inflammasome activation in COVID-19. EMBO Mol Med 2021; 13:e14150. [PMID: 34133077 PMCID: PMC8350892 DOI: 10.15252/emmm.202114150] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 01/12/2023] Open
Abstract
Innate immunity triggers responsible for viral control or hyperinflammation in COVID-19 are largely unknown. Here we show that the SARS-CoV-2 spike protein (S-protein) primes inflammasome formation and release of mature interleukin-1β (IL-1β) in macrophages derived from COVID-19 patients but not in macrophages from healthy SARS-CoV-2 naïve individuals. Furthermore, longitudinal analyses reveal robust S-protein-driven inflammasome activation in macrophages isolated from convalescent COVID-19 patients, which correlates with distinct epigenetic and gene expression signatures suggesting innate immune memory after recovery from COVID-19. Importantly, we show that S-protein-driven IL-1β secretion from patient-derived macrophages requires non-specific monocyte pre-activation in vivo to trigger NLRP3-inflammasome signaling. Our findings reveal that SARS-CoV-2 infection causes profound and long-lived reprogramming of macrophages resulting in augmented immunogenicity of the SARS-CoV-2 S-protein, a major vaccine antigen and potent driver of adaptive and innate immune signaling.
Collapse
Affiliation(s)
- Sebastian J Theobald
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Alexander Simonis
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Theodoros Georgomanolis
- Faculty of Medicine and University Hospital of CologneCologne Center for Genomics (CCG)University of CologneCologneGermany
| | - Christoph Kreer
- Laboratory of Experimental ImmunologyInstitute of VirologyFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Matthias Zehner
- Laboratory of Experimental ImmunologyInstitute of VirologyFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Hannah S Eisfeld
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Marie‐Christine Albert
- Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Institute for Medical Microbiology, Immunology and Hygiene (IMMIH)University of CologneCologneGermany
| | - Jason Chhen
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Susanne Motameny
- Faculty of Medicine and University Hospital of CologneCologne Center for Genomics (CCG)University of CologneCologneGermany
| | - Florian Erger
- Faculty of Medicine and University Hospital of CologneCologne Center for Genomics (CCG)University of CologneCologneGermany
- Faculty of MedicineInstitute of Human GeneticsUniversity Hospital CologneCologneGermany
| | - Julia Fischer
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- German Center for Infection Research (DZIF), Partner Site Bonn‐CologneCologneGermany
| | - Jakob J Malin
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Jessica Gräb
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Sandra Winter
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - Andromachi Pouikli
- Max Planck Research Group “Chromatin and Ageing”Max Planck Institute for Biology of AgeingCologneGermany
| | - Friederike David
- Faculty of Medicine and University Hospital of CologneCologne Center for Genomics (CCG)University of CologneCologneGermany
| | - Boris Böll
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Philipp Koehler
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| | - Kanika Vanshylla
- Laboratory of Experimental ImmunologyInstitute of VirologyFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Henning Gruell
- Laboratory of Experimental ImmunologyInstitute of VirologyFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Isabelle Suárez
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- German Center for Infection Research (DZIF), Partner Site Bonn‐CologneCologneGermany
| | - Michael Hallek
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Gerd Fätkenheuer
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- German Center for Infection Research (DZIF), Partner Site Bonn‐CologneCologneGermany
| | - Norma Jung
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- German Center for Infection Research (DZIF), Partner Site Bonn‐CologneCologneGermany
| | - Oliver A Cornely
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- German Center for Infection Research (DZIF), Partner Site Bonn‐CologneCologneGermany
| | - Clara Lehmann
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- German Center for Infection Research (DZIF), Partner Site Bonn‐CologneCologneGermany
| | - Peter Tessarz
- Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Max Planck Research Group “Chromatin and Ageing”Max Planck Institute for Biology of AgeingCologneGermany
| | - Janine Altmüller
- Faculty of Medicine and University Hospital of CologneCologne Center for Genomics (CCG)University of CologneCologneGermany
| | - Peter Nürnberg
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCologne Center for Genomics (CCG)University of CologneCologneGermany
| | - Hamid Kashkar
- Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Institute for Medical Microbiology, Immunology and Hygiene (IMMIH)University of CologneCologneGermany
| | - Florian Klein
- Laboratory of Experimental ImmunologyInstitute of VirologyFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- German Center for Infection Research (DZIF), Partner Site Bonn‐CologneCologneGermany
| | - Manuel Koch
- Medical FacultyInstitute for Dental Research and Oral Musculoskeletal BiologyUniversity of CologneCologneGermany
- Medical FacultyCenter for BiochemistryUniversity of CologneCologneGermany
| | - Jan Rybniker
- Department I of Internal MedicineFaculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Faculty of Medicine and University Hospital of CologneCenter for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
- German Center for Infection Research (DZIF), Partner Site Bonn‐CologneCologneGermany
| |
Collapse
|
90
|
Monastirioti A, Papadaki C, Rounis K, Kalapanida D, Mavroudis D, Agelaki S. A Prognostic Role for Circulating microRNAs Involved in Macrophage Polarization in Advanced Non-Small Cell Lung Cancer. Cells 2021; 10:cells10081988. [PMID: 34440757 PMCID: PMC8391493 DOI: 10.3390/cells10081988] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
Circulating microRNAs (miRNAs) are key regulators of the crosstalk between tumor cells and immune response. In the present study, miRNAs (let-7c, miR-26a, miR-30d, miR-98, miR-195, miR-202) reported to be involved in the polarization of macrophages were examined for associations with the outcomes of non-small cell lung cancer (NSCLC) patients (N = 125) treated with first-line platinum-based chemotherapy. RT-qPCR was used to analyze miRNA expression levels in the plasma of patients prior to treatment. In our results, disease progression was correlated with high miR-202 expression (HR: 2.335; p = 0.040). Additionally, high miR-202 expression was characterized as an independent prognostic factor for shorter progression-free survival (PFS, HR: 1.564; p = 0.021) and overall survival (OS, HR: 1.558; p = 0.024). Moreover, high miR-202 independently predicted shorter OS (HR: 1.989; p = 0.008) in the non-squamous (non-SqCC) subgroup, and high miR-26a was correlated with shorter OS in the squamous (SqCC) subgroup (10.07 vs. 13.53 months, p = 0.033). The results of the present study propose that the expression levels of circulating miRNAs involved in macrophage polarization are correlated with survival measures in NSCLC patients, and their role as potential biomarkers merits further investigation.
Collapse
Affiliation(s)
- Alexia Monastirioti
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Vassilika Vouton, 71003 Heraklion, Crete, Greece; (A.M.); (C.P.); (D.M.)
| | - Chara Papadaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Vassilika Vouton, 71003 Heraklion, Crete, Greece; (A.M.); (C.P.); (D.M.)
| | - Konstantinos Rounis
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Heraklion, Crete, Greece; (K.R.); (D.K.)
| | - Despoina Kalapanida
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Heraklion, Crete, Greece; (K.R.); (D.K.)
| | - Dimitrios Mavroudis
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Vassilika Vouton, 71003 Heraklion, Crete, Greece; (A.M.); (C.P.); (D.M.)
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Heraklion, Crete, Greece; (K.R.); (D.K.)
| | - Sofia Agelaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Vassilika Vouton, 71003 Heraklion, Crete, Greece; (A.M.); (C.P.); (D.M.)
- Department of Medical Oncology, University General Hospital of Heraklion, Vassilika Vouton, 71110 Heraklion, Crete, Greece; (K.R.); (D.K.)
- Correspondence: ; Tel.: +30-281-0392438
| |
Collapse
|
91
|
Moradi-Chaleshtori M, Shojaei S, Mohammadi-Yeganeh S, Hashemi SM. Transfer of miRNA in tumor-derived exosomes suppresses breast tumor cell invasion and migration by inducing M1 polarization in macrophages. Life Sci 2021; 282:119800. [PMID: 34245773 DOI: 10.1016/j.lfs.2021.119800] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/20/2021] [Accepted: 06/28/2021] [Indexed: 01/20/2023]
Abstract
AIMS Macrophage repolarization from M1 to M2 phenotype is one of the hallmarks of malignancy. M2 macrophages are the most represented population in the tumor microenvironment and play an active role in tumor progression. In recent years, microRNAs (miRNAs) have been identified as a regulator of macrophage polarization. MAIN METHODS In this study, miR-130 was delivered to M2 macrophages using tumor-derived exosomes. Then, we evaluated the macrophage polarization status by assessment of specific markers and cytokines for M1 and M2 phenotype. The phagocytosis ability of macrophages was also investigated. Additionally, we performed migration and invasion assays to detect the effect of macrophage reprogramming on breast cancer cells migration and invasion. KEY FINDINGS The findings of the current study indicated that exosomes efficiently delivered miR-130 into macrophages. Delivery of miR-130 into macrophages resulted in upregulation of M1 specific markers and cytokines, including CD86, Irf5, Nos2, TNF-α, and IL-1β and downregulation of M2 specific markers and cytokines, including CD206, Ym1, Arg, TGF-β, and IL-10. The phagocytosis ability of macrophages also enhanced after treatment with miRNA-loaded exosomes. Furthermore, migration and invasion assays demonstrated reduced ability of 4T1 breast cancer cells for migration and invasion after macrophages reprogramming. SIGNIFICANCE These observations suggest that repolarization of M2 macrophages to M1 phenotype using miRNA-containing exosomes can be a therapeutic strategy against tumor invasion and metastasis in breast cancer.
Collapse
Affiliation(s)
- Maryam Moradi-Chaleshtori
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Shojaei
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Mohammadi-Yeganeh
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyed Mahmoud Hashemi
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
92
|
The Role of miRNAs in Extracellular Matrix Repair and Chronic Fibrotic Lung Diseases. Cells 2021; 10:cells10071706. [PMID: 34359876 PMCID: PMC8304879 DOI: 10.3390/cells10071706] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
The lung extracellular matrix (ECM) plays a key role in the normal architecture of the lung, from embryonic lung development to mechanical stability and elastic recoil of the breathing adult lung. The lung ECM can modulate the biophysical environment of cells through ECM stiffness, porosity, topography and insolubility. In a reciprocal interaction, lung ECM dynamics result from the synthesis, degradation and organization of ECM components by the surrounding structural and immune cells. Repeated lung injury and repair can trigger a vicious cycle of aberrant ECM protein deposition, accompanied by elevated ECM stiffness, which has a lasting effect on cell and tissue function. The processes governing the resolution of injury repair are regulated by several pathways; however, in chronic lung diseases such as asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary disease (IPF) these processes are compromised, resulting in impaired cell function and ECM remodeling. Current estimates show that more than 60% of the human coding transcripts are regulated by miRNAs. miRNAs are small non-coding RNAs that regulate gene expressions and modulate cellular functions. This review is focused on the current knowledge of miRNAs in regulating ECM synthesis, degradation and topography by cells and their dysregulation in asthma, COPD and IPF.
Collapse
|
93
|
Longo V, Longo A, Adamo G, Fiannaca A, Picciotto S, La Paglia L, Romancino D, La Rosa M, Urso A, Cibella F, Bongiovanni A, Colombo P. 2,2'4,4'-Tetrabromodiphenyl Ether (PBDE-47) Modulates the Intracellular miRNA Profile, sEV Biogenesis and Their miRNA Cargo Exacerbating the LPS-Induced Pro-Inflammatory Response in THP-1 Macrophages. Front Immunol 2021; 12:664534. [PMID: 34025666 PMCID: PMC8138315 DOI: 10.3389/fimmu.2021.664534] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/22/2021] [Indexed: 12/30/2022] Open
Abstract
The 2,2’4,4’-tetrabromodiphenyl ether (PBDE-47) is one of the most prominent PBDE congeners detected in the environment and in animal and human tissues. Animal model experiments suggested the occurrence of PBDE-induced immunotoxicity leading to different outcomes and recently we demonstrated that this substance can impair macrophage and basophil activities. In this manuscript, we decided to further examine the effects induced by PBDE-47 treatment on innate immune response by looking at the intracellular expression profile of miRNAs as well as the biogenesis, cargo content and activity of human M(LPS) macrophage cell-derived small extracellular vesicles (sEVs). Microarray and in silico analysis demonstrated that PBDE-47 can induce some epigenetic effects in M(LPS) THP-1 cells modulating the expression of a set of intracellular miRNAs involved in biological pathways regulating the expression of estrogen-mediated signaling and immune responses with particular reference to M1/M2 differentiation. In addition to the cell-intrinsic modulation of intracellular miRNAs, we demonstrated that PBDE-47 could also interfere with the biogenesis of sEVs increasing their number and selecting a de novo population of sEVs. Moreover, PBDE-47 induced the overload of specific immune related miRNAs in PBDE-47 derived sEVs. Finally, culture experiments with naïve M(LPS) macrophages demonstrated that purified PBDE-47 derived sEVs can modulate macrophage immune response exacerbating the LPS-induced pro-inflammatory response inducing the overexpression of the IL-6 and the MMP9 genes. Data from this study demonstrated that PBDE-47 can perturb the innate immune response at different levels modulating the intracellular expression of miRNAs but also interfering with the biogenesis, cargo content and functional activity of M(LPS) macrophage cell-derived sEVs.
Collapse
Affiliation(s)
- Valeria Longo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Alessandra Longo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Giorgia Adamo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Antonino Fiannaca
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Sabrina Picciotto
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Laura La Paglia
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Daniele Romancino
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Massimo La Rosa
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Alfonso Urso
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Fabio Cibella
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Antonella Bongiovanni
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Paolo Colombo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| |
Collapse
|
94
|
Venosa A, Smith LC, Gow AJ, Zarbl H, Laskin JD, Laskin DL. Macrophage activation in the lung during the progression of nitrogen mustard induced injury is associated with histone modifications and altered miRNA expression. Toxicol Appl Pharmacol 2021; 423:115569. [PMID: 33971176 DOI: 10.1016/j.taap.2021.115569] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022]
Abstract
Activated macrophages have been implicated in lung injury and fibrosis induced by the cytotoxic alkylating agent, nitrogen mustard (NM). Herein, we determined if macrophage activation is associated with histone modifications and altered miRNA expression. Treatment of rats with NM (0.125 mg/kg, i.t.) resulted in increases in phosphorylation of H2A.X in lung macrophages at 1 d and 3 d post-exposure. This DNA damage response was accompanied by methylation of histone (H) 3 lysine (K) 4 and acetylation of H3K9, marks of transcriptional activation, and methylation of H3K36 and H3K9, marks associated with transcriptional repression. Increases in histone acetyl transferase and histone deacetylase were also observed in macrophages 1 d and 28 d post-NM exposure. PCR array analysis of miRNAs (miR)s involved in inflammation and fibrosis revealed unique and overlapping expression profiles in macrophages isolated 1, 3, 7, and 28 d post-NM. An IPA Core Analysis of predicted mRNA targets of differentially expressed miRNAs identified significant enrichment of Diseases and Functions related to cell cycle arrest, apoptosis, cell movement, cell adhesion, lipid metabolism, and inflammation 1 d and 28 d post NM. miRNA-mRNA interaction network analysis revealed highly connected miRNAs representing key upstream regulators of mRNAs involved in significantly enriched pathways including miR-34c-5p and miR-27a-3p at 1 d post NM and miR-125b-5p, miR-16-5p, miR-30c-5p, miR-19b-3p and miR-148b-3p at 28 d post NM. Collectively, these data show that NM promotes histone remodeling and alterations in miRNA expression linked to lung macrophage responses during inflammatory injury and fibrosis.
Collapse
Affiliation(s)
- Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - L Cody Smith
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ 08854, USA
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ 08854, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Helmut Zarbl
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA; Department of Environmental and Occupational Health and Justice, Rutgers University School of Public Health, Piscataway, NJ 08854, USA
| | - Jeffrey D Laskin
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA; Department of Environmental and Occupational Health and Justice, Rutgers University School of Public Health, Piscataway, NJ 08854, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Rutgers University Ernest Mario School of Pharmacy, Piscataway, NJ 08854, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
95
|
Trusinskis K, Lapsovs M, Paeglite S, Knoka E, Caunite L, Mazule M, Briede I, Jegere S, Kumsars I, Narbute I, Konrade I, Erglis A, Lejnieks A. Plasma circulating microRNAs in patients with stable coronary artery disease - Impact of different cardiovascular risk profiles and glomerular filtration rates. J Clin Transl Res 2021; 7:270-276. [PMID: 34104831 PMCID: PMC8177841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/27/2021] [Accepted: 03/27/2021] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND AND AIM Plasma circulating microRNA (miRNA)-126, -145, and -155 are associated with vascular remodeling, atherosclerotic lesion formation, and plaque vulnerability. In this study, we evaluated the levels of plasma circulating miRNAs in patients with stable coronary artery disease (CAD), different cardiovascular risk profiles, and different glomerular filtration rates (GFR). METHODS AND RESULTS Forty patients with stable CAD admitted for elective percutaneous coronary intervention (PCI) were enrolled in a prospective study. Before PCI, fasting blood samples were obtained to evaluate clinical parameters and miRNA-126 and miRNA-155 expression. The GFR was calculated by the MDRD and CKD-EPI formulas, and the severity of CAD was calculated according to the SYNTAX score. All these parameters were correlated with miRNAs. The association between miRNA levels and clinical characteristics was evaluated. The expression of miRNA-126 positively correlated with a higher SYNTAX score (r = 0.337; p=0.034); however, no significant correlations between miR-126, GFR, and clinical characteristics were observed. Higher plasma levels of miRNA-155 correlated with increased levels of triglycerides (r = 0.317; P = 0.049), C-peptide (r = 0.452; P = 0.011), and the HOMA index (r = 0.447; P = 0.012) and a higher body mass index (BMI) (r = 0.385; P = 0.015). GFR and miRNA-155 (MDRD - Rho=0.353; P = 0.027. CKD-EPI - Rho=0.357; P = 0.026) were found to have a moderate correlation, although miRNA-155 had no correlation with the SYNTAX score. CONCLUSION Plasma circulating miRNA-126 levels were increased in patients with severe atherosclerosis as determined by the SYNTAX score. Elevated miRNA-155 expression was observed in patients with Stage 1 GFR but was lower in patients with Stages 2 and 3 GFR. Plasma circulating miRNA-155 had positive correlations with higher levels of BMI, HOMA index, C-peptide, and triglycerides. RELEVANCE FOR PATIENTS Although further investigations are needed to confirm the role of miRNA-155 and miRNA-126, they may serve as potential biomarkers detecting severity of CAD, lowering of kidney function and metabolic syndrome.
Collapse
Affiliation(s)
- Karlis Trusinskis
- Latvian Centre of Cardiology, Pauls Stradins Clinical University Hospital, Riga, LV-1002, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, LV-1007, Latvia
| | - Maris Lapsovs
- Department of Internal Diseases, Riga Stradins University, Riga, LV-1007, Latvia
| | - Sandra Paeglite
- Department of Internal Diseases, Riga Stradins University, Riga, LV-1007, Latvia
| | - Evija Knoka
- Latvian Centre of Cardiology, Pauls Stradins Clinical University Hospital, Riga, LV-1002, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, LV-1007, Latvia
| | - Laima Caunite
- Latvian Centre of Cardiology, Pauls Stradins Clinical University Hospital, Riga, LV-1002, Latvia
- Department of Internal Diseases, Riga Stradins University, Riga, LV-1007, Latvia
| | - Mairita Mazule
- Department of Internal Diseases, Riga Stradins University, Riga, LV-1007, Latvia
- Faculty of Medicine, University of Latvia, Riga, LV-1050, Latvia
| | - Ieva Briede
- Latvian Centre of Cardiology, Pauls Stradins Clinical University Hospital, Riga, LV-1002, Latvia
| | - Sanda Jegere
- Latvian Centre of Cardiology, Pauls Stradins Clinical University Hospital, Riga, LV-1002, Latvia
| | - Indulis Kumsars
- Latvian Centre of Cardiology, Pauls Stradins Clinical University Hospital, Riga, LV-1002, Latvia
| | - Inga Narbute
- Latvian Centre of Cardiology, Pauls Stradins Clinical University Hospital, Riga, LV-1002, Latvia
| | - Ilze Konrade
- Department of Endocrinology and Internal Medicine, Riga East Clinical University Hospital, Riga, LV-1038, Latvia
| | - Andrejs Erglis
- Latvian Centre of Cardiology, Pauls Stradins Clinical University Hospital, Riga, LV-1002, Latvia
- Faculty of Medicine, University of Latvia, Riga, LV-1050, Latvia
| | - Aivars Lejnieks
- Department of Internal Diseases, Riga Stradins University, Riga, LV-1007, Latvia
- Department of Endocrinology and Internal Medicine, Riga East Clinical University Hospital, Riga, LV-1038, Latvia
| |
Collapse
|
96
|
Role of p53-miRNAs circuitry in immune surveillance and cancer development: A potential avenue for therapeutic intervention. Semin Cell Dev Biol 2021; 124:15-25. [PMID: 33875349 DOI: 10.1016/j.semcdb.2021.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/07/2021] [Accepted: 04/02/2021] [Indexed: 12/16/2022]
Abstract
The genome's guardian, p53, is a master regulatory transcription factor that occupies sequence-specific response elements in many genes and modulates their expression. The target genes transcribe both coding RNA and non-coding RNA involved in regulating several biological processes such as cell division, differentiation, and cell death. Besides, p53 also regulates tumor immunology via regulating the molecules related to the immune response either directly or via regulating other molecules, including microRNAs (miRNAs). At the post-transcriptional level, the regulations of genes by miRNAs have been an emerging mechanism. Interestingly, p53 and various miRNAs cross-talk at different regulation levels. The cross-talk between p53 and miRNAs creates loops, turns, and networks that can influence cell metabolism, cell fate, cellular homeostasis, and tumor formation. Further, p53-miRNAs circuit has also been insinuated in the regulation of immune surveillance machinery. There are several examples of p53-miRNAs circuitry where p53 regulates immunomodulatory miRNA expression, such as miR-34a and miR-17-92. Similarly, a reverse process occurs in which miRNAs such as miR-125b and miR-let-7 regulate the expression of p53. Thus, the p53-miRNAs circuitry connects the immunomodulatory pathways and may shift the pro-inflammatory balance towards the pro-tumorigenic condition. In this review, we discuss the influence of p53-miRNAs circuitry in modulating the immune response in cancer development. We assume that thorough studies on the p53-miRNAs circuitry in various cancers may prove useful in developing effective new cancer therapeutics for successfully combating this disease.
Collapse
|
97
|
Kimm MA, Klenk C, Alunni-Fabbroni M, Kästle S, Stechele M, Ricke J, Eisenblätter M, Wildgruber M. Tumor-Associated Macrophages-Implications for Molecular Oncology and Imaging. Biomedicines 2021; 9:biomedicines9040374. [PMID: 33918295 PMCID: PMC8066018 DOI: 10.3390/biomedicines9040374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages (TAMs) represent the largest group of leukocytes within the tumor microenvironment (TME) of solid tumors and orchestrate the composition of anti- as well as pro-tumorigenic factors. This makes TAMs an excellent target for novel cancer therapies. The plasticity of TAMs resulting in varying membrane receptors and expression of intracellular proteins allow the specific characterization of different subsets of TAMs. Those markers similarly allow tracking of TAMs by different means of molecular imaging. This review aims to provides an overview of the origin of tumor-associated macrophages, their polarization in different subtypes, and how characteristic markers of the subtypes can be used as targets for molecular imaging and theranostic approaches.
Collapse
Affiliation(s)
- Melanie A. Kimm
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Christopher Klenk
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Marianna Alunni-Fabbroni
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Sophia Kästle
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Matthias Stechele
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Michel Eisenblätter
- Department of Diagnostic and Interventional Radiology, Freiburg University Hospital, 79106 Freiburg, Germany;
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
- Correspondence: ; Tel.: +49-0-89-4400-76640
| |
Collapse
|
98
|
Recchiuti A, Patruno S, Mattoscio D, Isopi E, Pomilio A, Lamolinara A, Iezzi M, Pecce R, Romano M. Resolvin D1 and D2 reduce SARS-CoV-2-induced inflammatory responses in cystic fibrosis macrophages. FASEB J 2021; 35:e21441. [PMID: 33749902 PMCID: PMC8250053 DOI: 10.1096/fj.202001952r] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
An excessive, non‐resolving inflammatory response underlies severe COVID‐19 that may have fatal outcomes. Therefore, the investigation of endogenous pathways leading to resolution of inflammation is of interest to uncover strategies for mitigating inflammation in people with SARS‐CoV‐2 infection. This becomes particularly urgent in individuals with preexisting pathologies characterized by chronic respiratory inflammation and prone to bacterial infection, such as cystic fibrosis (CF). Here, we analyzed the immune responses to SARS‐CoV‐2 virion spike 1 glycoprotein (S1) of macrophages (MΦ) from volunteers with and without CF and tested the efficacy of resolvins (Rv) D1 and D2 in regulating the inflammatory and antimicrobial functions of MΦ exposed to S1. S1 significantly increased chemokine release, including interleukin (IL)‐8, in CF and non‐CF MΦ, while it enhanced IL‐6 and tumor necrosis factor (TNF)‐α in non‐CF MΦ, but not in CF cells. S1 also triggered the biosynthesis of RvD1 and modulated microRNAs miR‐16, miR‐29a, and miR‐103, known to control the inflammatory responses. RvD1 and RvD2 treatment abated S1‐induced inflammatory responses in CF and non‐CF MΦ, significantly reducing the release of select chemokines and cytokines including IL‐8 and TNF‐α. RvD1 and RvD2 both restored the expression of miR‐16 and miR‐29a, while selectively increasing miR‐223 and miR‐125a, which are involved in NF‐κB activation and MΦ inflammatory polarization. During Pseudomonas aeruginosa infection, S1 stimulated the MΦ phagocytic activity that was further enhanced by RvD1 and RvD2. These results provide a map of molecular responses to SARS‐CoV‐2 in MΦ, key determinants of COVID‐19‐related inflammation, unveiling some peculiarity in the response of cells from individuals with CF. They also demonstrate beneficial, regulatory actions of RvD1 and RvD2 on SARS‐CoV‐2‐induced inflammation.
Collapse
Affiliation(s)
- Antonio Recchiuti
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Sara Patruno
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Domenico Mattoscio
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Elisa Isopi
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Antonella Pomilio
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Alessia Lamolinara
- Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Department of Neurosciences, Imaging and Clinical Sciences, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Manuela Iezzi
- Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Department of Neurosciences, Imaging and Clinical Sciences, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Romina Pecce
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Mario Romano
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| |
Collapse
|
99
|
Walsh KB, Zimmerman KD, Zhang X, Demel SL, Luo Y, Langefeld CD, Wohleb E, Schulert G, Woo D, Adeoye O. miR-181a Mediates Inflammatory Gene Expression After Intracerebral Hemorrhage: An Integrated Analysis of miRNA-seq and mRNA-seq in a Swine ICH Model. J Mol Neurosci 2021; 71:1802-1814. [PMID: 33755911 DOI: 10.1007/s12031-021-01815-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/15/2021] [Indexed: 12/22/2022]
Abstract
Intracerebral hemorrhage (ICH) is a severe neurological disorder with no proven treatment. Inflammation after ICH contributes to clinical outcomes, but the relevant molecular mechanisms remain poorly understood. In studies of peripheral leukocyte counts and mRNA-sequencing (mRNA-seq), our group previously reported that monocytes and Interleukin-8 (IL-8) were important contributors to post-ICH inflammation. microRNA (miRNA) are powerful regulators of gene expression and promising therapeutic targets. We now report findings from an integrated analysis of miRNA-seq and mRNA-seq in peripheral blood mononuclear cells (PBMCs) from a swine ICH model. In 10 pigs, one PBMC sample was collected immediately prior to ICH induction and a second 6 h later; miRNA-seq and mRNA-seq were completed for each sample. An aggregate score calculation determined which miRNA regulated the differentially expressed mRNA. Networks of molecular interactions were generated for the combined miRNA/target mRNA. A total of 227 miRNA were identified, and 46 were differentially expressed after ICH (FDR < 0.05). The anti-inflammatory miR-181a was decreased post-ICH, and it was the most highly connected miRNA in the miRNA/mRNA bioinformatic network analysis. miR-181a has interconnected pathophysiology with IL-8 and monocytes; in prior studies, we found that IL-8 and monocytes contributed to post-ICH inflammation and ICH clinical outcome, respectively. miR-181a was a significant mediator of post-ICH inflammation and is promising for further study, including as a potential therapeutic target. This investigation also demonstrated feasible methodology for miRNA-seq/mRNA-seq analysis in swine that is innovative, and with unique challenges, compared with transcriptomics research in more established species.
Collapse
Affiliation(s)
- Kyle B Walsh
- University of Cincinnati Gardner Neuroscience Institute, Cincinnati, OH, USA.
- Department of Emergency Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Kip D Zimmerman
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Xiang Zhang
- Department of Environmental Health, University of Cincinnati, Cincinnati, OH, USA
| | - Stacie L Demel
- University of Cincinnati Gardner Neuroscience Institute, Cincinnati, OH, USA
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Yu Luo
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, OH, USA
| | - Carl D Langefeld
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Eric Wohleb
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- University of Cincinnati Neurobiology Research Center, Cincinnati, OH, USA
| | - Grant Schulert
- Division of Pediatric Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Daniel Woo
- University of Cincinnati Gardner Neuroscience Institute, Cincinnati, OH, USA
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Opeolu Adeoye
- Department of Emergency Medicine, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
100
|
Arora L, Pal D. Remodeling of Stromal Cells and Immune Landscape in Microenvironment During Tumor Progression. Front Oncol 2021; 11:596798. [PMID: 33763348 PMCID: PMC7982455 DOI: 10.3389/fonc.2021.596798] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
The molecular understanding of carcinogenesis and tumor progression rests in intra and inter-tumoral heterogeneity. Solid tumors confined with vast diversity of genetic abnormalities, epigenetic modifications, and environmental cues that differ at each stage from tumor initiation, progression, and metastasis. Complexity within tumors studied by conventional molecular techniques fails to identify different subclasses in stromal and immune cells in individuals and that affects immunotherapies. Here we focus on diversity of stromal cell population and immune inhabitants, whose subtypes create the complexity of tumor microenvironment (TME), leading primary tumors towards advanced-stage cancers. Recent advances in single-cell sequencing (epitope profiling) approach circumscribes phenotypic markers, molecular pathways, and evolutionary trajectories of an individual cell. We discussed the current knowledge of stromal and immune cell subclasses at different stages of cancer development with the regulatory role of non-coding RNAs. Finally, we reported the current therapeutic options in immunotherapies, advances in therapies targeting heterogeneity, and possible outcomes.
Collapse
Affiliation(s)
- Leena Arora
- Tissue Engineering and Regenerative Medicine Lab, Indian Institute of Technology Ropar, Rupnagar, India
| | - Durba Pal
- Tissue Engineering and Regenerative Medicine Lab, Indian Institute of Technology Ropar, Rupnagar, India
| |
Collapse
|