1
|
Xu H, Li X, Liu K, Huang P, Liu XJ. PM2.5 Promotes Macrophage-Mediated Inflammatory Response Through Airway Epithelial Cell-Derived Exosomal miR-155-5p. J Inflamm Res 2024; 17:8555-8567. [PMID: 39539727 PMCID: PMC11559224 DOI: 10.2147/jir.s482509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Background Airway epithelial cells (AECs) and alveolar macrophages are involved in airway inflammation. The direct effects of atmospheric fine-particulate-matter (PM2.5) on airway cells, such as AECs and alveolar macrophages, have been widely investigated, but the effect of cell-cell interaction on inflammatory response remains unclear. Exosomes play a crucial role in intercellular communication. However, the cellular interaction of exosomes in PM2.5-induced airway inflammation is unclear. Methods The PM2.5-induced human bronchial epithelial (BEAS-2B) cells and phorbol 12-myristate 13-acetate-induced macrophages (Mφ) were co-cultured and then the expression of IL-6, IL-1β, TNF-α and miRNA-155-5p were detected. Exosomes from PM2.5-exposed BEAS-2B cells were then co-cultured with Mφ to detect the expression of miR-155-5p and inflammatory cytokines, as well as cytokine signaling inhibitor-1 (SOCS1)/NFκB, and to detect the effect of the exosome inhibitor GW4869. Results After the co-culture of PM2.5-induced BEAS-2B cells and Mφ, the expression of Mφ-derived IL-6, IL-1β, and TNF-α, as well as miRNA-155-5p were upregulated. The expression of miRNA-155-5p was upregulated in BEAS-2B and BEAS-2B cell-derived exosomes after exposure to PM2.5. Furthermore, co-culturing exosomes derived from PM2.5-exposed BEAS-2B cells with Mφ, upregulated miR-155-5p and inflammatory cytokines, decreased cytokine signaling inhibitor-1 (SOCS1) expression, and activated NF-κB. In addition, adding exosome inhibitor GW4869 to PM2.5-interfered BEAS-2B cells co-culture with Mφ downregulated miRNA-155-5p expression, inhibited NF-κB, and reduced the levels of inflammatory factors. Conclusion PM2.5 promotes Mφ inflammation by upregulating miRNA-155-5P in exosomes obtained from BEAS-2B cells through miR-155-5P/SOCS1/NF-κB pathway. Exosomal miRNAs mediate cellular communication between BEAS-2B cells and Mφ, which may be a new mechanism of PM2.5-stimulated pulmonary inflammatory response.
Collapse
Affiliation(s)
- Hui Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Xin Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Kai Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Ping Huang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| | - Xiao-Ju Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, People’s Republic of China
- The First Hospital of Lanzhou University, Lanzhou, Gansu, People’s Republic of China
| |
Collapse
|
2
|
Qassim HA, Mohammed ST, Muhamed HJ. The impact of miRNA-155 in acute and chronic toxoplasmosis in Iraqi women. Acta Trop 2024; 255:107211. [PMID: 38678844 DOI: 10.1016/j.actatropica.2024.107211] [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: 12/20/2023] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 05/01/2024]
Abstract
Toxoplasmosis is a prevalent parasitic infection caused by Toxoplasma gondii known to induce complex immune responses, to control the infection. MicroRNAs (miRNAs) are a cluster of small noncoding RNAs that are reported to have regulatory functions in the immune response. The objective of this study is to assess the expression of miR-155 and its targets, Src homology-2 domain-containing inositol 5- phosphatase 1 (SHIP-1) and suppressor of cytokine signaling-1 (SOCS1), in non-pregnant Iraqi women seropositive for toxoplasmosis. The study included 55 non-pregnant women positive for toxoplasmosis (20 in the acute phase and 35 in the chronic phase) and 35 non-pregnant women negative for toxoplasmosis (control group). Serum samples were collected from all participants to investigate the expression of miR-155 by RT‒PCR, in addition to the levels of SOCS1 and SHIP-1 measured by ELISA. The results showed a significant increase in the expression of miR-155 in both groups of acute and chronic toxoplasmosis compared to the control group. Lower levels of SOCS1 and SHIP-1 were found in acutely infected women compared to those with chronic infection and non-infected women. These findings showed the possible critical impact of miR-155 on host immune response during T.gondii infection, proposing that miR-155 can be explored as a prospective target to support host immune response against infectious diseases, with special help in early detection and management of toxoplasmosis in high-risk immunocompromised patients. Further studies are needed to evaluate the molecular pathways by which miRNAs improve immunity against toxoplasmosis.
Collapse
Affiliation(s)
- Hiba A Qassim
- Department of Biology, College of Science, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Sabaa T Mohammed
- Department of Biology, College of Science, Al-Mustansiriyah University, Baghdad, Iraq
| | - Haider J Muhamed
- Department of Biology, College of Science, Al-Mustansiriyah University, Baghdad, Iraq
| |
Collapse
|
3
|
Yang Y, Zhang H, Wang Y, Xu J, Shu S, Wang P, Ding S, Huang Y, Zheng L, Yang Y, Xiong C. Promising dawn in the management of pulmonary hypertension: The mystery veil of gut microbiota. IMETA 2024; 3:e159. [PMID: 38882495 PMCID: PMC11170974 DOI: 10.1002/imt2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/15/2023] [Accepted: 11/25/2023] [Indexed: 06/18/2024]
Abstract
The gut microbiota is a complex community of microorganisms inhabiting the intestinal tract, which plays a vital role in human health. It is intricately involved in the metabolism, and it also affects diverse physiological processes. The gut-lung axis is a bidirectional pathway between the gastrointestinal tract and the lungs. Recent research has shown that the gut microbiome plays a crucial role in immune response regulation in the lungs and the development of lung diseases. In this review, we present the interrelated factors concerning gut microbiota and the associated metabolites in pulmonary hypertension (PH), a lethal disease characterized by elevated pulmonary vascular pressure and resistance. Our research team explored the role of gut-microbiota-derived metabolites in cardiovascular diseases and established the correlation between metabolites such as putrescine, succinate, trimethylamine N-oxide (TMAO), and N, N, N-trimethyl-5-aminovaleric acid with the diseases. Furthermore, we found that specific metabolites, such as TMAO and betaine, have significant clinical value in PH, suggesting their potential as biomarkers in disease management. In detailing the interplay between the gut microbiota, their metabolites, and PH, we underscored the potential therapeutic approaches modulating this microbiota. Ultimately, we endeavor to alleviate the substantial socioeconomic burden associated with this disease. This review presents a unique exploratory analysis of the link between gut microbiota and PH, intending to propel further investigations in the gut-lung axis.
Collapse
Affiliation(s)
- Yicheng Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Hanwen Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Yaoyao Wang
- State Key Laboratory of Cardiovascular Disease, Department of Nephrology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
- Department of Genetics University Medical Center Groningen, University of Groningen Groningen The Netherlands
| | - Songren Shu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiac Surgery Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Peizhi Wang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
- Center for Molecular Cardiology University of Zurich Zurich Switzerland
| | - Shusi Ding
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection The Capital Medical University Beijing China
| | - Yuan Huang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiac Surgery Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Lemin Zheng
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection The Capital Medical University Beijing China
- Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, School of Basic Medical Sciences, Health Science Center The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, Peking University Beijing China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Changming Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
4
|
Vaswani CM, Simone J, Pavelick JL, Wu X, Tan GW, Ektesabi AM, Gupta S, Tsoporis JN, Dos Santos CC. Tiny Guides, Big Impact: Focus on the Opportunities and Challenges of miR-Based Treatments for ARDS. Int J Mol Sci 2024; 25:2812. [PMID: 38474059 DOI: 10.3390/ijms25052812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/24/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is characterized by lung inflammation and increased membrane permeability, which represents the leading cause of mortality in ICUs. Mechanical ventilation strategies are at the forefront of supportive approaches for ARDS. Recently, an increasing understanding of RNA biology, function, and regulation, as well as the success of RNA vaccines, has spurred enthusiasm for the emergence of novel RNA-based therapeutics. The most common types of RNA seen in development are silencing (si)RNAs, antisense oligonucleotide therapy (ASO), and messenger (m)RNAs that collectively account for 80% of the RNA therapeutics pipeline. These three RNA platforms are the most mature, with approved products and demonstrated commercial success. Most recently, miRNAs have emerged as pivotal regulators of gene expression. Their dysregulation in various clinical conditions offers insights into ARDS pathogenesis and offers the innovative possibility of using microRNAs as targeted therapy. This review synthesizes the current state of the literature to contextualize the therapeutic potential of miRNA modulation. It considers the potential for miR-based therapeutics as a nuanced approach that incorporates the complexity of ARDS pathophysiology and the multifaceted nature of miRNA interactions.
Collapse
Affiliation(s)
- Chirag M Vaswani
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Julia Simone
- Department of Medicine, McMaster University, Hamilton, ON L8V 5C2, Canada
| | - Jacqueline L Pavelick
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xiao Wu
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Greaton W Tan
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Amin M Ektesabi
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sahil Gupta
- Faculty of Medicine, School of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - James N Tsoporis
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | - Claudia C Dos Santos
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Interdepartmental Division of Critical Care, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| |
Collapse
|
5
|
Al-Khazaleh AK, Zhou X, Bhuyan DJ, Münch GW, Al-Dalabeeh EA, Jaye K, Chang D. The Neurotherapeutic Arsenal in Cannabis sativa: Insights into Anti-Neuroinflammatory and Neuroprotective Activity and Potential Entourage Effects. Molecules 2024; 29:410. [PMID: 38257323 PMCID: PMC10821245 DOI: 10.3390/molecules29020410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Cannabis, renowned for its historical medicinal use, harbours various bioactive compounds-cannabinoids, terpenes, and flavonoids. While major cannabinoids like delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) have received extensive scrutiny for their pharmacological properties, emerging evidence underscores the collaborative interactions among these constituents, suggesting a collective therapeutic potential. This comprehensive review explores the intricate relationships and synergies between cannabinoids, terpenes, and flavonoids in cannabis. Cannabinoids, pivotal in cannabis's bioactivity, exhibit well-documented analgesic, anti-inflammatory, and neuroprotective effects. Terpenes, aromatic compounds imbuing distinct flavours, not only contribute to cannabis's sensory profile but also modulate cannabinoid effects through diverse molecular mechanisms. Flavonoids, another cannabis component, demonstrate anti-inflammatory, antioxidant, and neuroprotective properties, particularly relevant to neuroinflammation. The entourage hypothesis posits that combined cannabinoid, terpene, and flavonoid action yields synergistic or additive effects, surpassing individual compound efficacy. Recognizing the nuanced interactions is crucial for unravelling cannabis's complete therapeutic potential. Tailoring treatments based on the holistic composition of cannabis strains allows optimization of therapeutic outcomes while minimizing potential side effects. This review underscores the imperative to delve into the intricate roles of cannabinoids, terpenes, and flavonoids, offering promising prospects for innovative therapeutic interventions and advocating continued research to unlock cannabis's full therapeutic potential within the realm of natural plant-based medicine.
Collapse
Affiliation(s)
- Ahmad K. Al-Khazaleh
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia; (X.Z.); (D.J.B.); (G.W.M.); (K.J.)
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia; (X.Z.); (D.J.B.); (G.W.M.); (K.J.)
| | - Deep Jyoti Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia; (X.Z.); (D.J.B.); (G.W.M.); (K.J.)
- School of Science, Western Sydney University, Penrith, NSW 2751, Australia
| | - Gerald W. Münch
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia; (X.Z.); (D.J.B.); (G.W.M.); (K.J.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Penrith, NSW 2751, Australia
| | - Elaf Adel Al-Dalabeeh
- Department of Biological Sciences, School of Science, University of Jordan, Amman 11942, Jordan;
| | - Kayla Jaye
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia; (X.Z.); (D.J.B.); (G.W.M.); (K.J.)
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia; (X.Z.); (D.J.B.); (G.W.M.); (K.J.)
| |
Collapse
|
6
|
Prieto I, Kavanagh M, Jimenez-Castilla L, Pardines M, Lazaro I, Herrero del Real I, Flores-Muñoz M, Egido J, Lopez-Franco O, Gomez-Guerrero C. A mutual regulatory loop between miR-155 and SOCS1 influences renal inflammation and diabetic kidney disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102041. [PMID: 37842165 PMCID: PMC10571033 DOI: 10.1016/j.omtn.2023.102041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/23/2023] [Indexed: 10/17/2023]
Abstract
Diabetic kidney disease (DKD) is a common microvascular complication of diabetes, a global health issue. Hyperglycemia, in concert with cytokines, activates the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway to induce inflammation and oxidative stress contributing to renal damage. There is evidence of microRNA-155 (miR-155) involvement in diabetes complications, but the underlying mechanisms are unclear. In this study, gain- and loss-of-function experiments were conducted to investigate the interplay between miR-155-5p and suppressor of cytokine signaling 1 (SOCS1) in the regulation of the JAK/STAT pathway during renal inflammation and DKD. In experimental models of mesangial injury and diabetes, miR-155-5p expression correlated inversely with SOCS1 and positively with albuminuria and expression levels of cytokines and prooxidant genes. In renal cells, miR-155-5p mimic downregulated SOCS1 and promoted STAT1/3 activation, cytokine expression, and cell proliferation and migration. Conversely, both miR-155-5p antagonism and SOCS1 overexpression protected cells from inflammation and hyperglycemia damage. In vivo, SOCS1 gene delivery decreased miR-155-5p and kidney injury in diabetic mice. Moreover, therapeutic inhibition of miR-155-5p suppressed STAT1/3 activation and alleviated albuminuria, mesangial damage, and renal expression of inflammatory and fibrotic genes. In conclusion, modulation of the miR-155/SOCS1 axis protects kidneys against diabetic damage, thus highlighting its potential as therapeutic target for DKD.
Collapse
Affiliation(s)
- Ignacio Prieto
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigaciones Sanitarias-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid (UAM), 28040 Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - María Kavanagh
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigaciones Sanitarias-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid (UAM), 28040 Madrid, Spain
| | - Luna Jimenez-Castilla
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigaciones Sanitarias-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid (UAM), 28040 Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Marisa Pardines
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigaciones Sanitarias-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid (UAM), 28040 Madrid, Spain
| | - Iolanda Lazaro
- Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute-IMIM, 08003 Barcelona, Spain
| | - Isabel Herrero del Real
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigaciones Sanitarias-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid (UAM), 28040 Madrid, Spain
| | - Monica Flores-Muñoz
- Translational Medicine Lab, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa 91140, Veracruz, Mexico
| | - Jesus Egido
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigaciones Sanitarias-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid (UAM), 28040 Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| | - Oscar Lopez-Franco
- Translational Medicine Lab, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa 91140, Veracruz, Mexico
| | - Carmen Gomez-Guerrero
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigaciones Sanitarias-Fundacion Jimenez Diaz (IIS-FJD), Universidad Autonoma de Madrid (UAM), 28040 Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28029 Madrid, Spain
| |
Collapse
|
7
|
Zouei N, Dalimi A, Pirestani M, Ghaffarifar F. Assessment of tissue levels of miR-146a and proinflammatory cytokines in experimental cerebral toxoplasmosis following atovaquone and clindamycin treatment: An in vivo study. Microb Pathog 2023; 184:106340. [PMID: 37683834 DOI: 10.1016/j.micpath.2023.106340] [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: 06/12/2023] [Revised: 07/27/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
BACKGROUND Despite recent advances for treating cerebral toxoplasmosis (CT), monitoring the parasite burden and treatment response is still challenging. miRNAs are small non-coding RNAs with regulatory functions that can be used in diagnosis and treatment monitoring. We investigated the changes in miR-146a, BAG-1 gene, IL-6, and IL-10 tissue levels in the brain of BALB/c mice with chronic CT caused by the PRU strain of T. gondii following anti-parasitic and antibiotic treatment. METHOD Fifty-three 6-to 8-week-old BALB/c mice were infected using intraperitoneal inoculation of cerebral cysts of T. gondii PRU strain and then divided into five groups as follows: group 1 included mice treated with 100 mg/kg/d Atovaquone (AT), group 2 included mice treated with 400 mg/kg/d clindamycin (CL), group 3 included mice treated with combination therapy (AT + CL), group 4 included infected untreated mice as a positive control (PC), and; group 5 included uninfected untreated mice as negative control (NC). After the completion of the treatment course, tissue level of mir-146a, miR-155, BAG-1 gene, IL-6, and IL-10 was investigated with real-time polymerase chain reaction. The IL-6/IL-10 ratio was calculated as an indicator of immune response. Moreover, brain cyst numbers were counted on autopsy samples. RESULTS miR-146a, IL-6, IL-10, and BAG-1 genes were expressed in PC, but not in the NC group; miR-146a, IL-6, IL-10, and BAG-1 gene expression were significantly lower in AT, CL, and AT + CL compared with PC. MiR-146a and BAG-1 levels in AT and CL were not different statistically, however, they both had lower levels compared to AT + CL (P < 0.01). There was no difference in the expression of IL-6 and IL-10 between treatment groups. BAG-1 expression was significantly lower in AT, than in CL and AT + CL (P < 0.0089 and < 0.002, respectively). The PC group showed a higher ratio of IL-6/IL-10, although this increase was not statistically significant. It is noteworthy that the treatment with AT reduced this ratio; in the inter-group comparison, this ratio showed a decrease in the AT and AT + CL compared to the PC. The number of brain tissue cysts was significantly lower in AT, CL, and AT + CL, than in PC (p < 0.0001). AT had significantly lower brain cysts than CL and AT + CL (P < 0.0001). CONCLUSION It seems that the factors studied in the current research (microRNA and cytokines) are a suitable index for evaluating the response to antiparasitic and antibiotic treatment. However, more studies should be conducted in the future to confirm our findings.
Collapse
Affiliation(s)
- Nima Zouei
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolhossein Dalimi
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Majid Pirestani
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Ghaffarifar
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
8
|
Alghetaa H, Mohammed A, Singh N, Wilson K, Cai G, Putluri N, Nagarkatti M, Nagarkatti P. Resveratrol attenuates staphylococcal enterotoxin B-activated immune cell metabolism via upregulation of miR-100 and suppression of mTOR signaling pathway. Front Pharmacol 2023; 14:1106733. [PMID: 36909201 PMCID: PMC9999031 DOI: 10.3389/fphar.2023.1106733] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/16/2023] [Indexed: 03/14/2023] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is triggered by a variety of insults, such as bacterial and viral infections, including SARS-CoV-2, leading to high mortality. In the murine model of ARDS induced by Staphylococcal enterotoxin-B (SEB), our previous studies showed that while SEB triggered 100% mortality, treatment with Resveratrol (RES) completely prevented such mortality by attenuating inflammation in the lungs. In the current study, we investigated the metabolic profile of SEB-activated immune cells in the lungs following treatment with RES. RES-treated mice had higher expression of miR-100 in the lung mononuclear cells (MNCs), which targeted mTOR, leading to its decreased expression. Also, Single-cell RNA-seq (scRNA seq) unveiled the decreased expression of mTOR in a variety of immune cells in the lungs. There was also an increase in glycolytic and mitochondrial respiration in the cells from SEB + VEH group in comparison with SEB + RES group. Together these data suggested that RES alters the metabolic reprogramming of SEB-activated immune cells, through suppression of mTOR activation and its down- and upstream effects on energy metabolism. Also, miR-100 could serve as novel potential therapeutic molecule in the amelioration of ARDS.
Collapse
Affiliation(s)
- Hasan Alghetaa
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Baghdad, Baghdad, Iraq
| | - Amira Mohammed
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Baghdad, Baghdad, Iraq
| | - Narendra Singh
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Kiesha Wilson
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Goushuai Cai
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| | - Nagireddy Putluri
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
9
|
Allegra A, Murdaca G, Gammeri L, Ettari R, Gangemi S. Alarmins and MicroRNAs, a New Axis in the Genesis of Respiratory Diseases: Possible Therapeutic Implications. Int J Mol Sci 2023; 24:ijms24021783. [PMID: 36675299 PMCID: PMC9861898 DOI: 10.3390/ijms24021783] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 01/18/2023] Open
Abstract
It is well ascertained that airway inflammation has a key role in the genesis of numerous respiratory pathologies, including asthma, chronic obstructive pulmonary disease, and acute respiratory distress syndrome. Pulmonary tissue inflammation and anti-inflammatory responses implicate an intricate relationship between local and infiltrating immune cells and structural pulmonary cells. Alarmins are endogenic proteins discharged after cell injury in the extracellular microenvironment. The purpose of our review is to highlight the alterations in respiratory diseases involving some alarmins, such as high mobility group box 1 (HMGB1) and interleukin (IL)-33, and their inter-relationships and relationships with genetic non-coding material, such as microRNAs. The role played by these alarmins in some pathophysiological processes confirms the existence of an axis composed of HMGB1 and IL-33. These alarmins have been implicated in ferroptosis, the onset of type 2 inflammation and airway alterations. Moreover, both factors can act on non-coding genetic material capable of modifying respiratory function. Finally, we present an outline of alarmins and RNA-based therapeutics that have been proposed to treat respiratory pathologies.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence:
| | - Luca Gammeri
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
10
|
Sun Y, Hu B, Stanley G, Harris ZM, Gautam S, Homer R, Koff JL, Rajagopalan G. IFN- γ Is Protective in Cytokine Release Syndrome-associated Extrapulmonary Acute Lung Injury. Am J Respir Cell Mol Biol 2023; 68:75-89. [PMID: 36125351 PMCID: PMC9817908 DOI: 10.1165/rcmb.2022-0117oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/19/2022] [Indexed: 02/05/2023] Open
Abstract
The mechanisms by which excessive systemic activation of adaptive T lymphocytes, as in cytokine release syndrome (CRS), leads to innate immune cell-mediated acute lung injury (ALI) or acute respiratory distress syndrome, often in the absence of any infection, remains unknown. Here, we investigated the roles of IFN-γ and IL-17A, key T-cell cytokines significantly elevated in patients with CRS, in the immunopathogenesis of CRS-induced extrapulmonary ALI. CRS was induced in wild-type (WT), IL-17A- and IFN-γ knockout (KO) human leukocyte antigen-DR3 transgenic mice with 10 μg of the superantigen, staphylococcal enterotoxin B, given intraperitoneally. Several ALI parameters, including gene expression profiling in the lungs, were studied 4, 24, or 48 hours later. Systemic T-cell activation with staphylococcal enterotoxin B resulted in robust upregulation of several chemokines, S100A8/A9, matrix metalloproteases, and other molecules implicated in tissue damage, granulocyte as well as agranulocyte adhesion, and diapedesis in the lungs as early as 4 hours, which was accompanied by subsequent neutrophil/eosinophil lung infiltration and severe ALI in IFN-γ KO mice. These pathways were significantly underexpressed in IL-17A KO mice, which manifested mildest ALI and intermediate in WT mice. Neutralization of IFN-γ worsened ALI in WT and IL-17A KO mice, whereas neutralizing IL-17A did not mitigate lung injury in IFN-γ KO mice, suggesting a dominant protective role for IFN-γ in ALI and that IL-17A is dispensable. Ruxolitinib, a Janus kinase inhibitor, increased ALI severity in WT mice. Thus, our study identified novel mechanisms of ALI in CRS and its differential modulation by IFN-γ and IL-17A.
Collapse
Affiliation(s)
- Ying Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Buqu Hu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Gail Stanley
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Zachary M. Harris
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Samir Gautam
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | - Robert Homer
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; and
- Pathology and Laboratory Medicine Service, Veterans Affairs Connecticut HealthCare System, West Haven, Connecticut
| | - Jonathan L. Koff
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and
| | | |
Collapse
|
11
|
Mesenchymal Stem Cell-Secreted TGF-β1 Restores Treg/Th17 Skewing Induced by Lipopolysaccharide and Hypoxia Challenge via miR-155 Suppression. Stem Cells Int 2022; 2022:5522828. [PMID: 35313652 PMCID: PMC8934213 DOI: 10.1155/2022/5522828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 12/20/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
Background. Regulatory T cell (Treg)/T helper (Th) 17 skewing is important in the development of acute respiratory distress syndrome (ARDS). Immunomodulatory effects of mesenchymal stem cell- (MSC-) secreted transforming growth factor- (TGF-) β1 on CD4+ T cells are environment-sensitive and lack discussion in hypoxic and inflammatory conditions. Methods. Mouse splenic CD4+ T cells were precoated with anti-CD3 (5 μg/ml) and anti-CD28 (2 μg/ml) overnight. RAW264.7 cells were added as antigen-presenting cells (APCs). T cells with and without RAW264.7 cells were treated with various LPS concentrations of 0, 10, 100, and 1000 ng/ml or/and at hypoxia condition of 5% O2. Based on LPS (100 ng/ml) and hypoxia conditions (5% O2) as stimuli, MSCs were set as direct coculture or indirect coculture by transwell system. Anti-TGF-β1 neutralization antibody was added to explore the role of TGF-β1 among the soluble factors secreted by MSCs; miR-155 overexpression of CD4+ T cells was performed by transfection, and then, cells were added to the MSC-CD4+ T cell coculture system in hypoxic- and LPS-stimulated condition. After 48 hours, cells or supernatants were collected for detection of frequency of Treg and Th17 subsets, CD4+ T cell apoptosis and proliferation capacity assay by flow cytometry, secretion of INF-γ, IL-17A, IL-21, TGF-β1, and IL-10 by ELISA, and levels of miR-155, Rorc, Foxp3, and Ptpn2 mRNA expression of CD4+ T cells by RT-PCR. Results. MSCs could restore skewed Treg/Th17 induced by LPS and hypoxia compared to groups without MSCs with increased secretion of TGF-β1, IL-10, and IL-17A (
) and attenuate the increased expression of miR-155 in CD4+ T cells via cell-to-cell contact mechanism while TGF-β1 neutralization significantly inhibited the effects of MSCs restoring skewed Treg/Th17 and abolished its effect on miR-155 expression in CD4+ T cells. Conclusions. These findings suggested miR-155 suppression of CD4+ T cells mediated MSC-secreted TGF-β1 modulating skewed Treg/Th17 induced by LPS-hypoxia challenge, providing evidence when proposing future T lymphocyte-targeted cell therapy in a specific condition.
Collapse
|
12
|
Alharris E, Mohammed A, Alghetaa H, Zhou J, Nagarkatti M, Nagarkatti P. The Ability of Resveratrol to Attenuate Ovalbumin-Mediated Allergic Asthma Is Associated With Changes in Microbiota Involving the Gut-Lung Axis, Enhanced Barrier Function and Decreased Inflammation in the Lungs. Front Immunol 2022; 13:805770. [PMID: 35265071 PMCID: PMC8898895 DOI: 10.3389/fimmu.2022.805770] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/26/2022] [Indexed: 12/18/2022] Open
Abstract
Asthma is a chronic respiratory disease highly prevalent worldwide. Recent studies have suggested a role for microbiome-associated gut-lung axis in asthma development. In the current study, we investigated if Resveratrol (RES), a plant-based polyphenol, can attenuate ovalbumin (OVA)-induced murine allergic asthma, and if so, the role of microbiome in the gut-lung axis in this process. We found that RES attenuated allergic asthma with significant improvements in pulmonary functions in OVA-exposed mice when tested using plethysmography for frequency (F), mean volume (MV), specific airway resistance (sRaw), and delay time(dT). RES treatment also suppressed inflammatory cytokines in the lungs. RES modulated lung microbiota and caused an abundance of Akkermansia muciniphila accompanied by a reduction of LPS biosynthesis in OVA-treated mice. Furthermore, RES also altered gut microbiota and induced enrichment of Bacteroides acidifaciens significantly in the colon accompanied by an increase in butyric acid concentration in the colonic contents from OVA-treated mice. Additionally, RES caused significant increases in tight junction proteins and decreased mucin (Muc5ac) in the pulmonary epithelium of OVA-treated mice. Our results demonstrated that RES may attenuate asthma by inducing beneficial microbiota in the gut-lung axis and through the promotion of normal barrier functions of the lung.
Collapse
Affiliation(s)
| | | | | | | | | | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
13
|
El-Sayad M, Abdel Rahman M, Hussein N, Abdel Aziz R, El-Taweel HA, Abd El-Latif N. microRNA-155 Expression and Butyrylcholinesterase Activity in the Liver Tissue of Mice Infected with Toxoplasma gondii (Avirulent and Virulent Strains). Acta Parasitol 2021; 66:1167-1176. [PMID: 33840057 DOI: 10.1007/s11686-021-00383-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/23/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Toxoplasma gondii is an apicomplexan parasite that exhibits distinct strain-related virulence patterns in mice. It can induce hepatic inflammation. The present study investigated MicroRNA-155 (miRNA-155) expression and butyrylcholinesterase (BChE) activity in the liver tissue of mice infected with virulent and avirulent strains of T. gondii. METHODS Mice groups included: Group (A), uninfected controls; Group (B), infected with T. gondii avirulent strain (ME-49) and euthanized 7, 27, 47, or 67 days post-infection (pi); Group (C), infected by T. gondii virulent strain (RH) and euthanized 7 days pi; and Group (D), infected by T. gondii virulent strain (RH), treated 24 h pi with sulfamethoxazole-trimethoprim (150 mg/Kg/day and 30 mg/Kg/day, respectively) and euthanized 5, 10, or 20 days pi. miRNA-155 expression was estimated in the liver tissue using the reverse transcription real-time polymerase chain reaction and the ΔΔCt method. BChE activity was estimated in liver homogenates by Ellman's colorimetric method. Liver sections were examined histopathologically. RESULTS revealed a significant elevation in miRNA-155 expression and a significant reduction of BChE activity in all the infected untreated groups compared to the uninfected mice. In group B, the maximum upregulation of miRNA-155 expression and the least reduction in BChE activity were detected 7 days pi. In group D, complete restoration of normal levels occurred 20 days pi. Liver sections showed distinct histopathological patterns with detection of intracellular tachyzoites in group B. CONCLUSION miRNA-155 and BChE play a role in regulating host-parasite interaction in toxoplasmosis and may contribute to the pathogenesis of T. gondii induced hepatic damage.
Collapse
Affiliation(s)
- Mona El-Sayad
- Department of Parasitology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | - Neveen Hussein
- Department of Applied Medical Chemistry. Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Rawda Abdel Aziz
- Department of Applied Medical Chemistry. Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Hend A El-Taweel
- Department of Parasitology, Medical Research Institute, Alexandria University, Alexandria, Egypt.
| | - Naglaa Abd El-Latif
- Department of Parasitology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
14
|
Rodríguez Mesa XM, Moreno Vergara AF, Contreras Bolaños LA, Guevara Moriones N, Mejía Piñeros AL, Santander González SP. Therapeutic Prospects of Cannabinoids in the Immunomodulation of Prevalent Autoimmune Diseases. Cannabis Cannabinoid Res 2021; 6:196-210. [PMID: 34030476 PMCID: PMC8266560 DOI: 10.1089/can.2020.0183] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Introduction: Cannabinoids such as ▵-9-THC and CBD can downregulate the immune response by modulating the endocannabinoid system. This modulation is relevant for the treatment of prevalent autoimmune diseases (ADs), such as multiple sclerosis (MS), systemic lupus erythematosus (SLE), diabetes mellitus type 1 (DMT1), and rheumatoid arthritis (RA). These conditions require new therapeutic options with fewer side effects for the control of the autoimmune response. Objective: to conduct a literature review of preclinical scientific evidence that supports further clinical investigations for the use of cannabinoids (natural or synthetic) as potential immunomodulators of the immune response in ADs. Methodology: A systematic search was carried out in different databases using different MeSH terms, such as Cannabis sativa L., cannabinoids, immunomodulation, and ADs. Initially, 677 journal articles were found. After filtering by publication date (from 2000 to 2020 for SLE, DMT1, and RA; and 2010 to 2020 for MS) and removing the duplicate items, 200 articles were selected and analyzed by title and summary associated with the use of cannabinoids as immunomodulatory treatment for those diseases. Results: Evidence of the immunomodulatory effect of cannabinoids in the diseases previously mentioned, but SLE that did not meet the search criteria, was summarized from 24 journal articles. CBD was found to be one of the main modulators of the immune response. This molecule decreased the number of Th1 and Th17 proinflammatory cells and the production of the proinflammatory cytokines, interleukin (IL)-1, IL-12, IL-17, interferon (IFN)-γ, and tumor necrosis factor alpha, in mouse models of MS and DMT1. Additionally, new synthetic cannabinoid-like molecules, with agonist or antagonist activity on CB1, CB2, TRPV1, PPAR-α, and PPAR-γ receptors, have shown anti-inflammatory properties in MS, DMT1, and RA. Conclusion: Data from experimental animal models of AD showed that natural and synthetic cannabinoids downregulate inflammatory responses mediated by immune cells responsible for AD chronicity and progression. Although synthetic cannabinoid-like molecules were evaluated in just two clinical trials, they corroborated the potential use of cannabinoids to treat some ADs. Notwithstanding, new cannabinoid-based approaches are required to provide alternative treatments to patients affected by the large group of ADs.
Collapse
Affiliation(s)
- Xandy Melissa Rodríguez Mesa
- Phytoimmunomodulation Research Group, Juan N. Corpas University Foundation, Bogotá, Colombia
- Group of Plant Pharmacology and Alternative Therapeutics, Juan N. Corpas University Foundation, Bogotá, Colombia
| | - Andrés Felipe Moreno Vergara
- Phytoimmunomodulation Research Group, Juan N. Corpas University Foundation, Bogotá, Colombia
- Faculty of Medicine, Juan N. Corpas University Foundation, Bogotá, Colombia
| | - Leonardo Andrés Contreras Bolaños
- Phytoimmunomodulation Research Group, Juan N. Corpas University Foundation, Bogotá, Colombia
- Group of Plant Pharmacology and Alternative Therapeutics, Juan N. Corpas University Foundation, Bogotá, Colombia
| | - Natalia Guevara Moriones
- Phytoimmunomodulation Research Group, Juan N. Corpas University Foundation, Bogotá, Colombia
- Faculty of Medicine, Juan N. Corpas University Foundation, Bogotá, Colombia
| | - Antonio Luis Mejía Piñeros
- Group of Plant Pharmacology and Alternative Therapeutics, Juan N. Corpas University Foundation, Bogotá, Colombia
| | - Sandra Paola Santander González
- Phytoimmunomodulation Research Group, Juan N. Corpas University Foundation, Bogotá, Colombia
- Group of Plant Pharmacology and Alternative Therapeutics, Juan N. Corpas University Foundation, Bogotá, Colombia
| |
Collapse
|
15
|
Wang ZF, Yang YM, Fan H. Diagnostic value of miR-155 for acute lung injury/acute respiratory distress syndrome in patients with sepsis. J Int Med Res 2021; 48:300060520943070. [PMID: 32720541 PMCID: PMC7388133 DOI: 10.1177/0300060520943070] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective We aimed to investigate the diagnostic value of microRNA-155 (miR-155) for
acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) in
patients with sepsis. Methods In this prospective study, we used Spearman correlation analysis to
investigate relationships between miR-155 expression and inflammatory
factors, oxygenation ratio (PaO2/FiO2), and ALI/ARDS
score, and used area under the receiver operating characteristic curve
(AU-ROC) to evaluate miR-155's diagnostic accuracy for ALI/ARDS in patients
with sepsis. Results In total, 156 patients with sepsis were enrolled in our study, of which 41
had ALI and 32 had ARDS. miR-155 expression in plasma of patients with
sepsis and ALI/ARDS was significantly higher than that of patients with
sepsis but no ALI/ARDS. The miR-155 level in patients with sepsis and
ALI/ARDS was positively correlated with interleukin (IL)-1β and tumor
necrosis factor (TNF)-α levels and ALI/ARDS score, but negatively correlated
with PaO2/FiO2. The AU-ROC of plasma miR-155 for
diagnosis of sepsis with ALI/ARDS was 0.87, and plasma miR-155, IL-1β, and
TNF-α had high sensitivity and specificity for the diagnosis of sepsis with
ALI/ARDS. Conclusion miR-155 is highly expressed in plasma of patients with septic ALI/ARDS; it is
positively correlated with lung function and can be used for early
diagnosis.
Collapse
Affiliation(s)
- Zhou-Feng Wang
- Department of Respiratory and Critical Care, Ningbo First Hospital Longshan Hospital Medical and Health Group, Ningbo, Zhejiang Province, P. R. China
| | - Yu-Min Yang
- Department of Intensive Care Unit, Ningbo First Hospital, Ningbo, Zhejiang Province, P. R. China
| | - Heng Fan
- Department of Intensive Care Unit, Ningbo First Hospital, Ningbo, Zhejiang Province, P. R. China
| |
Collapse
|
16
|
Varikuti S, Verma C, Natarajan G, Oghumu S, Satoskar AR. MicroRNA155 Plays a Critical Role in the Pathogenesis of Cutaneous Leishmania major Infection by Promoting a Th2 Response and Attenuating Dendritic Cell Activity. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:809-816. [PMID: 33539779 PMCID: PMC8132173 DOI: 10.1016/j.ajpath.2021.01.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 01/10/2023]
Abstract
Interferon (IFN)-γ is indispensable in the resolution of cutaneous leishmaniasis (CL), while the Th2 cytokines IL-4, IL-10, and IL-13 mediate susceptibility. A recent study found that miR155, which promotes CD4+ Th1 response and IFN-γ production, is dispensable in the control of Leishmania donovani infection. Here, the role of miR155 in CL caused by L. major was investigated using miR155-deficient (miR155-/-) mice. Infection was controlled significantly quicker in the miR155-/- mice than in their wild-type (WT) counterparts, indicating that miR155 contributes to the pathogenesis of CL. Faster resolution of infection in miR155-/- mice was associated with increased levels of Th1-associated IL-12 and IFN-γ and reduced production of Th2- associated IL-4, IL-10, and IL-13. Concentrations of IFN-γ+CD8+ T cells and natural killer cells in draining lymph nodes were significantly higher in the L. major-infected miR155-/- mice than in the infected WT mice, as indicated by flow-cytometry. After in vitro IFN-γ stimulation, nitric oxide and IL-12 production were increased, IL-10 production was decreased, and parasite clearance was enhanced in L. major-infected miR155-/- DCs compared to those in WT DCs. Furthermore, IFN-γ production from activated miR155-/- T cells was significantly enhanced in L. major-infected miR155-/- DCs. Together, these findings demonstrate that miR155 promotes susceptibility to CL caused by L. major by promoting Th2 response and inhibiting DC function.
Collapse
Affiliation(s)
- Sanjay Varikuti
- Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio
| | - Chaitenya Verma
- Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio
| | - Gayathri Natarajan
- Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University Medical Center, Columbus, Ohio; Department of Microbiology, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
17
|
Killy B, Bodendorfer B, Mages J, Ritter K, Schreiber J, Hölscher C, Pracht K, Ekici A, Jäck HM, Lang R. DGCR8 deficiency impairs macrophage growth and unleashes the interferon response to mycobacteria. Life Sci Alliance 2021; 4:4/6/e202000810. [PMID: 33771876 PMCID: PMC8008949 DOI: 10.26508/lsa.202000810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 11/24/2022] Open
Abstract
The mycobacterial cell wall glycolipid trehalose-6,6-dimycolate (TDM) activates macrophages through the C-type lectin receptor MINCLE. Regulation of innate immune cells relies on miRNAs, which may be exploited by mycobacteria to survive and replicate in macrophages. Here, we have used macrophages deficient in the microprocessor component DGCR8 to investigate the impact of miRNA on the response to TDM. Deletion of DGCR8 in bone marrow progenitors reduced macrophage yield, but did not block macrophage differentiation. DGCR8-deficient macrophages showed reduced constitutive and TDM-inducible miRNA expression. RNAseq analysis revealed that they accumulated primary miRNA transcripts and displayed a modest type I IFN signature at baseline. Stimulation with TDM in the absence of DGCR8 induced overshooting expression of IFNβ and IFN-induced genes, which was blocked by antibodies to type I IFN. In contrast, signaling and transcriptional responses to recombinant IFNβ were unaltered. Infection with live Mycobacterium bovis Bacille Calmette-Guerin replicated the enhanced IFN response. Together, our results reveal an essential role for DGCR8 in curbing IFNβ expression macrophage reprogramming by mycobacteria.
Collapse
Affiliation(s)
- Barbara Killy
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Bodendorfer
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Kristina Ritter
- Infection Immunology, Forschungszentrum Borstel, Borstel, Germany
| | - Jonathan Schreiber
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Hölscher
- Infection Immunology, Forschungszentrum Borstel, Borstel, Germany.,German Center for Infection Research (DZIF), Partner Site Borstel, Borstel, Germany
| | - Katharina Pracht
- Division of Molecular Immunology, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arif Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
18
|
He B, Zhou W, Rui Y, Liu L, Chen B, Su X. MicroRNA-574-5p Attenuates Acute Respiratory Distress Syndrome by Targeting HMGB1. Am J Respir Cell Mol Biol 2021; 64:196-207. [PMID: 33202146 PMCID: PMC7874400 DOI: 10.1165/rcmb.2020-0112oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a critical condition with high mortality. HMGB1 (high-mobility group protein B1) is one of the key proinflammatory factors in the ARDS “inflammatory storm.” According to previous studies, some microRNAs (miRNAs) play important roles in this process. We aimed to determine the contributing miRNAs targeting the expression and release of HMGB1. miRNA expression in the peripheral blood of patients with ARDS was measured by miRNA microarray. miRNAs targeting HMGB1 were screened and explored for further study. In LPS-induced cell and mouse ARDS models, we explored the effect of this miRNA on the expression and secretion of HMGB1 by Western blot, real-time qPCR, and ELISA. The effects of this miRNA on the NF-κB signaling pathway, proinflammatory cytokines, and NLRP3 (nod-like receptor protein 3) inflammasome were detected by Western blot and real-time qPCR. In ARDS models, microRNA-574-5p (miR-574-5p) expression could be induced by the TLR4/NF-κB pathway upon LPS stimulation. It could suppress the inflammatory response by targeting HMGB1. Enforcing the expression of miR-574-5p or HMGB1 siRNA silencing inhibits the activation of NF-κB signaling pathway and the NLRP3 inflammasome. Moreover, overexpression of HMGB1 reversed the antiinflammatory effect of miR-574-5p. In ARDS mice, overexpression of miR-574-5p suppresses alveolar leukocytes infiltration, interstitial edema, protein effusion, and inflammation. This study demonstrated that miR-574-5p provided negative feedback to LPS-induced inflammation and relieved ARDS. It may provide new therapeutic strategies for ARDS.
Collapse
Affiliation(s)
- Binchan He
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.,The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; and
| | - Wei Zhou
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuwen Rui
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lulu Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bilin Chen
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xin Su
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Respiratory and Critical Care Medicine, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Riahi Rad Z, Riahi Rad Z, Goudarzi H, Goudarzi M, Mahmoudi M, Yasbolaghi Sharahi J, Hashemi A. MicroRNAs in the interaction between host-bacterial pathogens: A new perspective. J Cell Physiol 2021; 236:6249-6270. [PMID: 33599300 DOI: 10.1002/jcp.30333] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022]
Abstract
Gene expression regulation plays a critical role in host-pathogen interactions, and RNAs function is essential in this process. miRNAs are small noncoding, endogenous RNA fragments that affect stability and/or translation of mRNAs, act as major posttranscriptional regulators of gene expression. miRNA is involved in regulating many biological or pathological processes through targeting specific mRNAs, including development, differentiation, apoptosis, cell cycle, cytoskeleton organization, and autophagy. Deregulated microRNA expression is associated with many types of diseases, including cancers, immune disturbances, and infection. miRNAs are a vital section of the host immune response to bacterial-made infection. Bacterial pathogens suppress host miRNA expression for their benefit, promoting survival, replication, and persistence. The role played through miRNAs in interaction with host-bacterial pathogen has been extensively studied in the past 10 years, and knowledge about these staggering molecules' function can clarify the complicated and ambiguous interactions of the host-bacterial pathogen. Here, we review how pathogens prevent the host miRNA expression. We briefly discuss emerging themes in this field, including their role as biomarkers in identifying bacterial infections, as part of the gut microbiota, on host miRNA expression.
Collapse
Affiliation(s)
- Zohreh Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Yasbolaghi Sharahi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Liu J, Sala MA, Kim J. Dampening the Fire: A Negative Feedback Loop in Acute Respiratory Distress Syndrome. Am J Respir Cell Mol Biol 2021; 64:158-160. [PMID: 33522886 PMCID: PMC7874398 DOI: 10.1165/rcmb.2020-0487ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Jing Liu
- Department of Surgery, College of Medicine, Cancer Center, University of Illinois at Chicago, Chicago, Illinois
| | - Marc A Sala
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, and
| | - Jiyeon Kim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
21
|
Chang Y, Chen X, Tian Y, Gao X, Liu Z, Dong X, Wang L, He F, Zhou J. Downregulation of microRNA-155-5p prevents immune thrombocytopenia by promoting macrophage M2 polarization via the SOCS1-dependent PD1/PDL1 pathway. Life Sci 2020; 257:118057. [PMID: 32634427 DOI: 10.1016/j.lfs.2020.118057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 12/20/2022]
Abstract
AIMS We set about to investigate the potential role of microRNA-155-5p (miR-155-5p) in the development of immune thrombocytopenia (ITP), an idiopathic deficiency of blood platelets. MAIN METHODS Initially, RT-qPCR and Western blot analyses were carried out to determine the expression of miR-155-5p and SOCS1 in peripheral blood mononuclear cells (PBMCs) and macrophages from ITP patients. We undertook gain- and loss- function methods by transfection of macrophages and PBMCs with treated plasmids. The expression patterns of platelet-related factors were measured by ELISA, and the expressions of PD1, PDL1, and macrophage M2 marker CD206 and CD86 were also measured. The relationship between miR-155-5p and SOCS1 was determined using the dual-luciferase reporter gene assay. We also established an ITP mouse model to explore the roles of miR-155-5p and SOCS1 in vivo. KEY FINDINGS miR-155-5p was up-regulated, while SOCS1 was down-regulated in PBMCs and macrophages from ITP patients. SOCS1 was indicated as a target of miR-155-5p. Inhibition of miR-155-5p or up-regulation of SOCS1 facilitated macrophage M2 polarization as demonstrated by an increased M2/M1 ratio and suppressed expression of platelet-related factors. Furthermore, silencing of SOCS1 promoted ITP progression through blocking the PD1/PDL1 pathway, whilst upregulation of miR-155-5p remarkably increased the platelet abundance and suppressed SOCS1 expression in ITP model mice. SIGNIFICANCE Silencing of miR-155-5p could promote PD1/PDL1 pathway-mediated macrophage M2 polarization and prevent ITP via up-regulation of SOCS1, thus relieving ITP.
Collapse
Affiliation(s)
- Yuying Chang
- Department of Hematology, the 2(nd) Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Xi Chen
- Department of Hematology, the 2(nd) Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Yaoyao Tian
- Department of Hematology, the 2(nd) Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Xinyu Gao
- Department of Hematology, the 2(nd) Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Zhiyu Liu
- Flow Laboratory, the First Affiliated Hospital of Harbin Medical University, Harbin 150007, PR China
| | - Xiushuai Dong
- Department of Hematology, the 2(nd) Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Lianjie Wang
- Department of Hematology, the 2(nd) Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Fei He
- Department of Hematology, the 2(nd) Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Jin Zhou
- Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin 150007, PR China.
| |
Collapse
|
22
|
Lee TJ, Yuan X, Kerr K, Yoo JY, Kim DH, Kaur B, Eltzschig HK. Strategies to Modulate MicroRNA Functions for the Treatment of Cancer or Organ Injury. Pharmacol Rev 2020; 72:639-667. [PMID: 32554488 PMCID: PMC7300323 DOI: 10.1124/pr.119.019026] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cancer and organ injury-such as that occurring in the perioperative period, including acute lung injury, myocardial infarction, and acute gut injury-are among the leading causes of death in the United States and impose a significant impact on quality of life. MicroRNAs (miRNAs) have been studied extensively during the last two decades for their role as regulators of gene expression, their translational application as diagnostic markers, and their potential as therapeutic targets for disease treatment. Despite promising preclinical outcomes implicating miRNA targets in disease treatment, only a few miRNAs have reached clinical trials. This likely relates to difficulties in the delivery of miRNA drugs to their targets to achieve efficient inhibition or overexpression. Therefore, understanding how to efficiently deliver miRNAs into diseased tissues and specific cell types in patients is critical. This review summarizes current knowledge on various approaches to deliver therapeutic miRNAs or miRNA inhibitors and highlights current progress in miRNA-based disease therapy that has reached clinical trials. Based on ongoing advances in miRNA delivery, we believe that additional therapeutic approaches to modulate miRNA function will soon enter routine medical treatment of human disease, particularly for cancer or perioperative organ injury. SIGNIFICANCE STATEMENT: MicroRNAs have been studied extensively during the last two decades in cancer and organ injury, including acute lung injury, myocardial infarction, and acute gut injury, for their regulation of gene expression, application as diagnostic markers, and therapeutic potentials. In this review, we specifically emphasize the pros and cons of different delivery approaches to modulate microRNAs, as well as the most recent exciting progress in the field of therapeutic targeting of microRNAs for disease treatment in patients.
Collapse
Affiliation(s)
- Tae Jin Lee
- Departments of Neurosurgery (T.J.L., K.K., J.Y.Y., D.H.K., B.K.) and Anesthesiology (X.Y., H.K.E.), McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Xiaoyi Yuan
- Departments of Neurosurgery (T.J.L., K.K., J.Y.Y., D.H.K., B.K.) and Anesthesiology (X.Y., H.K.E.), McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Keith Kerr
- Departments of Neurosurgery (T.J.L., K.K., J.Y.Y., D.H.K., B.K.) and Anesthesiology (X.Y., H.K.E.), McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Ji Young Yoo
- Departments of Neurosurgery (T.J.L., K.K., J.Y.Y., D.H.K., B.K.) and Anesthesiology (X.Y., H.K.E.), McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Dong H Kim
- Departments of Neurosurgery (T.J.L., K.K., J.Y.Y., D.H.K., B.K.) and Anesthesiology (X.Y., H.K.E.), McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Balveen Kaur
- Departments of Neurosurgery (T.J.L., K.K., J.Y.Y., D.H.K., B.K.) and Anesthesiology (X.Y., H.K.E.), McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Holger K Eltzschig
- Departments of Neurosurgery (T.J.L., K.K., J.Y.Y., D.H.K., B.K.) and Anesthesiology (X.Y., H.K.E.), McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
23
|
Gluud M, Willerslev-Olsen A, Gjerdrum LMR, Lindahl LM, Buus TB, Andersen MH, Bonefeld CM, Krejsgaard T, Litvinov IV, Iversen L, Becker JC, Persson JL, Koralov SB, Litman T, Geisler C, Woetmann A, Odum N. MicroRNAs in the Pathogenesis, Diagnosis, Prognosis and Targeted Treatment of Cutaneous T-Cell Lymphomas. Cancers (Basel) 2020; 12:cancers12051229. [PMID: 32414221 PMCID: PMC7281391 DOI: 10.3390/cancers12051229] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 12/11/2022] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) represents a heterogeneous group of potentially devastating primary skin malignancies. Despite decades of intense research efforts, the pathogenesis is still not fully understood. In the early stages, both clinical and histopathological diagnosis is often difficult due to the ability of CTCL to masquerade as benign skin inflammatory dermatoses. Due to a lack of reliable biomarkers, it is also difficult to predict which patients will respond to therapy or progress towards severe recalcitrant disease. In this review, we discuss recent discoveries concerning dysregulated microRNA (miR) expression and putative pathological roles of oncogenic and tumor suppressive miRs in CTCL. We also focus on the interplay between miRs, histone deacetylase inhibitors, and oncogenic signaling pathways in malignant T cells as well as the impact of miRs in shaping the inflammatory tumor microenvironment. We highlight the potential use of miRs as diagnostic and prognostic markers, as well as their potential as therapeutic targets. Finally, we propose that the combined use of miR-modulating compounds with epigenetic drugs may provide a novel avenue for boosting the clinical efficacy of existing anti-cancer therapies in CTCL.
Collapse
Affiliation(s)
- Maria Gluud
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.G.); (A.W.-O.); (T.B.B.); (C.M.B.); (T.K.); (T.L.); (C.G.); (A.W.)
| | - Andreas Willerslev-Olsen
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.G.); (A.W.-O.); (T.B.B.); (C.M.B.); (T.K.); (T.L.); (C.G.); (A.W.)
| | - Lise Mette Rahbek Gjerdrum
- Department of Pathology, Zealand University Hospital, DK-4000 Roskilde, Denmark;
- Department of Clinical Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Lise M. Lindahl
- Department of Dermatology, Aarhus University Hospital, DK-8200 Aarhus, Denmark; (L.M.L.); (L.I.)
| | - Terkild B. Buus
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.G.); (A.W.-O.); (T.B.B.); (C.M.B.); (T.K.); (T.L.); (C.G.); (A.W.)
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology and Oncology, Copenhagen University Hospital, Herlev Hospital, DK-2730 Herlev, Denmark;
| | - Charlotte Menne Bonefeld
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.G.); (A.W.-O.); (T.B.B.); (C.M.B.); (T.K.); (T.L.); (C.G.); (A.W.)
| | - Thorbjorn Krejsgaard
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.G.); (A.W.-O.); (T.B.B.); (C.M.B.); (T.K.); (T.L.); (C.G.); (A.W.)
| | - Ivan V. Litvinov
- Division of Dermatology, McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, DK-8200 Aarhus, Denmark; (L.M.L.); (L.I.)
| | - Jürgen C. Becker
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Hospital Essen and Deutsches Krebsforschungszentrum (DKFZ), D-45141 Essen, Germany;
| | - Jenny L. Persson
- Department of Molecular Biology, Umea University, 90187 Umea, Sweden;
| | - Sergei B. Koralov
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA;
| | - Thomas Litman
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.G.); (A.W.-O.); (T.B.B.); (C.M.B.); (T.K.); (T.L.); (C.G.); (A.W.)
| | - Carsten Geisler
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.G.); (A.W.-O.); (T.B.B.); (C.M.B.); (T.K.); (T.L.); (C.G.); (A.W.)
| | - Anders Woetmann
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.G.); (A.W.-O.); (T.B.B.); (C.M.B.); (T.K.); (T.L.); (C.G.); (A.W.)
| | - Niels Odum
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark; (M.G.); (A.W.-O.); (T.B.B.); (C.M.B.); (T.K.); (T.L.); (C.G.); (A.W.)
- Correspondence: ; Tel.: +45-2875-7879
| |
Collapse
|
24
|
Chen L, Chen J, Xie G, Zhu L. MiR-222 inhibition alleviates Staphylococcal Enterotoxin B-induced inflammatory acute lung injury by targeting Foxo3. J Biosci 2020. [DOI: 10.1007/s12038-020-00037-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
25
|
MicroRNA-155 Participates in Smoke-Inhalation-Induced Acute Lung Injury through Inhibition of SOCS-1. Molecules 2020; 25:molecules25051022. [PMID: 32106541 PMCID: PMC7179228 DOI: 10.3390/molecules25051022] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/18/2020] [Accepted: 02/23/2020] [Indexed: 12/11/2022] Open
Abstract
Smoke inhalation causes acute lung injury (ALI), a severe clinical disease with high mortality. Accumulating evidence indicates that microRNA-155 (miR-155) and suppressor of cytokine signaling 1 (SOCS-1), as mediators of inflammatory response, are involved in the pathogenesis of ALI. In this paper, we explored the proinflammatory mechanism of miR-155 in smoke-inhalation-induced ALI. Our data revealed that smoke inhalation induces miR-155 expression, and miR-155 knockout (KO) significantly ameliorates smoke-inhalation-induced lung injury in mice. Neutrophil infiltration and myeloperoxidase (MPO), macrophage inflammatory protein 2 (MIP-2) and keratinocyte chemoattractant (KC) expressions were decreased in miR-155–/– mice after smoke inhalation as well. Real-time RT-PCR and immunoblotting results showed that SOCS-1 level was remarkably increased in miR-155–/– mice after smoke exposure. Furthermore, the experiments performed in isolated miR-155 KO pulmonary neutrophils demonstrated that the lack of SOCS-1 enhanced inflammatory cytokines (MIP-2 and KC) secretion in response to smoke stimulation. In conclusion, smoke induces increased expression of miR-155, and miR-155 is involved in inflammatory response to smoke-inhalation-induced lung injury by inhibiting the expression of SOCS-1.
Collapse
|
26
|
Keck J, Chambers JP, Kancharla A, Bashir DH, Henley L, Schenkel K, Castillo K, Neal Guentzel M, Gupta R, Arulanandam BP. The Role of MicroRNA-155 in Chlamydia muridarum Infected lungs. Microbes Infect 2020; 22:360-365. [PMID: 32084556 DOI: 10.1016/j.micinf.2020.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 01/14/2023]
Abstract
Our laboratory has investigated the role of an evolutionarily conserved RNA species called microRNAs (miRs) in regulation of anti-chlamydial protective immunity. MiRs including miR-155 expressed in specific immune effector cells are critical for antigen specific protective immunity and IFN-γ production. Using miR-155 deficient mice, and a murine pulmonary model for chlamydial infection, we report here 1) the effect of host miR-155 on bacterial burden, and 2) identify probable immune genes regulated by miR-155.
Collapse
Affiliation(s)
- Jonathon Keck
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - James P Chambers
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Aravind Kancharla
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Dona Haj Bashir
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Laura Henley
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Katherine Schenkel
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Kevin Castillo
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - M Neal Guentzel
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | - Rishein Gupta
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| | - Bernard P Arulanandam
- South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| |
Collapse
|
27
|
Mai LJ, Fu XX, He G, Zhao EN, Xue M. [Effect of asiaticoside on hyperoxia-induced bronchopulmonary dysplasia in neonatal rats and related mechanism]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:71-76. [PMID: 31948528 PMCID: PMC7389707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/11/2019] [Indexed: 11/04/2023]
Abstract
OBJECTIVE To study the protective effect of asiaticoside against hyperoxia-induced bronchopulmonary dysplasia in neonatal rats based on the microRNA-155 (miR-155)/suppressor of cytokine signaling-1 (SOCS1) axis. METHODS Neonatal rats were randomly divided into a control group, a model group, a low-dose asiaticoside group (10 mg/kg), a middle-dose asiaticoside group (25 mg/kg), a high-dose asiaticoside group (50 mg/kg), and a budesonide group (1.5 mg/kg), with 12 rats in each group. All rats except those in the control group were exposed to a high concentration of oxygen for 14 days to establish a neonatal rat model of bronchopulmonary dysplasia. The low-, middle-, and high-dose asiaticoside groups were given asiaticoside at different doses by gavage, and those in the budesonide group were given budesonide aerosol treatment. Hematoxylin and eosin staining was used to observe lung tissue development and measure radial alveolar count (RAC) and mean linear intercept (MLI). Superoxide dismutase (SOD) and malondialdehyde (MDA) detection kits were used to measure the levels of SOD and MDA in lung tissue. ELISA was used to measure the serum levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Quantitative real-time PCR was used to measure the mRNA expression of miR-155 and SOCS1 in lung tissue. Western blotting was used to measure the protein expression of SOCS1 in lung tissue. RESULTS Compared with the control group, the model group had the symptoms of bronchopulmonary dysplasia such as a disordered structure of lung tissue, enlargement of alveolar fusion, uneven alveolar septa, enlargement of average alveolar space, and a reduction in alveolar number. The model group also had significant increases in MLI, MDA level in lung tissue, serum levels of IL-6 and TNF-α, and miR-155 level in lung tissue (P<0.05) and significant reductions in RAC, SOD level, and mRNA and protein expression of SOCS1 in lung tissue (P<0.05). Compared with the model group, the low-, middle-, and high-dose asiaticoside groups and the budesonide group had significant improvement in the above symptoms of bronchopulmonary dysplasia, significant reductions in MLI, MDA level in lung tissue, serum levels of IL-6 and TNF-α, and miR-155 level in lung tissue (P<0.05), and significant increases in RAC, SOD level, and mRNA and protein expression of SOCS1 in lung tissue (P<0.05). Asiaticoside improved the above symptoms and indices in a dose-dependent manner. There were no significant differences in the above indices between the high-dose asiaticoside and budesonide groups (P>0.05). CONCLUSIONS Asiaticoside can alleviate inflammation injury induced by hyperoxia in neonatal rats and improve the symptoms of bronchopulmonary dysplasia in a dose-dependent manner, possibly by down-regulating the expression of miR-155 and up-regulating the expression of SOCS1.
Collapse
Affiliation(s)
- Lang-Jun Mai
- Department of Pediatrics, Danzhou People's Hospital, Danzhou, Hainan 571700, China.
| | | | | | | | | |
Collapse
|
28
|
Mai LJ, Fu XX, He G, Zhao EN, Xue M. [Effect of asiaticoside on hyperoxia-induced bronchopulmonary dysplasia in neonatal rats and related mechanism]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:71-76. [PMID: 31948528 PMCID: PMC7389707 DOI: 10.7499/j.issn.1008-8830.2020.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/11/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To study the protective effect of asiaticoside against hyperoxia-induced bronchopulmonary dysplasia in neonatal rats based on the microRNA-155 (miR-155)/suppressor of cytokine signaling-1 (SOCS1) axis. METHODS Neonatal rats were randomly divided into a control group, a model group, a low-dose asiaticoside group (10 mg/kg), a middle-dose asiaticoside group (25 mg/kg), a high-dose asiaticoside group (50 mg/kg), and a budesonide group (1.5 mg/kg), with 12 rats in each group. All rats except those in the control group were exposed to a high concentration of oxygen for 14 days to establish a neonatal rat model of bronchopulmonary dysplasia. The low-, middle-, and high-dose asiaticoside groups were given asiaticoside at different doses by gavage, and those in the budesonide group were given budesonide aerosol treatment. Hematoxylin and eosin staining was used to observe lung tissue development and measure radial alveolar count (RAC) and mean linear intercept (MLI). Superoxide dismutase (SOD) and malondialdehyde (MDA) detection kits were used to measure the levels of SOD and MDA in lung tissue. ELISA was used to measure the serum levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). Quantitative real-time PCR was used to measure the mRNA expression of miR-155 and SOCS1 in lung tissue. Western blotting was used to measure the protein expression of SOCS1 in lung tissue. RESULTS Compared with the control group, the model group had the symptoms of bronchopulmonary dysplasia such as a disordered structure of lung tissue, enlargement of alveolar fusion, uneven alveolar septa, enlargement of average alveolar space, and a reduction in alveolar number. The model group also had significant increases in MLI, MDA level in lung tissue, serum levels of IL-6 and TNF-α, and miR-155 level in lung tissue (P<0.05) and significant reductions in RAC, SOD level, and mRNA and protein expression of SOCS1 in lung tissue (P<0.05). Compared with the model group, the low-, middle-, and high-dose asiaticoside groups and the budesonide group had significant improvement in the above symptoms of bronchopulmonary dysplasia, significant reductions in MLI, MDA level in lung tissue, serum levels of IL-6 and TNF-α, and miR-155 level in lung tissue (P<0.05), and significant increases in RAC, SOD level, and mRNA and protein expression of SOCS1 in lung tissue (P<0.05). Asiaticoside improved the above symptoms and indices in a dose-dependent manner. There were no significant differences in the above indices between the high-dose asiaticoside and budesonide groups (P>0.05). CONCLUSIONS Asiaticoside can alleviate inflammation injury induced by hyperoxia in neonatal rats and improve the symptoms of bronchopulmonary dysplasia in a dose-dependent manner, possibly by down-regulating the expression of miR-155 and up-regulating the expression of SOCS1.
Collapse
Affiliation(s)
- Lang-Jun Mai
- Department of Pediatrics, Danzhou People's Hospital, Danzhou, Hainan 571700, China.
| | | | | | | | | |
Collapse
|
29
|
Zhang P, Yu J, Gui Y, Sun C, Han W. Inhibition of miRNA-222-3p Relieves Staphylococcal Enterotoxin B-Induced Liver Inflammatory Injury by Upregulating Suppressors of Cytokine Signaling 1. Yonsei Med J 2019; 60:1093-1102. [PMID: 31637892 PMCID: PMC6813146 DOI: 10.3349/ymj.2019.60.11.1093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/02/2019] [Accepted: 08/21/2019] [Indexed: 01/20/2023] Open
Abstract
PURPOSE Staphylococcal enterotoxin B (SEB) has been well-documented to induce liver injury. miRNA-222-3p (miR-222-3p) was implicated in SEB-induced lung injury and several liver injuries. This study aimed to explore the role of miR-222-3p in SEB-induced liver injury. MATERIALS AND METHODS Expression of miR-222-3p and suppressors of cytokine signaling 1 (SOCS1) was detected using real-time quantitative PCR and western blot. Liver injury was determined by levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and inflammatory cytokines, numbers of infiltrating mononuclear cells using AST/ALT assay kit, enzyme-linked immunosorbent assay (ELISA), and hematoxylin-eosin staining, respectively. Target binding between miR-222-3p and SOCS1 was predicted on targetScan software, and confirmed by luciferase reporter assay. RESULTS SEB induced liver injury in D-galactosamine (D-gal)-sensitized mice, as demonstrated by increased serum levels of AST and ALT, elevated release of interferon-gamma (INF-γ), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and IL-2, and promoted infiltrating immune cells into liver. Expression of miR-222-3p was dramatically upregulated, and SOCS1 was downregulated in SEB-induced liver injury both in mice and splenocytes. Moreover, miR-222-3p knockout (KO) mice exhibited alleviated liver injury accompanied with SOCS1 upregulation. Besides, splenocytes under SEB challenge released less INF-γ, TNF-α, IL-6, and IL-2 during miR-222-3p knockdown. Mechanically, SOCS1 was targeted and downregulated by miR-222-3p. Upregulation of SOCS1 attenuated INF-γ, TNF-α, IL-6, and IL-2 release in SEB-induced splenocytes; downregulation of SOCS1 could block the suppressive role of miR-222-3p knockdown in SEB-induced splenocytes. CONCLUSION Inhibition of miR-222-3p relieves SEB-induced liver inflammatory injury by upregulating SOCS1, thereby providing the first evidence of miR-222-3p in SEB-induced liver injury.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Clinical Laboratory, the Third People's Hospital of Dalian, Dalian, China
| | - Jingda Yu
- Department of Clinical Laboratory, the Baotou Medical College of Inner Mongolia University of Science and Technology, Inner Mongolia, China
| | - Yifang Gui
- Department of Clinical Laboratory, the Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Cui Sun
- Department of Clinical Laboratory, the Third People's Hospital of Dalian, Dalian, China
| | - Weiping Han
- Department of Clinical Laboratory, the Second Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
30
|
Al-Ghezi ZZ, Miranda K, Nagarkatti M, Nagarkatti PS. Combination of Cannabinoids, Δ9- Tetrahydrocannabinol and Cannabidiol, Ameliorates Experimental Multiple Sclerosis by Suppressing Neuroinflammation Through Regulation of miRNA-Mediated Signaling Pathways. Front Immunol 2019; 10:1921. [PMID: 31497013 PMCID: PMC6712515 DOI: 10.3389/fimmu.2019.01921] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/29/2019] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic and disabling disorder of the central nervous system (CNS) characterized by neuroinflammation leading to demyelination. Recently a combination of Δ9-tetrahydrocannabinol (THC) and Cannabidiol (CBD) extracted from Cannabis has been approved in many parts of the world to treat MS-related spasticity. THC+CBD combination was also shown to suppresses neuroinflammation, although the mechanisms remain to be further elucidated. In the current study, we demonstrate that THC+CBD combination therapy (10 mg/kg each) but not THC or CBD alone, attenuates murine experimental autoimmune encephalomyelitis (EAE) by reducing neuroinflammation and suppression of Th17 and Th1 cells. These effects were mediated through CB1 and CB2 receptors inasmuch as, THC+CBD failed to ameliorate EAE in mice deficient in CB1 and CB2. THC+CBD treatment also caused a decrease in the levels of brain infiltrating CD4+ T cells and pro-inflammatory molecules (IL-17, INF-γ, TNF-α, IL-1β, IL-6, and TBX21), while increasing anti-inflammatory phenotype such as FoxP3, STAT5b, IL-4, IL-10, and TGF-β. Also, the brain-derived cells showed increased apoptosis along with decreased percentage in G0/G1 phase with increased percentage in G2/M phase of cell cycle. miRNA microarray analysis of brain-derived CD4+ T cells revealed that THC+CBD treatment significantly down-regulated miR-21a-5p, miR-31-5p, miR-122-5p, miR-146a-5p, miR-150-5p, miR-155-5p, and miR-27b-5p while upregulating miR-706-5p and miR-7116. Pathway analysis showed that majority of the down-regulated miRs targeted molecules involved in cycle arrest and apoptosis such as CDKN2A, BCL2L11, and CCNG1, as well as anti-inflammatory molecules such as SOCS1 and FoxP3. Additionally, transfection studies involving miR-21 and use of Mir21-/- mice suggested that while this miR plays a critical role in EAE, additional miRs may also be involved in THC+CBD-mediated attenuation of EAE. Collectively, this study suggests that combination of THC+CBD suppresses neuroinflammation and attenuates clinical EAE development and that this effect is associated with changes in miRNA profile in brain-infiltrating cells.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/therapeutic use
- Brain/cytology
- Cannabidiol/therapeutic use
- Cells, Cultured
- Cytokines/genetics
- Cytokines/immunology
- Dronabinol/therapeutic use
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/genetics
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Signal Transduction
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Spleen/cytology
Collapse
Affiliation(s)
| | | | | | - Prakash S. Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
31
|
MicroRNA 155 Contributes to Host Immunity against Leishmania donovani but Is Not Essential for Resolution of Infection. Infect Immun 2019; 87:IAI.00307-19. [PMID: 31182615 DOI: 10.1128/iai.00307-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
CD4+ T helper 1 (Th1) cells producing interferon gamma (IFN-γ) are critical for the resolution of visceral leishmaniasis (VL). MicroRNA 155 (miR155) promotes CD4+ Th1 responses and IFN-γ production by targeting suppressor of cytokine signaling-1 (SOCS1) and Src homology-2 domain-containing inositol 5-phosphatase 1 (SHIP-1) and therefore could play a role in the resolution of VL. To determine the role of miR155 in VL, we monitored the course of Leishmania donovani infection in miR155 knockout (miR155KO) and wild-type (WT) C57BL/6 mice. miR155KO mice displayed significantly higher liver and spleen parasite loads than WT controls and showed impaired hepatic granuloma formation. However, parasite growth eventually declined in miR155KO mice, suggesting the induction of a compensatory miR155-independent antileishmanial pathway. Leishmania antigen-stimulated splenocytes from miR155KO mice produced significantly lower levels of Th1-associated IFN-γ than controls. Interestingly, at later time points, levels of Th2-associated interleukin-4 (IL-4) and IL-10 were also lower in miR155KO splenocyte supernatants than in WT mice. On the other hand, miR155KO mice displayed significantly higher levels of IFN-γ, iNOS, and TNF-α gene transcripts in their livers than WT mice, indicating that distinct organ-specific antiparasitic mechanisms were involved in control of L. donovani infection in miR155KO mice. Throughout the course of infection, organs of miR155KO mice showed significantly more PDL1-expressing Ly6Chi inflammatory monocytes than WT mice. Conversely, blockade of Ly6Chi inflammatory monocyte recruitment in miR155KO mice significantly reduced parasitic loads, indicating that these cells contributed to disease susceptibility. In conclusion, we found that miR155 contributes to the control of L. donovani but is not essential for infection resolution.
Collapse
|
32
|
Jiang K, Yang J, Guo S, Zhao G, Wu H, Deng G. Peripheral Circulating Exosome-Mediated Delivery of miR-155 as a Novel Mechanism for Acute Lung Inflammation. Mol Ther 2019; 27:1758-1771. [PMID: 31405809 DOI: 10.1016/j.ymthe.2019.07.003] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 06/29/2019] [Accepted: 07/03/2019] [Indexed: 12/25/2022] Open
Abstract
Emerging evidence has revealed that excessive activation of macrophages may result in an adverse lung inflammation involved in sepsis-related acute lung injury (ALI). However, it has never been clearly identified whether peripheral circulating serum exosomes participate in the pathogenesis of sepsis-related ALI. Therefore, the purposes of our study were to investigate the effect of serum exosomes on macrophage activation and elucidate a novel mechanism underlying sepsis-related ALI. Here we found that exosomes were abundant in the peripheral blood from ALI mice and selectively loaded microRNAs (miRNAs), such as miR-155. In vivo experiments revealed that intravenous injection of serum exosomes harvested from ALI mice, but not control mice, increased the number of M1 macrophages in the lung, and it caused lung inflammation in naive mice. In vitro, we demonstrated that serum exosomes from ALI mice delivered miR-155 to macrophages, stimulated nuclear factor κB (NF-κB) activation, and induced the production of tumor necrosis factor alpha (TNF-α) and interleukin (IL)-6. Furthermore, we also showed that serum exosome-derived miR-155 promoted macrophage proliferation and inflammation by targeting SHIP1 and SOCS1, respectively. Collectively, our data suggest the important role of circulating exosomes secreted into peripheral blood as a key mediator of septic lung injury via exosome-shuttling miR-155.
Collapse
Affiliation(s)
- Kangfeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Jing Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Haichong Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.
| |
Collapse
|
33
|
Wang T, Jiang L, Wei X, Dong Z, Liu B, Zhao J, Wang L, Xie P, Wang Y, Zhou S. Inhibition of miR-221 alleviates LPS-induced acute lung injury via inactivation of SOCS1/NF-κB signaling pathway. Cell Cycle 2019; 18:1893-1907. [PMID: 31208297 DOI: 10.1080/15384101.2019.1632136] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The role of inflammation response has been well documented in the development of acute lung injury (ALI). However, little is known about the functions of miRNAs in the regulation of inflammation in ALI. The aim of this study was to explore the effects of miRNAs in the regulation of inflammation in ALI and to elucidate the biomolecular mechanisms responsible for these effects. The expression profiles of miRNAs in lung tissues from lipopolysaccharide (LPS)-induced ALI mice model were analyzed using a microarray. It was observed that microRNA-221-3p (miR-221) was significantly increased in lung tissues in ALI mice. The inhibition of miR-221 attenuated lung injury including decreased lung W/D weight ratio and lung permeability and survival rates of ALI mice, as well as apoptosis, whereas its agomir-mediated upregulation exacerbated the lung injury. Concomitantly, miR-221 inhibition significantly reduced LPS-induced pulmonary inflammation, while LPS-induced pulmonary inflammation was aggravated by miR-221 upregulation. Of note, suppressor of cytokine signaling-1 (SOCS1), an effective suppressor of the NF-κB signaling pathway, was found to be a direct target of miR-221 in RAW264.7 cells. Overexpression of SOCS1 by pcDNA-SOCS1 plasmids markedly reversed the miR-221 inhibition-mediated inhibitory effects on inflammation and apoptosis in LPS-treated RAW264.7 cells. Finally, it was found that miR-221 inhibition suppressed LPS induced the activation of the NF-κB signaling pathway, as demonstrated by downregulation of phosphorylated-IκBα, p-p65 and upregulation of IκBα, whilst miR-221 overexpression had an opposite result in ALI mice. Our findings demonstrate that inhibition of miR-221 can alleviate LPS-induced inflammation via inactivation of SOCS1/NF-κB signaling pathway in ALI mice.
Collapse
Affiliation(s)
- Tao Wang
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Lihua Jiang
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Xiaoyong Wei
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Zhenghua Dong
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Bo Liu
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Junbo Zhao
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Lijuan Wang
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Peilin Xie
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Yuxia Wang
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Shangyou Zhou
- a Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| |
Collapse
|
34
|
MicroRNA-155 regulates lipopolysaccharide-induced mucin 5AC overproduction via a suppressor of cytokine signaling 1-mediated mechanism in human bronchial epithelial cells. Respir Physiol Neurobiol 2019; 264:12-18. [DOI: 10.1016/j.resp.2019.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/25/2019] [Accepted: 03/20/2019] [Indexed: 01/15/2023]
|
35
|
Menard KL, Haskins BE, Denkers EY. Impact of Toxoplasma gondii Infection on Host Non-coding RNA Responses. Front Cell Infect Microbiol 2019; 9:132. [PMID: 31157172 PMCID: PMC6530353 DOI: 10.3389/fcimb.2019.00132] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/12/2019] [Indexed: 12/12/2022] Open
Abstract
As an intracellular microbe, Toxoplasma gondii must establish a highly intimate relationship with its host to ensure success as a parasite. Many studies over the last decade-and-a-half have highlighted how the host reshapes its immunoproteome to survive infection, and conversely how the parasite regulates host responses to ensure persistence. The role of host non-protein-coding RNA during infection is a vast and largely unexplored area of emerging interest. The potential importance of this facet of the host-parasite interaction is underscored by current estimates that as much as 80% of the host genome is transcribed into non-translated RNA. Here, we review the current state of knowledge with respect to two major classes of non-coding RNA, microRNA (miRNA) and long non-coding RNA (lncRNA), in the host response to T. gondii infection. These two classes of regulatory RNA are known to have profound and widespread effects on cell function. However, their impact on infection and immunity is not well-understood, particularly for the response to T. gondii. Nevertheless, numerous miRNAs have been identified that are upregulated by Toxoplasma, and emerging evidence suggests a functional role during infection. While the field of lncRNA is in its infancy, it is already clear that Toxoplasma is also a strong trigger for this class of regulatory RNA. Non-coding RNA responses induced by T. gondii are likely to be major determinants of the host's ability to resist infection and the parasite's ability to establish long-term latency.
Collapse
Affiliation(s)
- Kayla L Menard
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| | - Breanne E Haskins
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| | - Eric Y Denkers
- Department of Biology, Center for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
36
|
Aguilar C, Mano M, Eulalio A. Multifaceted Roles of microRNAs in Host-Bacterial Pathogen Interaction. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0002-2019. [PMID: 31152522 PMCID: PMC11026079 DOI: 10.1128/microbiolspec.bai-0002-2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a well-characterized class of small noncoding RNAs that act as major posttranscriptional regulators of gene expression. Accordingly, miRNAs have been associated with a wide range of fundamental biological processes and implicated in human diseases. During the past decade, miRNAs have also been recognized for their role in the complex interplay between the host and bacterial pathogens, either as part of the host response to counteract infection or as a molecular strategy employed by bacteria to subvert host pathways for their own benefit. Importantly, the characterization of downstream miRNA targets and their underlying mechanisms of action has uncovered novel molecular factors and pathways relevant to infection. In this article, we review the current knowledge of the miRNA response to bacterial infection, focusing on different bacterial pathogens, including Salmonella enterica, Listeria monocytogenes, Mycobacterium spp., and Helicobacter pylori, among others.
Collapse
Affiliation(s)
- Carmen Aguilar
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- Functional Genomics and RNA-Based Therapeutics Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
- RNA & Infection Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
37
|
MiR-155 expressed in bone marrow-derived lymphocytes promoted lipopolysaccharide-induced acute lung injury through Ang-2-Tie-2 pathway. Biochem Biophys Res Commun 2019; 510:352-357. [DOI: 10.1016/j.bbrc.2019.01.079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 01/16/2019] [Indexed: 11/19/2022]
|
38
|
Alharris E, Singh NP, Nagarkatti PS, Nagarkatti M. Role of miRNA in the regulation of cannabidiol-mediated apoptosis in neuroblastoma cells. Oncotarget 2019; 10:45-59. [PMID: 30713602 PMCID: PMC6343753 DOI: 10.18632/oncotarget.26534] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/13/2018] [Indexed: 12/28/2022] Open
Abstract
Neuroblastoma (NBL) is one of the most common childhood cancers that originate from the immature nerve cells of the sympathetic system. Studies with NBL cancers have also shown that miRNAs are dysregulated and may play a critical role in pathogenesis. Cannabidiol (CBD) is a non-psychoactive compound found in marijuana which has been previously shown by our laboratory and others to induce apoptosis in cancer cells. However, there are no studies reported to test if CBD mediates these effects through regulation of miRNA. In the current study, therefore, we investigated if CBD induces apoptosis in human NBL cell lines, SH SY5Y and IMR-32, and if it is regulated by miRNA. Our data demonstrated that CBD induces apoptosis in NBL cells through activation of serotonin and vanilloid receptors. We also found that caspase-2 and -3 played an important role in the induction of apoptosis. CBD also significantly reduced NBL cell migration and invasion in vitro. Furthermore, CBD blocked mitochondrial respiration and caused a shift in metabolism towards glycolysis. CBD altered the expression of miRNA specifically, down-regulating hsa-let-7a and upregulating hsa-mir-1972. Downregulation of let-7a increased expression of target caspase-3, and growth arrest specific-7 (GAS-7) genes. Upregulation of hsa-mir-1972 caused decreased expression of BCL2L1 and SIRT2 genes. Together, our studies suggest that CBD-mediated apoptosis in NBL cells is regulated by miRNA.
Collapse
Affiliation(s)
- Esraah Alharris
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Narendra P Singh
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| |
Collapse
|
39
|
Alharris E, Alghetaa H, Seth R, Chatterjee S, Singh NP, Nagarkatti M, Nagarkatti P. Resveratrol Attenuates Allergic Asthma and Associated Inflammation in the Lungs Through Regulation of miRNA-34a That Targets FoxP3 in Mice. Front Immunol 2018; 9:2992. [PMID: 30619345 PMCID: PMC6306424 DOI: 10.3389/fimmu.2018.02992] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/04/2018] [Indexed: 01/01/2023] Open
Abstract
Asthma is a chronic inflammatory disease of airways mediated by T-helper 2 (Th2) cells involving complex signaling pathways. Although resveratrol has previously been shown to attenuate allergic asthma, the role of miRNA in this process has not been studied. We investigated the effect of resveratrol on ovalbumin-induced experimental allergic asthma in mice. To that end, BALB/c mice were immunized with ovalbumin (OVA) intraperitoneally followed by oral gavage of vehicle (OVA-veh) or resveratrol (100 mg/kg body) (OVA-res). On day 7, the experimental groups received intranasal challenge of OVA followed by 7 days of additional oral gavage of vehicle or resveratrol. At day 15, all mice were euthanized and bronchioalveolar fluid (BALF), serum and lung infiltrating cells were collected and analyzed. The data showed that resveratrol significantly reduced IL-5, IL-13, and TGF-β in the serum and BALF in mice with OVA-induced asthma. Also, we saw a decrease in CD3+CD4+, CD3+CD8+, and CD4+IL-4+ cells with increase in CD4+CD25+FOXP3+ cells in pulmonary inflammatory cell infiltrate in OVA-res group when compared to OVA-veh. miRNA expression arrays using lung infiltrating cells showed that resveratrol caused significant alterations in miRNA expression, specifically downregulating the expression of miR-34a. Additionally, miR-34a was found to target FOXP3, as evidenced by enhanced expression of FOXP3 in the lung tissue. Also, transfection studies showed that miR-34a inhibitor upregulated FOXP3 expression while miR-34a-mimic downregulated FOXP3 expression. The current study suggests that resveratrol attenuates allergic asthma by downregulating miR-34a that induces increased expression of FOXP3, a master regulator of Treg development and functions.
Collapse
Affiliation(s)
- Esraah Alharris
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Hasan Alghetaa
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Ratanesh Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina,Columbia, SC, United States
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina,Columbia, SC, United States
| | - Narendra P. Singh
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States,*Correspondence: Prakash Nagarkatti
| |
Collapse
|
40
|
Kadhim S, Singh NP, Zumbrun EE, Cui T, Chatterjee S, Hofseth L, Abood A, Nagarkatti P, Nagarkatti M. Resveratrol-Mediated Attenuation of Staphylococcus aureus Enterotoxin B-Induced Acute Liver Injury Is Associated With Regulation of microRNA and Induction of Myeloid-Derived Suppressor Cells. Front Microbiol 2018; 9:2910. [PMID: 30619104 PMCID: PMC6304356 DOI: 10.3389/fmicb.2018.02910] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022] Open
Abstract
Resveratrol (RES) is a polyphenolic compound found abundantly in plant products including red grapes, peanuts, and mulberries. Because of potent anti-inflammatory properties of RES, we investigated whether RES can protect from Staphylococcal enterotoxin B (SEB)-induced acute liver injury in mice. SEB is a potent super antigen that induces robust inflammation and releases inflammatory cytokines that can be fatal. We observed that SEB caused acute liver injury in mice with increases in enzyme aspartate transaminase (AST) levels, and massive infiltration of immune cells into the liver. Treatment with RES (100 mg/kg body weight) attenuated SEB-induced acute liver injury, as indicated by decreased AST levels and cellular infiltration in the liver. Interestingly, RES treatment increased the number of myeloid derived suppressor cells (MDSCs) in the liver. RES treatment led to alterations in the microRNA (miR) profile in liver mononuclear cells (MNCs) of mice exposed to SEB, and pathway analysis indicated these miRs targeted many inflammatory pathways. Of these, we identified miR-185, which was down-regulated by RES, to specifically target Colony Stimulating Factor (CSF1) using transfection studies. Moreover, the levels of CSF1 were significantly increased in RES-treated SEB mice. Because CSF1 is critical in MDSC induction, our studies suggest that RES may induce MDSCs by down-regulating miR-185 leading to increase the expression of CSF1. The data presented demonstrate for the first time that RES can effectively attenuates SEB-induced acute liver injury and that this may result from its action on miRs and induction of MDSCs.
Collapse
Affiliation(s)
- Sabah Kadhim
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Narendra P. Singh
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Elizabeth E. Zumbrun
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Taixing Cui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| | - Lorne Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Abduladheem Abood
- College of Dental Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
41
|
Aguilar C, Mano M, Eulalio A. MicroRNAs at the Host-Bacteria Interface: Host Defense or Bacterial Offense. Trends Microbiol 2018; 27:206-218. [PMID: 30477908 DOI: 10.1016/j.tim.2018.10.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/17/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Abstract
MicroRNAs are a class of small noncoding RNAs that act as major post-transcriptional regulators of gene expression. They are currently recognized for their important role in the intricate interaction between host and bacterial pathogens, either as part of the host immune response to neutralize infection, or as a molecular strategy employed by bacteria to hijack host pathways for their own benefit. Here, we summarize recent advances on the function of miRNAs during infection of mammalian hosts by bacterial pathogens, highlighting key cellular pathways. In addition, we discuss emerging themes in this field, including the participation of miRNAs in host-microbiota crosstalk and cell-to-cell communication.
Collapse
Affiliation(s)
- Carmen Aguilar
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- Functional Genomics and RNA-based Therapeutics Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany; RNA & Infection Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
42
|
Hu RS, He JJ, Elsheikha HM, Zhang FK, Zou Y, Zhao GH, Cong W, Zhu XQ. Differential Brain MicroRNA Expression Profiles After Acute and Chronic Infection of Mice With Toxoplasma gondii Oocysts. Front Microbiol 2018; 9:2316. [PMID: 30333806 PMCID: PMC6176049 DOI: 10.3389/fmicb.2018.02316] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/11/2018] [Indexed: 12/16/2022] Open
Abstract
Brain microRNAs (miRNAs) change in abundance in response to Toxoplasma gondii infection. However, their precise role in the pathogenesis of cerebral infection with T. gondii oocyst remains unclear. We studied the abundance of miRNAs in the brain of mice on days 11 and 33 post-infection (dpi) in order to identify miRNA pattern specific to early (11 dpi) and late (33 dpi) T. gondii infection. Mice were challenged with T. gondii oocysts (Type II strain) and on 11 and 33 dpi, the expression of miRNAs in mouse brain was investigated using small RNA (sRNA) sequencing. miRNA expression was confirmed by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed to identify the biological processes, molecular functions, and cellular components, as well as pathways involved in infection. More than 1,500 miRNAs (1,352 known and 150 novel miRNAs) were detected in the infected and control mice. The expression of miRNAs varied across time after infection; 3, 38, and 108 differentially expressed miRNAs (P < 0.05) were detected during acute infection, chronic infection and chronic vs. acute infection, respectively. GO analysis showed that chronically infected mice had more predicted targets of dysregulated miRNAs than acutely infected mice. KEGG analysis indicated that most predicted targets were involved in immune- or disease-related pathways. Our data indicate that T. gondii infection alters the abundance of miRNAs in mouse brain particularly at the chronic stage, probably to fine-tune conditions required for the establishment of a latent brain infection.
Collapse
Affiliation(s)
- Rui-Si Hu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, The University of Nottingham, Loughborough, United Kingdom
| | - Fu-Kai Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yang Zou
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Guang-Hui Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Wei Cong
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,College of Marine Science, Shandong University at Weihai, Weihai, China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
43
|
Ling Y, Li ZZ, Zhang JF, Zheng XW, Lei ZQ, Chen RY, Feng JH. RETRACTED: MicroRNA-494 inhibition alleviates acute lung injury through Nrf2 signaling pathway via NQO1 in sepsis-associated acute respiratory distress syndrome. Life Sci 2018; 210:1-8. [PMID: 30121199 PMCID: PMC9673760 DOI: 10.1016/j.lfs.2018.08.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 11/24/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figures 5G,H+I, which appear to have a similar phenotype as many other publications, as detailed here: https://pubpeer.com/publications/7C9483B2551952AD53CCFCE206C4EB; and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. The journal requested that the corresponding author comment on these concerns and provide the raw data. The authors did not respond to this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Yun Ling
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Zheng-Zhao Li
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Jian-Feng Zhang
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China.
| | - Xiao-Wen Zheng
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Zhuo-Qing Lei
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Ru-Yan Chen
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Ji-Hua Feng
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| |
Collapse
|
44
|
Lin X, Chen L, Li H, Liu Y, Guan Y, Li X, Jia Z, Lin X, Jia J, Sun Y, Xiao D. miR-155 accelerates proliferation of mouse hepatocytes during liver regeneration by directly targeting SOCS1. Am J Physiol Gastrointest Liver Physiol 2018; 315:G443-G453. [PMID: 29792529 DOI: 10.1152/ajpgi.00072.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Liver regeneration after two-thirds partial hepatectomy (PH) is a clinically significant repair process for restoring proper liver architecture. Although microRNA-155 (miR-155) has been found to serve as a crucial microRNA regulator that controls liver cell function and proliferation, little is known about its specific role in the regenerating liver. Using a mouse model with miR-155 overexpression or miR-155 knockout, we investigated the molecular mechanisms of miR-155 in liver regeneration. We found a marked induction of miR-155 in C57BL/6 mice after PH. Furthermore, RL-m155 mice showed enhanced liver regeneration as a result of accelerated progression of hepatocytes into the cell cycle, mainly through an increase in cyclin levels. However, proliferation of hepatocytes was delayed in miR-155-deficient livers. Expression of suppressor of cytokine signaling 1 (SOCS1) was dramatically downregulated in the process of liver regeneration, and enhancement of SOCS1 contributed to impaired proliferation of hepatocytes. Additionally, in vitro and in vivo experiments showed that adenovirus- or adeno-associated virus-mediated overexpression of SOCS1 attenuated improved liver regeneration induced by miR-155 overexpression. Our study shows that miR-155 is a pro-proliferative regulator in liver regeneration by facilitating the cell cycle and directly targeting SOCS1. NEW & NOTEWORTHY Our findings suggest a microRNA-155 (miR-155)-mediated positive regulation pattern in liver regeneration. A series of in vivo and in vitro studies showed that miR-155 upregulation enhanced partial hepatectomy-induced proliferation of hepatocytes by promoting the cell cycle without inducing DNA damage or apoptosis. Suppressor of cytokine signaling 1, a target gene of miR-155, antagonized the proliferation-promoting effect of miR-155. Therefore, pharmacological intervention targeting miR-155 may be therapeutically beneficial in various liver diseases.
Collapse
Affiliation(s)
- Xia Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University , Guangzhou , China
| | - Li Chen
- Department of Endocrinology, The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Haiyan Li
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University , Guangzhou , China
| | - Yu Liu
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University , Guangzhou , China
| | - Yanhong Guan
- Department of Endocrinology, The Second Affiliated Hospital, Guangzhou Medical University , Guangzhou , China
| | - Xiaoyan Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University , Guangzhou , China
| | - Zhenchang Jia
- School of Laboratory Medicine and Biotechnology, Southern Medical University , Guangzhou , China
| | - Xiaolin Lin
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University , Guangzhou , China
| | - Junshuang Jia
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University , Guangzhou , China
| | - Yan Sun
- Zhongshan School of Medicine, Sun Yat-sen University , Guangzhou , China
| | - Dong Xiao
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy Research and Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, Southern Medical University , Guangzhou , China.,Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University , Guangzhou , China
| |
Collapse
|
45
|
Alghetaa H, Mohammed A, Sultan M, Busbee P, Murphy A, Chatterjee S, Nagarkatti M, Nagarkatti P. Resveratrol protects mice against SEB-induced acute lung injury and mortality by miR-193a modulation that targets TGF-β signalling. J Cell Mol Med 2018. [PMID: 29512867 PMCID: PMC5908132 DOI: 10.1111/jcmm.13542] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Staphylococcal enterotoxin B (SEB) is a potent superantigen produced by Staphylococcus aureus that triggers a strong immune response, characterized by cytokine storm, multi‐organ failure, and often death. When inhaled, SEB can cause acute lung injury (ALI) and respiratory failure. In this study, we investigated the effect of resveratrol (RES), a phytoallexin, on SEB‐driven ALI and mortality in mice. We used a dual‐exposure model of SEB in C3H/HeJ mice, which caused 100% mortality within the first 5 days of exposure, and treatment with RES resulted in 100% survival of these mice up to 10 days post‐SEB exposure. RES reduced the inflammatory cytokines in the serum and lungs, as well as T cell infiltration into the lungs caused by SEB. Treatment with RES also caused increased production of transforming growth factor‐beta (TGF‐β) in the blood and lungs. RES altered the miRNA profile in the immune cells isolated from the lungs. Of these, miR‐193a was strongly induced by SEB and was down‐regulated by RES treatment. Furthermore, transfection studies and pathway analyses revealed that miR‐193a targeted several molecules involved in TGF‐β signalling (TGFβ2, TGFβR3) and activation of apoptotic pathways death receptor‐6 (DR6). Together, our studies suggest that RES can effectively neutralize SEB‐mediated lung injury and mortality through potential regulation of miRNA that promote anti‐inflammatory activities.
Collapse
Affiliation(s)
- Hasan Alghetaa
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Amira Mohammed
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Muthanna Sultan
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Philip Busbee
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Angela Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Saurabh Chatterjee
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
46
|
Ferreira-Duarte AP, Pinheiro-Torres AS, Anhê GF, Condino-Neto A, Antunes E, DeSouza IA. MHC Class II Activation and Interferon-γ Mediate the Inhibition of Neutrophils and Eosinophils by Staphylococcal Enterotoxin Type A (SEA). Front Cell Infect Microbiol 2017; 7:518. [PMID: 29322036 PMCID: PMC5733477 DOI: 10.3389/fcimb.2017.00518] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 12/04/2017] [Indexed: 12/30/2022] Open
Abstract
Staphylococcal enterotoxins are classified as superantigens that act by linking T-cell receptor with MHC class II molecules, which are expressed on classical antigen-presenting cells (APC). Evidence shows that MHC class II is also expressed in neutrophils and eosinophils. This study aimed to investigate the role of MHC class II and IFN-γ on chemotactic and adhesion properties of neutrophils and eosinophils after incubation with SEA. Bone marrow (BM) cells obtained from BALB/c mice were resuspended in culture medium, and incubated with SEA (3–30 ng/ml; 1–4 h), after which chemotaxis and adhesion were evaluated. Incubation with SEA significantly reduced the chemotactic and adhesive responses in BM neutrophils activated with IL-8 (200 ng/ml). Likewise, SEA significantly reduced the chemotactic and adhesive responses of BM eosinophils activated with eotaxin (300 ng/ml). The inhibitory effects of SEA on cell chemotaxis and adhesion were fully prevented by prior incubation with an anti-MHC class II blocking antibody (2 μg/ml). SEA also significantly reduced the intracellular Ca2+ levels in IL-8- and eotaxin-activated BM cells. No alterations of MAC-1, VLA4, and LFA-1α expressions were observed after SEA incubation. In addition, SEA elevated by 3.5-fold (P < 0.05) the INF-γ levels in BM cells. Incubation of BM leukocytes with IFN-γ (10 ng/ml, 2 h) reduced both neutrophil and eosinophil chemotaxis and adhesion, which were prevented by prior incubation with anti-MHC class II antibody (2 μg/ml). In conclusion, SEA inhibits neutrophil and eosinophil by MHC class II-dependent mechanism, which may be modulated by concomitant release of IFN-γ.
Collapse
Affiliation(s)
- Ana P Ferreira-Duarte
- Department of Biology and Physiology, Faculty of Medicine of Jundiai, Jundiai, Brazil
| | | | - Gabriel F Anhê
- Department of Pharmacology, State University of Campinas, Campinas, Brazil
| | - Antônio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edson Antunes
- Department of Pharmacology, State University of Campinas, Campinas, Brazil
| | - Ivani A DeSouza
- Department of Biology and Physiology, Faculty of Medicine of Jundiai, Jundiai, Brazil
| |
Collapse
|
47
|
Neudecker V, Yuan X, Bowser JL, Eltzschig HK. MicroRNAs in mucosal inflammation. J Mol Med (Berl) 2017; 95:935-949. [PMID: 28726085 DOI: 10.1007/s00109-017-1568-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/28/2017] [Accepted: 07/04/2017] [Indexed: 12/12/2022]
Abstract
Of the total human body's surface, the majority is internal surface, belonging to the lungs (100 m2) and intestinal tract (400 m2). In comparison, the external surface area, belonging to the skin, comprises less than 1% (2 m2). Continuous exposure of the mucosal surface to external factors (e.g., pathogens, food particles) requires tight regulation to maintain homeostasis. MicroRNAs (miRNAs) have gained noticeable attention as playing important roles in maintaining the steady-state of tissues by modulating immune functions and inflammatory responses. Accordingly, associations have been found between miRNA expression levels and human health conditions and diseases. These findings have important implications in inflammatory diseases involving pulmonary and intestinal mucosa, such as acute lung injury or inflammatory bowel disease. In this review, we highlight the known biology of miRNAs and discuss the role of miRNAs in modulating mucosal defense and homeostasis. Additionally, we discuss miRNAs serving as potential therapeutic targets to treat immunological conditions, particularly mucosal inflammation.
Collapse
Affiliation(s)
- Viola Neudecker
- Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany.
| | - Xiaoyi Yuan
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jessica L Bowser
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, the University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| |
Collapse
|
48
|
Acosta AC, Santos SJD, Albuquerque L, Soares KDA, Mota RA, Medeiros ESD. Frequência de genes codificadores de toxinas em Staphylococcus aureus isolados de leite de tanques expansão comunitários. PESQUISA VETERINARIA BRASILEIRA 2017. [DOI: 10.1590/s0100-736x2017000700007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
RESUMO: A capacidade de produção de toxinas pelo Staphylococcus aureus no leite e produtos derivados está relacionado com surtos de intoxicação alimentar. Objetivou-se nesta pesquisa, estudar a ocorrência de genes que codificam para enterotoxinas estafilocócicas (sea, seb, sed, seg, seh e sei) e toxinas α e β hemolítica (hla e hlb) em S. aureus isolados de 53 amostras de leite de tanques expansão comunitários no Estado de Alagoas, Brasil. Foram identificados 27 isolados (50,94%) como S. aureus pela amplificação do gene nuc. 13/27 isolados (48,1%) foram positivos para pelo menos um gene das enterotoxinas estudadas, sendo as frequências dos genes sea 33,3%, seh 18,5%, sei 11,1% e sed 7,4%; não entanto não foram identificados os genes seb e seg nestas bactérias. Para as toxinas hemolíticas, 51,9% dos isolados portavam ambos genes (hla e hlb), sendo a frequência para o gene hla de 81,5% e para o gene hlb de 51,9%. A frequência de genes das toxinas avaliadas é alta o que constitui um risco potencial para a saúde pública em especial, as enterotoxinas por serem termoestáveis e estarem asssociados com surtos de intoxicação alimentar.
Collapse
|
49
|
Genistein Protects Against Ox-LDL-Induced Inflammation Through MicroRNA-155/SOCS1-Mediated Repression of NF-ĸB Signaling Pathway in HUVECs. Inflammation 2017; 40:1450-1459. [DOI: 10.1007/s10753-017-0588-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
50
|
Kumar Kingsley SM, Vishnu Bhat B. Role of MicroRNAs in the development and function of innate immune cells. Int Rev Immunol 2017; 36:154-175. [DOI: 10.1080/08830185.2017.1284212] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- S. Manoj Kumar Kingsley
- Department of Neonatology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - B. Vishnu Bhat
- Department of Neonatology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|