1
|
Liu D, Zhang Y, Zhang Y, Huang Q, Meng W, Gao J, Mo X, Tian H, Li S. Chloroquine Alleviates Atherosclerosis by Modulating Regulatory T Cells Through the ATM/AMPK/mTOR Signaling Pathway in ApoE -/- Mice. Exp Clin Endocrinol Diabetes 2023; 131:676-685. [PMID: 38056492 DOI: 10.1055/a-2201-8728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
BACKGROUND Clinical observation suggests the atheroprotective effect of chloroquine and its derivatives, while its mechanism remains unclear. This study aimed to observe the protective effect of chloroquine against atherosclerosis and explore the underlying mechanism. METHODS Ataxia telangiectasia mutated (ATM) wild-type or haploinsufficient apolipoprotein-E-knockout (ATM+/+ApoE-/- or ATM+/-ApoE-/-) mice were treated with different dosages of chloroquine. Anti-CD25 antibody was used to deplete natural Tregs in ATM+/+ApoE-/- mice. The atherosclerotic burden in different groups of mice was comprehensively evaluated by H&E staining and Masson staining. The effect of chloroquine on the regulatory T cells (Tregs) was assessed in vivo and in vitro by flow cytometry and immunohistochemical staining. The expression of related proteins was detected by real-time polymerase chain reaction and western blotting. RESULTS In ATM+/+ApoE-/- mice, chloroquine alleviated atherosclerotic lesions, stabilized the plaque, and increased Treg counts in the atherosclerotic lesions and spleens. However, in ATM haploinsufficient mice (ATM+/-ApoE-/-), chloroquine no longer prevented atherosclerosis or impacted Treg counts. Abolishing Treg cells using an anti-CD25 antibody in vivo abrogated the atheroprotective effect of chloroquine. In vitro, chloroquine promoted the differentiation of Tregs from naïve T cells, which was accompanied by enhanced ATM/AMP-activated protein kinase (AMPK) activity and reduced downstream mammalian target of rapamycin (mTOR) activity. DISCUSSION These findings suggest that chloroquine ameliorates atherosclerosis and stabilizes plaque by modulating Tregs differentiation through the regulation of the ATM/AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Dan Liu
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Center for Stem Cell Research and Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Yiyi Zhang
- Department of Endocrinology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences, Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China
| | - Qiaorong Huang
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wentong Meng
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xianming Mo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Haoming Tian
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Sheyu Li
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Li W, Wu M, Li Y, Shen J. Reactive nitrogen species as therapeutic targets for autophagy/mitophagy modulation to relieve neurodegeneration in multiple sclerosis: Potential application for drug discovery. Free Radic Biol Med 2023; 208:37-51. [PMID: 37532065 DOI: 10.1016/j.freeradbiomed.2023.07.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023]
Abstract
Multiple sclerosis (MS) is a neuroinflammatory disease with limited therapeutic effects, eventually developing into handicap. Seeking novel therapeutic strategies for MS is timely important. Active autophagy/mitophagy could mediate neurodegeneration, while its roles in MS remain controversial. To elucidate the exact roles of autophagy/mitophagy and reveal its in-depth regulatory mechanisms, we conduct a systematic literature study and analyze the factors that might be responsible for divergent results obtained. The dynamic change levels of autophagy/mitophagy appear to be a determining factor for final neuron fate during MS pathology. Excessive neuronal autophagy/mitophagy contributes to neurodegeneration after disease onset at the active MS phase. Reactive nitrogen species (RNS) serve as key regulators for redox-related modifications and participate in autophagy/mitophagy modulation in MS. Nitric oxide (•NO) and peroxynitrite (ONOO-), two representative RNS, could nitrate or nitrosate Drp1/parkin/PINK1 pathway, activating excessive mitophagy and aggravating neuronal injury. Targeting RNS-mediated excessive autophagy/mitophagy could be a promising strategy for developing novel anti-MS drugs. In this review, we highlight the important roles of RNS-mediated autophagy/mitophagy in neuronal injury and review the potential therapeutic compounds with the bioactivities of inhibiting RNS-mediated autophagy/mitophagy activation and attenuating MS progression. Overall, we conclude that reactive nitrogen species could be promising therapeutic targets to regulate autophagy/mitophagy for multiple sclerosis treatment.
Collapse
Affiliation(s)
- Wenting Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Yuzhen Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
| | - Jiangang Shen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
3
|
Műzes G, Sipos F. Autoimmunity and Carcinogenesis: Their Relationship under the Umbrella of Autophagy. Biomedicines 2023; 11:biomedicines11041130. [PMID: 37189748 DOI: 10.3390/biomedicines11041130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
The immune system and autophagy share a functional relationship. Both innate and adaptive immune responses involve autophagy and, depending on the disease’s origin and pathophysiology, it may have a detrimental or positive role on autoimmune disorders. As a “double-edged sword” in tumors, autophagy can either facilitate or impede tumor growth. The autophagy regulatory network that influences tumor progression and treatment resistance is dependent on cell and tissue types and tumor stages. The connection between autoimmunity and carcinogenesis has not been sufficiently explored in past studies. As a crucial mechanism between the two phenomena, autophagy may play a substantial role, though the specifics remain unclear. Several autophagy modifiers have demonstrated beneficial effects in models of autoimmune disease, emphasizing their therapeutic potential as treatments for autoimmune disorders. The function of autophagy in the tumor microenvironment and immune cells is the subject of intensive study. The objective of this review is to investigate the role of autophagy in the simultaneous genesis of autoimmunity and malignancy, shedding light on both sides of the issue. We believe our work will assist in the organization of current understanding in the field and promote additional research on this urgent and crucial topic.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
4
|
Kartik S, Pal R, Chaudhary MJ, Nath R, Kumar M, Binwal M, Bawankule DU. Neuroprotective role of chloroquine via modulation of autophagy and neuroinflammation in MPTP-induced Parkinson's disease. Inflammopharmacology 2023; 31:927-941. [PMID: 36715843 DOI: 10.1007/s10787-023-01141-z] [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: 07/14/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is a neuro-motor ailment that strikes adults in their older life and results in both motor and non-motor impairments. In neuronal and glial cells, PD has recently been linked to a dysregulated autophagic system and cerebral inflammation. Chloroquine (CQ), an anti-malarial drug, has been demonstrated to suppress autophagy in a variety of diseases, including cerebral ischemia, Alzheimer's disease (AD), and Traumatic brain injury (TBI), while its involvement in PD is still unclear. BALB/c mice were randomly allocated to one of four groups: 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP), CQ treatment with or without MPTP, or control. The CQ treatment group received CQ (intraperitoneally, 8 mg/kg body weight) after 1 h of MPTP induction on day 1, and it lasted for 7 days. CQ therapy preserves dopamine levels stable, inhibits tyrosine hydroxylase (TH) positive dopaminergic cell death, and lowers oxidative stress. CQ reduces the behavioural, motor, and cognitive deficits caused by MPTP after injury. Furthermore, CQ therapy slowed aberrant neuronal autophagy (microtubule-associated protein-1 light chain 3B; LC3B & Beclin1) and lowered expression levels of the inflammatory cytokines interleukin 1 (IL-1β) and tumour necrosis factor (TNF-α) in the mice brain. In addition, CQ's antioxidant and anti-inflammatory effects were also tested in MPTP-mediated cell death in PC12 cells, demonstrating that CQ has a neurorestorative impact by successfully rescuing MPTP-induced ROS generation and cell loss. Our findings show that CQ's can help to prevent dopaminergic degeneration and improve neurological function after MPTP intoxication by lowering the harmful effects of neuronal autophagy and cerebral inflammation.
Collapse
Affiliation(s)
- Shipra Kartik
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India
| | - Rishi Pal
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India.
| | - Manju J Chaudhary
- Department of Physiology, Government Medical College, Tirwa Road, Kannauj, UP, India
| | - Rajendra Nath
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India
| | - Madhu Kumar
- Department of Pathology, King George's Medical University, Lucknow, UP, 226003, India
| | - Monika Binwal
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, UP, 226015, India
| | - D U Bawankule
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, UP, 226015, India
| |
Collapse
|
5
|
Mills JA, Humphries J, Simpson JD, Sonderegger SE, Thurecht KJ, Fletcher NL. Modulating Macrophage Clearance of Nanoparticles: Comparison of Small-Molecule and Biologic Drugs as Pharmacokinetic Modifiers of Soft Nanomaterials. Mol Pharm 2022; 19:4080-4097. [PMID: 36069540 DOI: 10.1021/acs.molpharmaceut.2c00528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nanomedicines show benefits in overcoming the limitations of conventional drug delivery systems by reducing side effects, toxicity, and exhibiting enhanced pharmacokinetic (PK) profiles to improve the therapeutic window of small-molecule drugs. However, upon administration, many nanoparticles (NPs) prompt induction of host innate immune responses, which in combination with other clearance pathways such as renal and hepatic, eliminate up to 99% of the administered dose. Here, we explore a drug predosing strategy to transiently suppress the mononuclear phagocyte system (MPS), subsequently improving the PK profile and biological behaviors exhibited by a model NP system [hyperbranched polymers (HBPs)] in an immunocompetent mouse model. In vitro assays allowed the identification of five drug candidates that attenuated cellular association. Predosing of lead compounds chloroquine (CQ) and zoledronic acid (ZA) further showed increased HBP retention within the circulatory system of mice, as shown by both fluorescence imaging and positron emission tomography-computed tomography. Flow cytometric evaluation of spleen and liver tissue cells following intravenous administration further demonstrated that CQ and ZA significantly reduced HBP association with myeloid cells by 23 and 16%, respectively. The results of this study support the use of CQ to pharmacologically suppress the MPS to improve NP PKs.
Collapse
Affiliation(s)
- Jessica A Mills
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - James Humphries
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Joshua D Simpson
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Stefan E Sonderegger
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nicholas L Fletcher
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, Queensland 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology and ARC Training Centre for Innovation in Biomedical Imaging Technologies, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
6
|
Murase R, Shingu Y, Wakasa S. Cardioprotective effects of chloroquine pretreatment on ischemic and reperfusion injury via activation of ERK1/2 in isolated rat hearts. Mol Biol Rep 2022; 49:9429-9436. [PMID: 35896843 DOI: 10.1007/s11033-022-07801-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/15/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE Several therapeutic agents have been found to prevent myocardial ischemic and reperfusion (I/R) injury after cardiac surgery; however, no drug is routinely used to afford cardioprotective benefits in clinical settings. Herein, we aimed to determine whether chloroquine (CQ) pretreatment attenuates I/R injury after global ischemia in isolated rat hearts and elucidate mechanisms underlying the effects of CQ. METHODS Isolated rat hearts were subjected to 30-min global ischemia, followed by 60-min reperfusion with Krebs-Henseleit buffer (KHB). Immediately before ischemia, 10 mL of pretreatment solutions (KHB, n = 4 or KHB + CQ [100 μM], n = 4) were injected through the aortic root. Cardiac function was examined based on the rate pressure product (RPP). Myocardial apoptosis was evaluated using TUNEL staining. To assess the reperfusion ischemia salvage kinase pathway, protein expression levels of AKT and extracellular signal-regulated kinase (ERK1/2) were determined using western blotting. To investigate the role of ERK1/2, an ERK1/2 selective inhibitor was used in eight additional rats. RESULTS The recovery rate of the RPP was higher in the KHB + CQ group than in the KHB group 60 min after I/R (KHB, 44 ± 3% vs. KHB + CQ, 69 ± 7%; P = 0.019, d = 2.2). CQ pretreatment reduced apoptosis and enhanced the phosphorylation of ERK1/2; however, AKT phosphorylation was unaltered. In addition, the ERK1/2 inhibitor abolished CQ-mediated cardioprotective effects. CONCLUSIONS CQ pretreatment showed protective effects on cardiac function after I/R by activating ERK1/2.
Collapse
Affiliation(s)
- Ryota Murase
- Department of Cardiovascular and Thoracic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kitaku, Sapporo, 060-8638, Japan
| | - Yasushige Shingu
- Department of Cardiovascular and Thoracic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kitaku, Sapporo, 060-8638, Japan.
| | - Satoru Wakasa
- Department of Cardiovascular and Thoracic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kitaku, Sapporo, 060-8638, Japan
| |
Collapse
|
7
|
Mills JA, Liu F, Jarrett TR, Fletcher NL, Thurecht KJ. Nanoparticle based medicines: approaches for evading and manipulating the mononuclear phagocyte system and potential for clinical translation. Biomater Sci 2022; 10:3029-3053. [PMID: 35419582 DOI: 10.1039/d2bm00181k] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
For decades, nanomedicines have been reported as a potential means to overcome the limitations of conventional drug delivery systems by reducing side effects, toxicity and the non-ideal pharmacokinetic behaviour typically exhibited by small molecule drugs. However, upon administration many nanoparticles prompt induction of host inflammatory responses due to recognition and uptake by macrophages, eliminating up to 95% of the administered dose. While significant advances in nanoparticle engineering and consequent therapeutic efficacy have been made, it is becoming clear that nanoparticle recognition by the mononuclear phagocyte system (MPS) poses an impassable junction in the current framework of nanoparticle development. Hence, this has negative consequences on the clinical translation of nanotechnology with respect to therapeutic efficacy, systemic toxicity and economic benefit. In order to improve the translation of nanomedicines from bench-to-bedside, there is a requirement to either modify nanomedicines in terms of how they interact with intrinsic processes in the body, or modulate the body to be more accommodating for nanomedicine treatments. Here we provide an overview of the current standard for design elements of nanoparticles, as well as factors to consider when producing nanomedicines that have minimal MPS-nanoparticle interactions; we explore this landscape across the cellular to tissue and organ levels. Further, rather than designing materials to suit the body, a growing research niche involves modulating biological responses to administered nanomaterials. We here discuss how developing strategic methods of MPS 'pre-conditioning' with small molecule or biological drugs, as well as implementing strategic dosing regimens, such as 'decoy' nanoparticles, is essential to increasing nanoparticle therapeutic efficacy. By adopting such a perspective, we hope to highlight the increasing trends in research dedicated to improving nanomedicine translation, and subsequently making a positive clinical impact.
Collapse
Affiliation(s)
- Jessica A Mills
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia. .,Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australia
| | - Feifei Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia. .,Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australia.,ARC Centre for Innovation in Biomedical Imaging Technology, Australia
| | - Thomas R Jarrett
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia. .,Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australia.,ARC Centre for Innovation in Biomedical Imaging Technology, Australia
| | - Nicholas L Fletcher
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia. .,Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia. .,Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD 4072, Australia.,ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australia.,ARC Centre for Innovation in Biomedical Imaging Technology, Australia
| |
Collapse
|
8
|
Cheng YJ, Cheng XY, Zhang YM, Wang F, Wang X, Meng LQ, Liu G, Cui Z, Zhao MH. Effects of hydroxychloroquine on proteinuria in membranous nephropathy. J Nephrol 2021; 35:1145-1157. [PMID: 34846713 DOI: 10.1007/s40620-021-01182-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Many patients with primary membranous nephropathy have severe proteinuria unresponsive to optimized renin-angiotensin-aldosterone system inhibitors (RAASi). We evaluated the efficacy and safety of hydroxychloroquine as an adjunctive agent in membranous nephropathy (MN) treatments. METHODS We prospectively recruited 126 patients with biopsy-proven primary membranous nephropathy and urinary protein 1-8 g/day while receiving optimized RAASi treatment for ≥ 3 months and well-controlled blood pressure. Forty-three patients received hydroxychloroquine and RAASi (hydroxychloroquine-RAASi group), and 83 patients received RAASi alone (RAASi group). Treatment responses, including proteinuria reduction, complete and partial remission rates, and autoantibody against phospholipase A2 receptor (anti-PLA2R) levels, were compared between the two groups at 6 months and over the long term. RESULTS At 6 months, the effective response rate (proteinuria reduction > 30%) (57.5% vs. 28.9%, P = 0.002), clinical remission rate (35.0% vs. 15.7%, P = 0.015), and percentage change in proteinuria (- 51.7% vs. - 21.9%, P < 0.001) were higher, and the rate of switching to immunosuppressants (25.0% vs. 45.8%, P = 0.027) was lower in the hydroxychloroquine-RAASi group than in the RAASi group. Hydroxychloroquine administration was an independent protective factor with an effective response (OR 0.37, P = 0.021). In the long term, the clinical remission rate was higher in the HCQ-RAASi group (62.5% vs. 38.6%, P = 0.013). Hydroxychloroquine therapy was associated with a higher rate of anti-PLA2R reduction (< 20 U/ml) (HR 0.28, P = 0.031). We observed no serious adverse events associated with hydroxychloroquine. CONCLUSIONS Hydroxychloroquine could be an option for patients with membranous nephropathy seeking to achieve proteinuria reduction and anti-PLA2R antibody reduction in addition to optimized RAASi. Randomized controlled trials are needed to confirm these findings. TRIAL REGISTRATION ChiCTR2100045947, 20210430, retrospectively registered.
Collapse
Affiliation(s)
- Yan-Jiao Cheng
- Renal Division, Peking University First Hospital, Beijing, 100034, People's Republic of China.,Institute of Nephrology, Peking University, Beijing, People's Republic of China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - Xu-Yang Cheng
- Renal Division, Peking University First Hospital, Beijing, 100034, People's Republic of China. .,Institute of Nephrology, Peking University, Beijing, People's Republic of China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China. .,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China.
| | - Yi-Miao Zhang
- Renal Division, Peking University First Hospital, Beijing, 100034, People's Republic of China.,Institute of Nephrology, Peking University, Beijing, People's Republic of China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - Fang Wang
- Renal Division, Peking University First Hospital, Beijing, 100034, People's Republic of China.,Institute of Nephrology, Peking University, Beijing, People's Republic of China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - Xin Wang
- Renal Division, Peking University First Hospital, Beijing, 100034, People's Republic of China.,Institute of Nephrology, Peking University, Beijing, People's Republic of China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - Li-Qiang Meng
- Renal Division, Peking University First Hospital, Beijing, 100034, People's Republic of China.,Institute of Nephrology, Peking University, Beijing, People's Republic of China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - Gang Liu
- Renal Division, Peking University First Hospital, Beijing, 100034, People's Republic of China.,Institute of Nephrology, Peking University, Beijing, People's Republic of China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - Zhao Cui
- Renal Division, Peking University First Hospital, Beijing, 100034, People's Republic of China. .,Institute of Nephrology, Peking University, Beijing, People's Republic of China. .,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China. .,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China.
| | - Ming-Hui Zhao
- Renal Division, Peking University First Hospital, Beijing, 100034, People's Republic of China.,Institute of Nephrology, Peking University, Beijing, People's Republic of China.,Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,Key Laboratory of CKD Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China.,Peking-Tsinghua Center for Life Sciences, Beijing, 100871, People's Republic of China
| |
Collapse
|
9
|
Wu MY, Wang EJ, Feng D, Li M, Ye RD, Lu JH. Pharmacological insights into autophagy modulation in autoimmune diseases. Acta Pharm Sin B 2021; 11:3364-3378. [PMID: 34900523 PMCID: PMC8642426 DOI: 10.1016/j.apsb.2021.03.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 12/21/2022] Open
Abstract
As a cellular bulk degradation and survival mechanism, autophagy is implicated in diverse biological processes. Genome-wide association studies have revealed the link between autophagy gene polymorphisms and susceptibility of autoimmune diseases including systemic lupus erythematosus (SLE) and inflammatory bowel disease (IBD), indicating that autophagy dysregulation may be involved in the development of autoimmune diseases. A series of autophagy modulators have displayed protective effects on autoimmune disease models, highlighting the emerging role of autophagy modulators in treating autoimmune diseases. This review explores the roles of autophagy in the autoimmune diseases, with emphasis on four major autoimmune diseases [SLE, rheumatoid arthritis (RA), IBD, and experimental autoimmune encephalomyelitis (EAE)]. More importantly, the therapeutic potentials of small molecular autophagy modulators (including autophagy inducers and inhibitors) on autoimmune diseases are comprehensively analyzed.
Collapse
Affiliation(s)
- Ming-Yue Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 9999078, China
| | - Er-Jin Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 9999078, China
| | - Du Feng
- Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, College of Basic Medical Science, Guangzhou Medical University, Guangzhou 510000, China
| | - Min Li
- School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510000, China
| | - Richard D. Ye
- Kobilka Institute of Innovative Drug Discovery, School of Life and Health Sciences, the Chinese University of Hong Kong, Shenzhen 518000, China
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 9999078, China
| |
Collapse
|
10
|
Yang G, Van Kaer L. Therapeutic Targeting of Immune Cell Autophagy in Multiple Sclerosis: Russian Roulette or Silver Bullet? Front Immunol 2021; 12:724108. [PMID: 34531871 PMCID: PMC8438236 DOI: 10.3389/fimmu.2021.724108] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) in which the immune system damages the protective insulation surrounding nerve fibers that project from neurons. The pathological hallmark of MS is multiple areas of myelin loss accompanied by inflammation within the CNS, resulting in loss of cognitive function that ultimately leads to paralysis. Recent studies in MS have focused on autophagy, a cellular self-eating process, as a potential target for MS treatment. Here, we review the contribution of immune cell autophagy to the pathogenesis of experimental autoimmune encephalomyelitis (EAE), the prototypic animal model of MS. A better understanding of the role of autophagy in different immune cells to EAE might inform the development of novel therapeutic approaches in MS and other autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Guan Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
11
|
Durán N, Nakazato G, Durán M, Berti IR, Castro GR, Stanisic D, Brocchi M, Fávaro WJ, Ferreira-Halder CV, Justo GZ, Tasic L. Multi-target drug with potential applications: violacein in the spotlight. World J Microbiol Biotechnol 2021; 37:151. [PMID: 34398340 DOI: 10.1007/s11274-021-03120-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/02/2021] [Indexed: 11/28/2022]
Abstract
The aim of the current review is to address updated research on a natural pigment called violacein, with emphasis on its production, biological activity and applications. New information about violacein's action mechanisms as antitumor agent and about its synergistic action in drug delivery systems has brought new alternatives for anticancer therapy. Thus, violacein is introduced as reliable drug capable of overcoming at least three cancer hallmarks, namely: proliferative signaling, cell death resistance and metastasis. In addition, antimicrobial effects on several microorganisms affecting humans and other animals turn violacein into an attractive drug to combat resistant pathogens. Emphasis is given to effects of violacein combined with different agents, such as antibiotics, anticancer agents and nanoparticles. Although violacein is well-known for many decades, it remains an attractive compound. Thus, research groups have been making continuous effort to help improving its production in recent years, which can surely enable its pharmaceutical and chemical application as multi-task compound, even in the cosmetics and food industries.
Collapse
Affiliation(s)
- Nelson Durán
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil. .,Nanomedicine Research Unit (Nanomed), Center for Natural and Human Sciences (CCNH), Universidade Federal do ABC (UFABC), Santo André, SP, Brazil.
| | - Gerson Nakazato
- Laboratory of Basic and Applied Bacteriology, Department of Microbiology, Biology Sciences Center, Universidade Estadual de Londrina (UEL), Londrina, PR, Brazil
| | - Marcela Durán
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil.,Nanomedicine Research Unit (Nanomed), Center for Natural and Human Sciences (CCNH), Universidade Federal do ABC (UFABC), Santo André, SP, Brazil
| | - Ignasio R Berti
- Nanobiomaterials Laboratory, Department of Chemistry, School of Sciences, Institute of Applied Biotechnology CINDEFI (UNLPCONICET, CCT La Plata),, Universidad Nacional de La Plata, La Plata, Argentina
| | - Guillermo R Castro
- Nanobiomaterials Laboratory, Department of Chemistry, School of Sciences, Institute of Applied Biotechnology CINDEFI (UNLPCONICET, CCT La Plata),, Universidad Nacional de La Plata, La Plata, Argentina
| | - Danijela Stanisic
- Biological Chemistry Laboratory, Institute of Chemistry, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Marcelo Brocchi
- Laboratory of Tropical Diseases, Department of Genetic, Evolution and Bioagents , Biology Institute, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Wagner J Fávaro
- Laboratory of Urogenital Carcinogenesis and Immunotherapy, Department of Structural and Functional Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Carmen V Ferreira-Halder
- Departamento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Giselle Z Justo
- Departamento de Ciências Farmacêuticas (Campus Diadema) e Departamento de Bioquímica (Campus São Paulo), Universidade Federal de São Paulo (UNIFESP), 3 de Maio, 100, São Paulo, SP, 04044-020, Brazil.
| | - Ljubica Tasic
- Biological Chemistry Laboratory, Institute of Chemistry, Universidade Estadual de Campinas, Campinas, SP, Brazil
| |
Collapse
|
12
|
Krueger J, Santinon F, Kazanova A, Issa ME, Larrivee B, Kremer R, Milhalcioiu C, Rudd CE. Hydroxychloroquine (HCQ) decreases the benefit of anti-PD-1 immune checkpoint blockade in tumor immunotherapy. PLoS One 2021; 16:e0251731. [PMID: 34181666 PMCID: PMC8238207 DOI: 10.1371/journal.pone.0251731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/03/2021] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy using checkpoint blockade (ICB) with antibodies such as anti-PD-1 has revolutionised the treatment of many cancers. Despite its use to treat COVID-19 patients and autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis, the effect of hydroxychloroquine (HCQ) on cancer immunotherapy has not been examined. In this study, remarkably, we find that HCQ alone, or in combination with azithromycin (AZ), at doses used to treat patients, decreased the therapeutic benefit of anti-PD-1 in cancer immunotherapy. No deleterious effect was seen on untreated tumors. Mechanistically, HCQ and HCQ/AZ inhibited PD-L1 expression on tumor cells, while specifically targeting the anti-PD-1 induced increase in progenitor CD8+CD44+PD-1+TCF1+ tumor infiltrating T cells (TILs) and the generation of CD8+CD44+PD-1+ effectors. Surprisingly, it also impaired the appearance of a subset of terminally exhausted CD8+ TILs. No effect was seen on the presence of CD4+ T cells, FoxP3+ regulatory T cells (Tregs), thymic subsets, B cells, antibody production, myeloid cells, or the vasculature of mice. This study indicates for the first time that HCQ and HCQ/AZ negatively impact the ability of anti-PD-1 checkpoint blockade to promote tumor rejection.
Collapse
Affiliation(s)
- Janna Krueger
- Division of Immuno-Oncology, Research Center Maisonneuve-Rosemont Hospital (CR-HMR) Montreal, Quebec, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, Quebec, Canada
| | - Francois Santinon
- Division of Immuno-Oncology, Research Center Maisonneuve-Rosemont Hospital (CR-HMR) Montreal, Quebec, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, Quebec, Canada
| | - Alexandra Kazanova
- Division of Immuno-Oncology, Research Center Maisonneuve-Rosemont Hospital (CR-HMR) Montreal, Quebec, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, Quebec, Canada
| | - Mark E. Issa
- Division of Immuno-Oncology, Research Center Maisonneuve-Rosemont Hospital (CR-HMR) Montreal, Quebec, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, Quebec, Canada
| | - Bruno Larrivee
- Department of Ophthalmology, Universite de Montreal, Montreal, Quebec, Canada
| | - Richard Kremer
- Division of Experimental Medicine, Department of Medicine, McGill University Health Center, Montreal, Canada
| | - Catalin Milhalcioiu
- Division of Medical Oncology, Department of Medicine, McGill University Health Center, Montreal, Canada
| | - Christopher E. Rudd
- Division of Immuno-Oncology, Research Center Maisonneuve-Rosemont Hospital (CR-HMR) Montreal, Quebec, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, Quebec, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University Health Center, Montreal, Canada
| |
Collapse
|
13
|
Wang Y, Zheng J, Islam MS, Yang Y, Hu Y, Chen X. The role of CD4 +FoxP3 + regulatory T cells in the immunopathogenesis of COVID-19: implications for treatment. Int J Biol Sci 2021; 17:1507-1520. [PMID: 33907514 PMCID: PMC8071774 DOI: 10.7150/ijbs.59534] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/15/2021] [Indexed: 01/08/2023] Open
Abstract
The severe cases of Coronavirus Disease 2019 (COVID-19) frequently exhibit excessive inflammatory responses, acute respiratory distress syndrome (ARDS), coagulopathy, and organ damage. The most striking immunopathology of advanced COVID-19 is cytokine release syndrome or "cytokine storm" that is attributable to the deficiencies in immune regulatory mechanisms. CD4+FoxP3+ regulatory T cells (Tregs) are central regulators of immune responses and play an indispensable role in the maintenance of immune homeostasis. Tregs are likely involved in the attenuation of antiviral defense at the early stage of infection and ameliorating inflammation-induced organ injury at the late stage of COVID-19. In this article, we review and summarize the current understanding of the change of Tregs in patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and discuss the potential role of Tregs in the immunopathology of COVID-19. The emerging concept of Treg-targeted therapies, including both adoptive Treg transfer and low dose of IL-2 treatment, is introduced. Furthermore, the potential Treg-boosting effect of therapeutic agents used in the treatment of COVID-19, including dexamethasone, vitamin D, tocilizumab and sarilumab, chloroquine, hydroxychloroquine, azithromycin, adalimumab and tetrandrine, is discussed. The problems in the current study of Treg cells in COVID-19 and future perspectives are also addressed.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR 999078, China
| |
Collapse
|
14
|
Fouladseresht H, Doroudchi M, Rokhtabnak N, Abdolrahimzadehfard H, Roudgari A, Sabetian G, Paydar S. Predictive monitoring and therapeutic immune biomarkers in the management of clinical complications of COVID-19. Cytokine Growth Factor Rev 2021; 58:32-48. [PMID: 33199179 PMCID: PMC7544568 DOI: 10.1016/j.cytogfr.2020.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022]
Abstract
The coronavirus disease-2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), appears with a wide spectrum of mild-to-critical clinical complications. Many clinical and experimental findings suggest the role of inflammatory mechanisms in the immunopathology of COVID-19. Hence, cellular and molecular mediators of the immune system can be potential targets for predicting, monitoring, and treating the progressive complications of COVID-19. In this review, we assess the latest cellular and molecular data on the immunopathology of COVID-19 according to the pathological evidence (e.g., mucus and surfactants), dysregulations of pro- and anti-inflammatory mediators (e.g., cytokines and chemokines), and impairments of innate and acquired immune system functions (e.g., mononuclear cells, neutrophils and antibodies). Furthermore, we determine the significance of immune biomarkers for predicting, monitoring, and treating the progressive complications of COVID-19. We also discuss the clinical importance of recent immune biomarkers in COVID-19, and at the end of each section, recent clinical trials in immune biomarkers for COVID-19 are mentioned.
Collapse
Affiliation(s)
- Hamed Fouladseresht
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Rokhtabnak
- Department of Biology, Faculty of Science, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Hossein Abdolrahimzadehfard
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Roudgari
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Golnar Sabetian
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahram Paydar
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
15
|
Relevance of Autophagy and Mitophagy Dynamics and Markers in Neurodegenerative Diseases. Biomedicines 2021; 9:biomedicines9020149. [PMID: 33557057 PMCID: PMC7913851 DOI: 10.3390/biomedicines9020149] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/18/2022] Open
Abstract
During the past few decades, considerable efforts have been made to discover and validate new molecular mechanisms and biomarkers of neurodegenerative diseases. Recent discoveries have demonstrated how autophagy and its specialized form mitophagy are extensively associated with the development, maintenance, and progression of several neurodegenerative diseases. These mechanisms play a pivotal role in the homeostasis of neural cells and are responsible for the clearance of intracellular aggregates and misfolded proteins and the turnover of organelles, in particular, mitochondria. In this review, we summarize recent advances describing the importance of autophagy and mitophagy in neurodegenerative diseases, with particular attention given to multiple sclerosis, Parkinson’s disease, and Alzheimer’s disease. We also review how elements involved in autophagy and mitophagy may represent potential biomarkers for these common neurodegenerative diseases. Finally, we examine the possibility that the modulation of autophagic and mitophagic mechanisms may be an innovative strategy for overcoming neurodegenerative conditions. A deeper knowledge of autophagic and mitophagic mechanisms could facilitate diagnosis and prognostication as well as accelerate the development of therapeutic strategies for neurodegenerative diseases.
Collapse
|
16
|
Roth-Walter F, Adcock IM, Benito-Villalvilla C, Bianchini R, Bjermer L, Boyman O, Caramori G, Cari L, Fan Chung K, Diamant Z, Eguiluz-Gracia I, Knol EF, Kolios A, Levi-Schaffer F, Nocentini G, Palomares O, Redegeld F, Van Esch B, Stellato C. Immune modulation via T regulatory cell enhancement: Disease-modifying therapies for autoimmunity and their potential for chronic allergic and inflammatory diseases-An EAACI position paper of the Task Force on Immunopharmacology (TIPCO). Allergy 2021; 76:90-113. [PMID: 32593226 DOI: 10.1111/all.14478] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022]
Abstract
Therapeutic advances using targeted biologicals and small-molecule drugs have achieved significant success in the treatment of chronic allergic, autoimmune, and inflammatory diseases particularly for some patients with severe, treatment-resistant forms. This has been aided by improved identification of disease phenotypes. Despite these achievements, not all severe forms of chronic inflammatory and autoimmune diseases are successfully targeted, and current treatment options, besides allergen immunotherapy for selected allergic diseases, fail to change the disease course. T cell-based therapies aim to cure diseases through the selective induction of appropriate immune responses following the delivery of engineered, specific cytotoxic, or regulatory T cells (Tregs). Adoptive cell therapies (ACT) with genetically engineered T cells have revolutionized the oncology field, bringing curative treatment for leukemia and lymphoma, while therapies exploiting the suppressive functions of Tregs have been developed in nononcological settings, such as in transplantation and autoimmune diseases. ACT with Tregs are also being considered in nononcological settings such as cardiovascular disease, obesity, and chronic inflammatory disorders. After describing the general features of T cell-based approaches and current applications in autoimmune diseases, this position paper reviews the experimental models testing or supporting T cell-based approaches, especially Treg-based approaches, in severe IgE-mediated responses and chronic respiratory airway diseases, such as severe asthma and COPD. Along with an assessment of challenges and unmet needs facing the application of ACT in these settings, this article underscores the potential of ACT to offer curative options for patients with severe or treatment-resistant forms of these immune-driven disorders.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Ian M Adcock
- Molecular Cell Biology Group, National Heart & Lung Institute, Imperial College London, London, UK
| | - Cristina Benito-Villalvilla
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Rodolfo Bianchini
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
| | - Leif Bjermer
- Department of Respiratory Medicine and Allergology, Lung and Allergy research, Allergy, Asthma and COPD Competence Center, Lund University, Lund, Sweden
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gaetano Caramori
- Department of Biomedical Sciences, Dentistry and Morphological and Functional Imaging (BIOMORF), Respiratory Medicine Unit, University of Messina, Messina, Italy
| | - Luigi Cari
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Kian Fan Chung
- Experimental Studies Medicine at National Heart & Lung Institute, Imperial College London & Royal Brompton & Harefield NHS Trust, London, UK
| | - Zuzana Diamant
- Department of Respiratory Medicine and Allergology, Institute for Clinical Science, Skane University Hospital, Lund, Sweden
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Department of Clinical Pharmacy & Pharmacology, University Groningen, University Medical Center Groningen and QPS-NL, Groningen, Netherlands
| | - Ibon Eguiluz-Gracia
- Allergy Unit, Hospital Regional Universitario de Málaga-Instituto de Investigación Biomédica de Málaga (IBIMA)-ARADyAL, Málaga, Spain
| | - Edward F Knol
- Departments of Immunology and Dermatology/Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Antonios Kolios
- Department of Immunology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Francesca Levi-Schaffer
- Pharmacology Unit, Faculty of Medicine, Institute for Drug Research, The Hebrew University of Jerusalem, Israel
| | - Giuseppe Nocentini
- Department of Medicine, Section of Pharmacology, University of Perugia, Perugia, Italy
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Frank Redegeld
- Faculty of Science, Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Betty Van Esch
- Faculty of Science, Division of Pharmacology, Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| |
Collapse
|
17
|
Nutma E, Marzin MC, Cillessen SA, Amor S. Autophagy in white matter disorders of the CNS: mechanisms and therapeutic opportunities. J Pathol 2020; 253:133-147. [PMID: 33135781 PMCID: PMC7839724 DOI: 10.1002/path.5576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/21/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022]
Abstract
Autophagy is a constitutive process that degrades, recycles and clears damaged proteins or organelles, yet, despite activation of this pathway, abnormal proteins accumulate in neurons in neurodegenerative diseases and in oligodendrocytes in white matter disorders. Here, we discuss the role of autophagy in white matter disorders, including neurotropic infections, inflammatory diseases such as multiple sclerosis, and in hereditary metabolic disorders and acquired toxic‐metabolic disorders. Once triggered due to cell stress, autophagy can enhance cell survival or cell death that may contribute to oligodendrocyte damage and myelin loss in white matter diseases. For some disorders, the mechanisms leading to myelin loss are clear, whereas the aetiological agent and pathological mechanisms are unknown for other myelin disorders, although emerging studies indicate that a common mechanism underlying these disorders is dysregulation of autophagic pathways. In this review we discuss the alterations in the autophagic process in white matter disorders and the potential use of autophagy‐modulating agents as therapeutic approaches in these pathological conditions. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Manuel C Marzin
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Saskia Agm Cillessen
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands.,Department of Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
18
|
CCR6 blockade on regulatory T cells ameliorates experimental model of multiple sclerosis. Cent Eur J Immunol 2020; 45:256-266. [PMID: 33437177 PMCID: PMC7790011 DOI: 10.5114/ceji.2020.101241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/30/2019] [Indexed: 01/28/2023] Open
Abstract
Regulatory T cells (Tregs) play a significant role in limiting damage of tissue affected by autoimmune process, which has been demonstrated in various experimental models for multiple sclerosis (MS) (mostly experimental autoimmune encephalomyelitis – EAE), rheumatoid arthritis, and type 1 diabetes. In this study, we demonstrated that Tregs increasingly migrate to central nervous system (CNS) during subsequent phases of EAE (preclinical, initial attack, and remission). In contrast, in peripheral tissues (blood, lymph nodes, and spleen), a significant accumulation of Tregs is mostly present during EAE remission. Moreover, an increased expression of CCR6 on Tregs in the CNS, blood, lymph nodes, and spleen in all phases of EAE was observed. The highest expression of CCR6 on Tregs from the CNS, lymph nodes, and spleen was noted during the initial attack of EAE, whereas in the blood, the peak expression of CCR6 was detected during the preclinical phase. The presence of Tregs in the CNS during EAE was confirmed by immunohistochemistry. To analyze additional functional significance of CCR6 expression on Tregs for EAE pathology, we modulated the clinical course of this MS model using Tregs with blocked CCR6. EAE mice, which received CCR6-deficient Tregs showed significant amelioration of disease severity. This observation suggests that CCR6 on Tregs may be a potential target for future therapeutic interventions in MS.
Collapse
|
19
|
Martinez GP, Zabaleta ME, Di Giulio C, Charris JE, Mijares MR. The Role of Chloroquine and Hydroxychloroquine in Immune Regulation and Diseases. Curr Pharm Des 2020; 26:4467-4485. [DOI: 10.2174/1381612826666200707132920] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are derivatives of the heterocyclic aromatic compound
quinoline. These economical compounds have been used as antimalarial agents for many years. Currently,
they are used as monotherapy or in conjunction with other therapies for the treatment of autoimmune diseases
such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjögren's syndrome (SS) and antiphospholipid
antibody syndrome (APS). Based on its effects on the modulation of the autophagy process, various
clinical studies suggest that CQ and HCQ could be used in combination with other chemotherapeutics for the
treatment of various types of cancer. Furthermore, the antiviral effects showed against Zika, Chikungunya, and
HIV are due to the annulation of endosomal/lysosomal acidification. Recently, CQ and HCQ were approved for
the U.S. Food and Drug Administration (FDA) for the treatment of infected patients with the coronavirus SARSCoV-
2, causing the disease originated in December 2019, namely COVID-2019. Several mechanisms have been
proposed to explain the pharmacological effects of these drugs: 1) disruption of lysosomal and endosomal pH, 2)
inhibition of protein secretion/expression, 3) inhibition of antigen presentation, 4) decrease of proinflammatory
cytokines, 5) inhibition of autophagy, 6) induction of apoptosis and 7) inhibition of ion channels activation. Thus,
evidence has shown that these structures are leading molecules that can be modified or combined with other
therapeutic agents. In this review, we will discuss the most recent findings in the mechanisms of action of CQ and
HCQ in the immune system, and the use of these antimalarial drugs on diseases.
Collapse
Affiliation(s)
- Gricelis P. Martinez
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| | - Mercedes E. Zabaleta
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| | - Camilo Di Giulio
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| | - Jaime E. Charris
- Organic Synthesis Laboratory, Faculty of Pharmacy, Central University of Venezuela, 47206, Los Chaguaramos 1041-A, Caracas, Venezuela
| | - Michael R. Mijares
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, 50109, Los Chaguaramos 1050-A, Caracas, Venezuela
| |
Collapse
|
20
|
Elucidating the Pivotal Immunomodulatory and Anti-Inflammatory Potentials of Chloroquine and Hydroxychloroquine. J Immunol Res 2020; 2020:4582612. [PMID: 33062720 PMCID: PMC7533005 DOI: 10.1155/2020/4582612] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/03/2020] [Indexed: 12/30/2022] Open
Abstract
Chloroquine (CQ) and hydroxychloroquine (HCQ) are derivatives of 4-aminoquinoline compounds with over 60 years of safe clinical usage. CQ and HCQ are able to inhibit the production of cytokines such as interleukin- (IL-) 1, IL-2, IL-6, IL-17, and IL-22. Also, CQ and HCQ inhibit the production of interferon- (IFN-) α and IFN-γ and/or tumor necrotizing factor- (TNF-) α. Furthermore, CQ blocks the production of prostaglandins (PGs) in the intact cell by inhibiting substrate accessibility of arachidonic acid necessary for the production of PGs. Moreover, CQ affects the stability between T-helper cell (Th) 1 and Th2 cytokine secretion by augmenting IL-10 production in peripheral blood mononuclear cells (PBMCs). Additionally, CQ is capable of blocking lipopolysaccharide- (LPS-) triggered stimulation of extracellular signal-modulated extracellular signal-regulated kinases 1/2 in human PBMCs. HCQ at clinical levels effectively blocks CpG-triggered class-switched memory B-cells from differentiating into plasmablasts as well as producing IgG. Also, HCQ inhibits cytokine generation from all the B-cell subsets. IgM memory B-cells exhibits the utmost cytokine production. Nevertheless, CQ triggers the production of reactive oxygen species. A rare, but serious, side effect of CQ or HCQ in nondiabetic patients is hypoglycaemia. Thus, in critically ill patients, CQ and HCQ are most likely to deplete all the energy stores of the body leaving the patient very weak and sicker. We advocate that, during clinical usage of CQ and HCQ in critically ill patients, it is very essential to strengthen the CQ or HCQ with glucose infusion. CQ and HCQ are thus potential inhibitors of the COVID-19 cytokine storm.
Collapse
|
21
|
Prasad H, Rao R. Endosomal Acid-Base Homeostasis in Neurodegenerative Diseases. Rev Physiol Biochem Pharmacol 2020; 185:195-231. [PMID: 32737755 PMCID: PMC7614123 DOI: 10.1007/112_2020_25] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Neurodegenerative disorders are debilitating and largely untreatable conditions that pose a significant burden to affected individuals and caregivers. Overwhelming evidence supports a crucial preclinical role for endosomal dysfunction as an upstream pathogenic hub and driver in Alzheimer's disease (AD) and related neurodegenerative disorders. We present recent advances on the role of endosomal acid-base homeostasis in neurodegeneration and discuss evidence for converging mechanisms. The strongest genetic risk factor in sporadic AD is the ε4 allele of Apolipoprotein E (ApoE4), which potentiates pre-symptomatic endosomal dysfunction and prominent amyloid beta (Aβ) pathology, although how these pathways are linked mechanistically has remained unclear. There is emerging evidence that the Christianson syndrome protein NHE6 is a prominent ApoE4 effector linking endosomal function to Aβ pathologies. By functioning as a dominant leak pathway for protons, the Na+/H+ exchanger activity of NHE6 limits endosomal acidification and regulates β-secretase (BACE)-mediated Aβ production and LRP1 receptor-mediated Aβ clearance. Pathological endosomal acidification may impact both Aβ generation and clearance mechanisms and emerges as a promising therapeutic target in AD. We also offer our perspective on the complex role of endosomal acid-base homeostasis in the pathogenesis of neurodegeneration and its therapeutic implications for neuronal rescue and repair strategies.
Collapse
Affiliation(s)
- Hari Prasad
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India, Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rajini Rao
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
22
|
Vabret N, Britton GJ, Gruber C, Hegde S, Kim J, Kuksin M, Levantovsky R, Malle L, Moreira A, Park MD, Pia L, Risson E, Saffern M, Salomé B, Esai Selvan M, Spindler MP, Tan J, van der Heide V, Gregory JK, Alexandropoulos K, Bhardwaj N, Brown BD, Greenbaum B, Gümüş ZH, Homann D, Horowitz A, Kamphorst AO, Curotto de Lafaille MA, Mehandru S, Merad M, Samstein RM. Immunology of COVID-19: Current State of the Science. Immunity 2020; 52:910-941. [PMID: 32505227 PMCID: PMC7200337 DOI: 10.1016/j.immuni.2020.05.002] [Citation(s) in RCA: 1143] [Impact Index Per Article: 285.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has affected millions of people worldwide, igniting an unprecedented effort from the scientific community to understand the biological underpinning of COVID19 pathophysiology. In this Review, we summarize the current state of knowledge of innate and adaptive immune responses elicited by SARS-CoV-2 infection and the immunological pathways that likely contribute to disease severity and death. We also discuss the rationale and clinical outcome of current therapeutic strategies as well as prospective clinical trials to prevent or treat SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Nicolas Vabret
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Graham J Britton
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Conor Gruber
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samarth Hegde
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joel Kim
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Kuksin
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Levantovsky
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Louise Malle
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alvaro Moreira
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luisanna Pia
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emma Risson
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Saffern
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bérengère Salomé
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Myvizhi Esai Selvan
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew P Spindler
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Tan
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Verena van der Heide
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jill K Gregory
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Nina Bhardwaj
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian D Brown
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Greenbaum
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zeynep H Gümüş
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dirk Homann
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amir Horowitz
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alice O Kamphorst
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Saurabh Mehandru
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Robert M Samstein
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
23
|
Thome R, Boehm A, Ishikawa LLW, Casella G, Munhoz J, Ciric B, Zhang GX, Rostami A. Comprehensive Analysis of the Immune and Stromal Compartments of the CNS in EAE Mice Reveal Pathways by Which Chloroquine Suppresses Neuroinflammation. Brain Sci 2020; 10:brainsci10060348. [PMID: 32516999 PMCID: PMC7349328 DOI: 10.3390/brainsci10060348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) are neuroinflammatory diseases of the central nervous system (CNS), where leukocytes and CNS resident cells play important roles in disease development and pathogenesis. The antimalarial drug chloroquine (CQ) has been shown to suppress EAE by modulating dendritic cells (DCs) and Th17 cells. However, the mechanism of action by which CQ modulates EAE is far from being elucidated. Here, we comprehensively analyzed the CNS of CQ and PBS-treated EAE mice to identify and characterize the cells that are affected by CQ. Our results show that leukocytes are largely modulated by CQ and have a reduction in the expression of inflammatory markers. Intriguingly, CQ vastly modulated the CNS resident cells astrocytes, oligodendrocytes (OLs) and microglia (MG), with the latter producing IL-10 and IL-12p70. Overall, our results show a panoramic view of the cellular components that are affect by CQ and provide further evidence that drug repurposing of CQ will be beneficial to MS patients.
Collapse
|
24
|
Kim SB, Huh K, Heo JY, Joo EJ, Kim YJ, Choi WS, Kim YJ, Seo YB, Yoon YK, Ku NS, Jeong SJ, Kim SH, Peck KR, Yeom JS. Interim Guidelines on Antiviral Therapy for COVID-19. Infect Chemother 2020; 52:281-304. [PMID: 32342676 PMCID: PMC7335642 DOI: 10.3947/ic.2020.52.2.281] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Indexed: 12/15/2022] Open
Abstract
Since the first case was reported in Wuhan, Hubei Province, China on December 12, 2019, Coronavirus disease 2019 (COVID-19) has spread widely to other countries since January 2020. As of April 16, 2020, 10635 confirmed cases have been reported, with 230 deaths in Korea. COVID-19 patients may be asymptomatic or show various clinical manifestations, including acute symptoms such as fever, fatigue, sore throat; pneumonia presenting as acute respiratory distress syndrome; and multiple organ failure. As COVID-19 has such varied clinical manifestations and case fatality rates, no standard antiviral therapy regimen has been established other than supportive therapy. In the present guideline, we aim to introduce potentially helpful antiviral and other drug therapies based on in vivo and in vitro research and clinical experiences from many countries.
Collapse
Affiliation(s)
- Sun Bean Kim
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyungmin Huh
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung Yeon Heo
- Department of Infectious Diseases, Ajou University school of Medicine, Suwon, Korea
| | - Eun Jeong Joo
- Division of Infectious Diseases, Department of Internal Medicine, Sungkyunkwan University School of Medicine, Kangbuk Samsung hospital, Seoul, Korea
| | - Youn Jeong Kim
- Division of Infectious Diseases, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Won Suk Choi
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Yae Jean Kim
- Division of Infectious Diseases and Immunodeficiency, Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yu Bin Seo
- Division of Infectious Diseases, Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Young Kyung Yoon
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nam Su Ku
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Su Jin Jeong
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyong Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Sup Yeom
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
25
|
Sarma P, Kaur H, Kumar H, Mahendru D, Avti P, Bhattacharyya A, Prajapat M, Shekhar N, Kumar S, Singh R, Singh A, Dhibar DP, Prakash A, Medhi B. Virological and clinical cure in COVID-19 patients treated with hydroxychloroquine: A systematic review and meta-analysis. J Med Virol 2020; 92:776-785. [PMID: 32297988 PMCID: PMC7262144 DOI: 10.1002/jmv.25898] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/12/2020] [Accepted: 04/13/2020] [Indexed: 12/23/2022]
Abstract
Following the demonstration of the efficacy of hydroxychloroquine against severe acute respiratory syndrome coronavirus 2 in vitro, many trials started to evaluate its efficacy in clinical settings. However, no systematic review and meta-analysis have addressed the issue of the safety and efficacy of hydroxychloroquine (HCQ) in coronavirus disease 2019. We conducted a systematic review and meta-analysis with the objectives of evaluation of safety and efficacy of HCQ alone or in combination in terms of "time to clinical cure," "virological cure," "death or clinical worsening of disease," "radiological progression," and safety. RevMan was used for meta-analysis. We searched 16 literature databases out of which seven studies (n = 1358) were included in the systematic review. In terms of clinical cure, two studies reported possible benefit in "time to body temperature normalization" and one study reported less "cough days" in the HCQ arm. Treatment with HCQ resulted in less number of cases showing the radiological progression of lung disease (odds ratio [OR], 0.31, 95% confidence interval [CI], 0.11-0.9). No difference was observed in virological cure (OR, 2.37, 95% CI, 0.13-44.53), death or clinical worsening of disease (OR, 1.37, 95% CI, 1.37-21.97), and safety (OR, 2.19, 95% CI, 0.59-8.18), when compared with the control/conventional treatment. Five studies reported either the safety or efficacy of HCQ + azithromycin. Although seems safe and effective, more data are required for a definitive conclusion. HCQ seems to be promising in terms of less number of cases with radiological progression with a comparable safety profile to control/conventional treatment. We need more data to come to a definite conclusion.
Collapse
Affiliation(s)
- Phulen Sarma
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Hardeep Kaur
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Harish Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Dhruv Mahendru
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Pramod Avti
- Department of Biophysics, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Anusuya Bhattacharyya
- Department of Ophthalmology, Government Medical College and Hospital, Chandigarh, India
| | - Manisha Prajapat
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Nishant Shekhar
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Subodh Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Rahul Singh
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Ashutosh Singh
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Deba Prasad Dhibar
- Department of Internal medicine, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Ajay Prakash
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Research, Chandigarh, India
| |
Collapse
|
26
|
Chloroquine reduces Th17 cell differentiation by stimulating T-bet expression in T cells. Cell Mol Immunol 2020; 18:779-780. [PMID: 32313209 PMCID: PMC7170033 DOI: 10.1038/s41423-020-0432-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 11/09/2022] Open
|
27
|
Varisli L, Cen O, Vlahopoulos S. Dissecting pharmacological effects of chloroquine in cancer treatment: interference with inflammatory signaling pathways. Immunology 2020; 159:257-278. [PMID: 31782148 PMCID: PMC7011648 DOI: 10.1111/imm.13160] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
Chloroquines are 4-aminoquinoline-based drugs mainly used to treat malaria. At pharmacological concentrations, they have significant effects on tissue homeostasis, targeting diverse signaling pathways in mammalian cells. A key target pathway is autophagy, which regulates macromolecule turnover in the cell. In addition to affecting cellular metabolism and bioenergetic flow equilibrium, autophagy plays a pivotal role at the interface between inflammation and cancer progression. Chloroquines consequently have critical effects in tissue metabolic activity and importantly, in key functions of the immune system. In this article, we will review the work addressing the role of chloroquines in the homeostasis of mammalian tissue, and the potential strengths and weaknesses concerning their use in cancer therapy.
Collapse
Affiliation(s)
- Lokman Varisli
- Union of Education and Science Workers (EGITIM SEN), Diyarbakir Branch, Diyarbakir, Turkey
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir, Turkey
| | - Osman Cen
- Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Natural Sciences, Joliet Jr College, Joliet, IL, USA
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
28
|
Mohammadi Barzelighi H, Daraei B, Dastan F. Approaches for the Treatment of SARS-CoV-2 Infection: A Pharmacologic View and Literature Review. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:258-281. [PMID: 33680028 PMCID: PMC7757982 DOI: 10.22037/ijpr.2020.113821.14506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The emergence of a novel Coronavirus disease (COVID-19) inducing acute respiratory distress syndrome (ARDS) was identified in Hubei province of China in December 2019 and rapidly spread worldwide as pandemic and became a public health concern. COVID-19 disease is caused by a new virus known as SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), which has recently offered many challenges and efforts to identify effective drugs for its prevention and treatment. Currently, there is no proven effective approach and medication against this virus. Quickly expanding clinical trials and studies on Coronavirus disease 2019 increase our knowledge regarding SARS-CoV-2 virus and introduce several potential drugs targeting virus moiety or host cell elements. Overall, 3 stages were suggested for SARS-CoV-2 infection according to the disease severity, clinical manifestations, and treatment outcomes, including mild, moderate, and severe. This review aimed to classify and summarize several medications and potential therapies according to the disease 3 stages; however, it is worth noting that no medication and therapy has been effective so far.
Collapse
Affiliation(s)
| | - Bahram Daraei
- Department of Toxicology and Pharmacology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farzaneh Dastan
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Autophagy Is a Tolerance-Avoidance Mechanism that Modulates TCR-Mediated Signaling and Cell Metabolism to Prevent Induction of T Cell Anergy. Cell Rep 2019; 24:1136-1150. [PMID: 30067971 PMCID: PMC6109966 DOI: 10.1016/j.celrep.2018.06.065] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/04/2018] [Accepted: 06/15/2018] [Indexed: 11/25/2022] Open
Abstract
Mocholi et al. show that, following T cell activation, activation of
autophagy constitutes a tolerance-avoidance mechanism that, through modulation
of cell metabolism and specific signaling pathways, allows T cells to engage in
effector responses and avoid anergy. In vivo inhibition of autophagy in T cells
induces tolerance and prevents autoimmunity. In response to activation, CD4+ T cells upregulate autophagy.
However, the functional consequences of that upregulation have not been fully
elucidated. In this study, we identify autophagy as a tolerance-avoidance
mechanism. Our data show that inhibition of autophagy during CD4+ T
cell activation induces a long-lasting state of hypo-responsiveness that is
accompanied by the expression of an anergic gene signature. Cells unable to
induce autophagy after T cell receptor (TCR) engagement show inefficient
mitochondrial respiration and decreased turnover of the protein tyrosine
phosphatase PTPN1, which translates into defective TCR-mediated signaling.
In vivo, inhibition of autophagy during antigen priming
induces T cell anergy and decreases the severity of disease in an experimental
autoimmune encephalomyelitis mouse model. Interestingly, CD4+ T cells
isolated from the synovial fluid of juvenile idiopathic arthritis patients,
while resistant to suboptimal stimulation-induced anergy, can be tolerized with
autophagy inhibitors. We propose that autophagy constitutes a
tolerance-avoidance mechanism, which determines CD4+ T cell fate.
Collapse
|
30
|
Park TY, Jang Y, Kim W, Shin J, Toh HT, Kim CH, Yoon HS, Leblanc P, Kim KS. Chloroquine modulates inflammatory autoimmune responses through Nurr1 in autoimmune diseases. Sci Rep 2019; 9:15559. [PMID: 31664129 PMCID: PMC6820774 DOI: 10.1038/s41598-019-52085-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/12/2019] [Indexed: 12/19/2022] Open
Abstract
For over a half-century the anti-malarial drug chloroquine (CQ) has been used as a therapeutic agent, alone or in combination, to treat autoimmune diseases. However, neither the underlying mechanism(s) of action nor their molecular target(s) are well defined. The orphan nuclear receptor Nurr1 (also known as NR4A2) is an essential transcription factor affecting the development and maintenance of midbrain dopaminergic neurons. In this study, using in vitro T cell differentiation models, we demonstrate that CQ activates TREG cell differentiation and induces Foxp3 gene expression in a Nurr1-dependent manner. Remarkably, CQ appears to induce Nurr1 function by two distinct mechanisms: firstly, by direct binding to Nurr1’s ligand-binding domain and promoting its transcriptional activity and secondly by upregulation of Nurr1 expression through the CREB signaling pathway. In contrast, CQ suppressed gene expression and differentiation of pathogenic TH17 cells. Importantly, using a valid animal model of inflammatory bowel disease (IBD), we demonstrated that CQ promotes Foxp3 expression and differentiation of TREG cells in a Nurr1-dependent manner, leading to significant improvement of IBD-related symptoms. Taken together, these data suggest that CQ ameliorates autoimmune diseases via regulating Nurr1 function/expression and that Nurr1 is a promising target for developing effective therapeutics of human inflammatory autoimmune diseases.
Collapse
Affiliation(s)
- Tae-Yoon Park
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, Massachusetts, 02478, USA
| | - Yongwoo Jang
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, Massachusetts, 02478, USA
| | - Woori Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, Massachusetts, 02478, USA
| | - Joon Shin
- School of Biological Sciences, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Hui Ting Toh
- School of Biological Sciences, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Chun-Hyung Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, Massachusetts, 02478, USA
| | - Ho Sup Yoon
- School of Biological Sciences, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Pierre Leblanc
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, Massachusetts, 02478, USA.
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, 115 Mill Street, Belmont, Massachusetts, 02478, USA. .,Program in Neuroscience and Harvard Stem Cell Institute, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
31
|
Thome R, Bonfanti AP, Rasouli J, Mari ER, Zhang GX, Rostami A, Verinaud L. Chloroquine-treated dendritic cells require STAT1 signaling for their tolerogenic activity. Eur J Immunol 2018; 48:1228-1234. [PMID: 29572810 DOI: 10.1002/eji.201747362] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/20/2018] [Accepted: 03/14/2018] [Indexed: 01/09/2023]
Abstract
MS and EAE are T cell-driven autoimmune diseases of the CNS where IL-17-producing Th17 cells promote damage and are pathogenic. Conversely, tolerogenic DCs induce Treg cells and suppress Th17 cells. Chloroquine (CQ) suppresses EAE through the modulation of DCs by unknown mechanisms. Here, we show that STAT 1 is necessary for CQ-induced tolerogenic DCs (tolDCs) to efficiently suppress EAE. We observed that CQ induces phosphorylation of STAT1 in DCs in vivo and in vitro. Genetic blockage of STAT1 abrogated the suppressive activity of CQ-treated DCs. Opposed to its WT counterparts, CQ-treated STAT1-/- BMDCs were unable to suppress Th17 cells and increased EAE severity. Our findings show that STAT1 is a major signaling pathway in CQ-induced tolDCs and may shed light on new therapeutic avenues for the induction of tolDCs in autoimmune diseases such as MS.
Collapse
Affiliation(s)
- Rodolfo Thome
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, Brazil.,Department of Neurology, Thomas Jefferson University, Philadelphia, USA
| | - Amanda Pires Bonfanti
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, Brazil
| | - Javad Rasouli
- Department of Neurology, Thomas Jefferson University, Philadelphia, USA
| | | | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, USA
| | | | - Liana Verinaud
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, Brazil
| |
Collapse
|
32
|
Cong Y, Hart BJ, Gross R, Zhou H, Frieman M, Bollinger L, Wada J, Hensley LE, Jahrling PB, Dyall J, Holbrook MR. MERS-CoV pathogenesis and antiviral efficacy of licensed drugs in human monocyte-derived antigen-presenting cells. PLoS One 2018; 13:e0194868. [PMID: 29566060 PMCID: PMC5864050 DOI: 10.1371/journal.pone.0194868] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 03/12/2018] [Indexed: 12/21/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) presents an emerging threat to public health worldwide by causing severe respiratory disease in humans with high virulence and case fatality rate (about 35%) since 2012. Little is known about the pathogenesis and innate antiviral response in primary human monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs) upon MERS-CoV infection. In this study, we assessed MERS-CoV replication as well as induction of inflammatory cytokines and chemokines in MDMs and immature and mature MDDCs. Immature MDDCs and MDMs were permissive for MERS-CoV infection, while mature MDDCs were not, with stimulation of proinflammatory cytokine and chemokine upregulation in MDMs, but not in MDDCs. To further evaluate the antiviral activity of well-defined drugs in primary antigen presenting cells (APCs), three compounds (chloroquine, chlorpromazine and toremifine), each with broad-spectrum antiviral activity in immortalized cell lines, were evaluated in MDMs and MDDCs to determine their antiviral effect on MERS-CoV infection. While chloroquine was not active in these primary cells, chlorpromazine showed strong anti-MERS-CoV activity, but it was associated with high cytotoxicity narrowing the potential window for drug utilization. Unlike in established cells, toremifene had marginal activity when tested in antigen presenting cells, with high apparent cytotoxicity, also limiting its potential as a therapeutic option. These results demonstrate the value of testing drugs in primary cells, in addition to established cell lines, before initiating preclinical or clinical studies for MERS treatment and the importance of carefully assessing cytotoxicity in drug screen assays. Furthermore, these studies also highlight the role of APCs in stimulating a robust protective immune response to MERS-CoV infection.
Collapse
Affiliation(s)
- Yu Cong
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Brit J. Hart
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Robin Gross
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Huanying Zhou
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Matthew Frieman
- Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Laura Bollinger
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Jiro Wada
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Lisa E. Hensley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Peter B. Jahrling
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
- Emerging Viral Pathogen Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Julie Dyall
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Michael R. Holbrook
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| |
Collapse
|
33
|
Umbilical cord-derived mesenchymal stem cells reversed the suppressive deficiency of T regulatory cells from peripheral blood of patients with multiple sclerosis in a co-culture - a preliminary study. Oncotarget 2018; 7:72537-72545. [PMID: 27705922 PMCID: PMC5341927 DOI: 10.18632/oncotarget.12345] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 09/12/2016] [Indexed: 01/01/2023] Open
Abstract
The immunoregulatory function of T regulatory cells (Tregs) is impaired in multiple sclerosis (MS). Recent studies have shown that umbilical cord-derived mesenchymal stem cells (UC-MSCs) exert regulatory effect on the functions of immune cells. Thus, we investigated whether UC-MSCs could improve the impaired function of Tregs from MS patients. Co-cultures of UC-MSCs with PBMCs of MS patients were performed for 3 days. Flow cytometry was used to determine the frequency of Tregs. A cell proliferation assay was used to evaluate the suppressive capacity of Tregs. ELISA was conducted for cytokine analysis in the co-cultures. Our results showed that UC-MSCs significantly increased the frequency of CD4+CD25+CD127low/− Tregs in resting CD4+ T cells (p<0.01) from MS, accompanied by the significantly augmented production of cytokine prostaglandin E2, transforming growth factor (−β1, and interleukin-10, along with a reduced interferon-γ production in these co-cultures (p<0.05 - 0.01). More importantly, UC-MSC-primed Tregs of MS patients significantly inhibited the proliferation of PHA-stimulated autologous and allogeneic CD4+CD25− T effector cells (Teffs) from MS patients and healthy individuals compared to non-UC-MSC-primed (naïve) Tregs from the same MS patients (p<0.01). Furthermore, no remarkable differences in suppressing the proliferation of PHA-stimulated CD4+CD25− Teffs was observed in UC-MSC-primed Tregs from MS patients and naïve Tregs from healthy subjects. The impaired suppressive function of Tregs from MS can be completely reversed in a co-culture by UC-MSC modulation. This report is the first to demonstrate that functional defects of Tregs in MS can be repaired in vitro using a simple UC-MSC priming approach.
Collapse
|
34
|
Tang TT, Lv LL, Pan MM, Wen Y, Wang B, Li ZL, Wu M, Wang FM, Crowley SD, Liu BC. Hydroxychloroquine attenuates renal ischemia/reperfusion injury by inhibiting cathepsin mediated NLRP3 inflammasome activation. Cell Death Dis 2018; 9:351. [PMID: 29500339 PMCID: PMC5834539 DOI: 10.1038/s41419-018-0378-3] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/28/2018] [Accepted: 02/01/2018] [Indexed: 12/27/2022]
Abstract
Inflammation is a major contributor to the pathogenesis of ischemic acute kidney injury (AKI), which complicates the post-operative outcomes of large numbers of hospitalized surgical patients. Hydroxychloroquine (HCQ), a well-known anti-malarial drug, is commonly used in clinical practice for its anti-inflammatory actions. However, little is known about its role in renal ischemia/reperfusion (I/R) injury. In the current study, mice were subjected to I/R injury and HCQ was administered for seven days by gavage prior to surgery. In parallel, HK-2 human renal proximal tubule cells were prophylactically treated with HCQ and then were exposed to hypoxia/reoxygenation (H/R). The results showed that HCQ significantly attenuated renal dysfunction evidenced by blunted decreases in serum creatinine and kidney injury molecular-1 expression and the improvement of HK-2 cell viability. Additionally, HCQ markedly reduced macrophage and neutrophil infiltration, pro-inflammatory cytokine production, and NLRP3 inflammasome activation. Mechanistic studies showed that HCQ could inhibit the priming of the NLRP3 inflammasome by down-regulating I/R or H/R-induced NF-κB signaling. Moreover, HCQ reduced cathepsin (CTS) B, CTSD and CTSL activity, and their redistribution from lysosomes to cytoplasm. CTSB and CTSL (not CTSD) were implicated in I/R triggered NLRP3 inflammasome activation. Notably, we found that HCQ attenuated renal injury through downregulation of CTSB and CTSL-mediated NLRP3 inflammasome activation. This study provides new insights into the anti-inflammatory effect of HCQ in the treatment of AKI.
Collapse
Affiliation(s)
- Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China.
| | - Ming-Ming Pan
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yi Wen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Min Wu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Feng-Mei Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Steve D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, NC, United States
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China.
| |
Collapse
|
35
|
Kim N, Park CS, Im SA, Kim JW, Lee JH, Park YJ, Song S, Lee CK. Minocycline promotes the generation of dendritic cells with regulatory properties. Oncotarget 2018; 7:52818-52831. [PMID: 27463004 PMCID: PMC5288151 DOI: 10.18632/oncotarget.10810] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 07/13/2016] [Indexed: 12/02/2022] Open
Abstract
Minocycline, which has long been used as a broad-spectrum antibiotic, also exhibits non-antibiotic properties such as inhibition of inflammation and angiogenesis. In this study, we show that minocycline significantly enhances the generation of dendritic cells (DCs) from mouse bone marrow (BM) cells when used together with GM-CSF and IL-4. DCs generated from BM cells in the presence of minocycline (Mino-DCs) demonstrate the characteristics of regulatory DCs. Compared with control DCs, Mino-DCs are resistant to subsequent maturation stimuli, impaired in MHC class II-restricted exogenous Ag presentation, and show decreased cytokine secretion. Mino-DCs also show decreased ability to prime allogeneic-specific T cells, while increasing the expansion of CD4+CD25+Foxp3+ T regulatory cells both in vitro and in vivo. In addition, pretreatment with MOG35-55 peptide-pulsed Mino-DCs ameliorates clinical signs of experimental autoimmune encephalitis induced by MOG peptide injection. Our study identifies minocycline as a new pharmacological agent that could be potentially used to increase the production of regulatory DCs for cell therapy to treat autoimmune disorders, allergy, and transplant rejection.
Collapse
Affiliation(s)
- Narae Kim
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Chan-Su Park
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Sun-A Im
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Ji-Wan Kim
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Jae-Hee Lee
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Young-Jun Park
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Sukgil Song
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| | - Chong-Kil Lee
- College of Pharmacy, Chungbuk National University, Cheongju, South Korea
| |
Collapse
|
36
|
Val F, Avalos S, Gomes AA, Zerpa JEA, Fontecha G, Siqueira AM, Bassat Q, Alecrim MGC, Monteiro WM, Lacerda MVG. Are respiratory complications of Plasmodium vivax malaria an underestimated problem? Malar J 2017; 16:495. [PMID: 29273053 PMCID: PMC5741897 DOI: 10.1186/s12936-017-2143-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/18/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Respiratory complications are uncommon, but often life-threatening features of Plasmodium vivax malaria. This study aimed to estimate the prevalence and lethality associated with such complications among P. vivax malaria patients in a tertiary hospital in the Western Brazilian Amazon, and to identify variables associated with severe respiratory complications, intensive care need and death. Medical records from 2009 to 2016 were reviewed aiming to identify all patients diagnosed with P. vivax malaria and respiratory complications. Prevalence, lethality and risk factors associated with WHO defined respiratory complications, intensive care need and death were assessed. RESULTS A total of 587 vivax malaria patients were hospitalized during the study period. Thirty (5.1%) developed respiratory complications. Thirteen (43.3%) developed severe respiratory complications, intensive care was required for 12 (40%) patients and 5 (16.6%) died. On admission, anaemia and thrombocytopaenia were common findings, whereas fever was unusual. Patients presented different classes of parasitaemia and six were aparasitaemic on admission. Time to respiratory complications occurred after anti-malarials administration in 18 (60%) patients and progressed very rapidly. Seventeen patients (56.7%) had comorbidities and/or concomitant conditions, which were significantly associated to higher odds of developing severe respiratory complications, need for intensive care and death (p < 0.05). CONCLUSION Respiratory complications were shown to be associated with significant mortality in this population. Patients with comorbidities and/or concomitant conditions require special attention to avoid this potential life-threatening complication.
Collapse
Affiliation(s)
- Fernando Val
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil. .,Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil.
| | - Sara Avalos
- Microbiology Research Institute, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - André Alexandre Gomes
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil.,Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - José Evelio Albornoz Zerpa
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil.,Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Gustavo Fontecha
- Microbiology Research Institute, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - André Machado Siqueira
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Quique Bassat
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain.,ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain.,Pediatric Infectious Diseases Unit, Pediatrics Department, Hospital Sant Joan de Déu (University of Barcelona), Barcelona, Spain.,Universidad Europea de Madrid, Madrid, Spain
| | - Maria Graças Costa Alecrim
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil.,Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil
| | - Wuelton Marcelo Monteiro
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil. .,Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil.
| | - Marcus Vinícius Guimarães Lacerda
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Amazonas, Brazil.,Universidade do Estado do Amazonas, Manaus, Amazonas, Brazil.,Instituto de Pesquisas Leônidas and Maria Deane, Fundação Oswaldo Cruz, Manaus, Amazonas, Brazil
| |
Collapse
|
37
|
Lee JH, Park CS, Jang S, Kim JW, Kim SH, Song S, Kim K, Lee CK. Tolerogenic dendritic cells are efficiently generated using minocycline and dexamethasone. Sci Rep 2017; 7:15087. [PMID: 29118423 PMCID: PMC5678112 DOI: 10.1038/s41598-017-15569-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 10/30/2017] [Indexed: 01/31/2023] Open
Abstract
Tolerogenic dendritic cells (tDCs) represent a promising tool for cellular therapy against autoimmune diseases, allergies, and transplantation rejection. Numerous pharmacological agents are known to induce tDC generation. Minocycline, which has long been used as a broad-spectrum antibiotic, was recently shown to significantly increase the generation of DCs with regulatory properties. Here, we examined the effect of the combination of minocycline with dexamethasone, rapamycin, vitamin D3, and interleukin (IL)-10, which are all known inducers of tDC generation. The highest number of tDCs was generated when minocycline and dexamethasone were used together with granulocyte colony-stimulating factor (GM-SCF) and IL-4. The tolerogenicity of the minocycline/dexamethasone-conditioned tDCs was much better than or at least equal to those of the tDCs generated with either one of these agents, as assessed through in vitro phenotypic and functional assays. In addition, pretreatment with MOG35-55 peptide-pulsed minocycline/dexamethasone-conditioned tDCs significantly ameliorated the clinical signs of experimental autoimmune encephalitis induced by MOG peptide injection in a murine model. These results confirmed that tDCs with potent tolerogenic properties could be efficiently generated by the combined use of minocycline and dexamethasone, along with GM-CSF and IL-4. Our results would help in the development of ex vivo tDC-based immunotherapies.
Collapse
Affiliation(s)
- Jae-Hee Lee
- College of Pharmacy, Chungbuk National University, Cheongju, 28644, South Korea
| | - Chan-Su Park
- College of Pharmacy, Chungbuk National University, Cheongju, 28644, South Korea
| | - Sundong Jang
- College of Pharmacy, Chungbuk National University, Cheongju, 28644, South Korea
| | - Ji-Wan Kim
- College of Pharmacy, Chungbuk National University, Cheongju, 28644, South Korea
| | - Sang-Hyeon Kim
- College of Pharmacy, Chungbuk National University, Cheongju, 28644, South Korea
| | - Sukgil Song
- College of Pharmacy, Chungbuk National University, Cheongju, 28644, South Korea
| | - Kyungjae Kim
- College of Pharmacy, Sahmyook University, Seoul, 01795, South Korea
| | - Chong-Kil Lee
- College of Pharmacy, Chungbuk National University, Cheongju, 28644, South Korea.
| |
Collapse
|
38
|
Samaras P, Tusup M, Nguyen-Kim TDL, Seifert B, Bachmann H, von Moos R, Knuth A, Pascolo S. Phase I study of a chloroquine-gemcitabine combination in patients with metastatic or unresectable pancreatic cancer. Cancer Chemother Pharmacol 2017; 80:1005-1012. [PMID: 28980060 DOI: 10.1007/s00280-017-3446-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 09/23/2017] [Indexed: 11/30/2022]
Abstract
PURPOSE Following a previously published pre-clinical validation, this phase I study evaluated the safety, maximum tolerated dose, anti-tumour activity and immune status of a gemcitabine-chloroquine combination as a first- or late-line treatment in patients with metastatic or unresectable pancreatic cancer. METHODS In this 3 + 3 dose escalation study, patients received a single weekly standard dose of intravenous gemcitabine, followed by single weekly oral intake of 100, 200 or 300 mg of chloroquine. Tumour response was assessed using the Response Evaluation Criteria in Solid Tumors version 1.1. Immune status was evaluated by RT-PCR to measure the relative expression of immune-related genes in peripheral blood mononuclear cells (PBMCs). RESULTS Overall, nine patients [median age 72 years; interquartile range (IQR), 68-78 years] were treated. No dose-limiting toxicities as defined in the protocol were observed. Three patients experienced partial response, and two patients had stable disease. The median time to progression was 4 months (95% CI 0.8-7.2), and the median overall survival was 7.6 months (95% CI 5.3-9.9). Among 86 assayed immune genes, three were significantly differentially expressed in PBMCs from responding versus non-responding patients: interferon-gamma receptor-1, toll-like receptor 2, and beta-2 microglobulin. CONCLUSIONS The addition of chloroquine to gemcitabine was well tolerated and showed promising effects on the clinical response to the anti-cancer chemotherapy. Based on these initial results, the efficacy of the gemcitabine-chloroquine combination should be further assessed.
Collapse
Affiliation(s)
- Panagiotis Samaras
- Department of Oncology, University Hospital of Zürich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Marina Tusup
- Department of Dermatology, University Hospital of Zürich, Gloriastrasse 31, 8091, Zurich, Switzerland
| | - Thi Dan Linh Nguyen-Kim
- Department of Diagnostic and Interventional Radiology, University Hospital of Zürich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Burkhardt Seifert
- Department of Biostatistics at the Epidemiology, Biostatistics and Prevention Institute, University of Zürich, Zurich, Switzerland
| | - Helga Bachmann
- Department of Oncology, University Hospital of Zürich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Roger von Moos
- Department of Oncology, University Hospital of Zürich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Alexander Knuth
- Department of Oncology, University Hospital of Zürich, Rämistrasse 100, 8091, Zurich, Switzerland.,National Center for Cancer Care and Research NCCCR, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Steve Pascolo
- Department of Oncology, University Hospital of Zürich, Rämistrasse 100, 8091, Zurich, Switzerland. .,Department of Dermatology, University Hospital of Zürich, Gloriastrasse 31, 8091, Zurich, Switzerland. .,Department of Dermatology, University Hospital of Zürich, Gloriastrasse 31, 8091, Zurich, Switzerland.
| |
Collapse
|
39
|
Masuelli L, Granato M, Benvenuto M, Mattera R, Bernardini R, Mattei M, d'Amati G, D'Orazi G, Faggioni A, Bei R, Cirone M. Chloroquine supplementation increases the cytotoxic effect of curcumin against Her2/neu overexpressing breast cancer cells in vitro and in vivo in nude mice while counteracts it in immune competent mice. Oncoimmunology 2017; 6:e1356151. [PMID: 29147611 DOI: 10.1080/2162402x.2017.1356151] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/05/2017] [Accepted: 07/07/2017] [Indexed: 01/21/2023] Open
Abstract
Autophagy is usually a pro-survival mechanism in cancer cells, especially in the course of chemotherapy, thus autophagy inhibition may enhance the chemotherapy-mediated anti-cancer effect. However, since autophagy is strongly involved in the immunogenicity of cell death by promoting ATP release, its inhibition may reduce the immune response against tumors, negatively influencing the overall outcome of chemotherapy. In this study, we evaluated the in vitro and in vivo anti-cancer effect of curcumin (CUR) against Her2/neu overexpressing breast cancer cells (TUBO) in the presence or in the absence of the autophagy inhibitor chloroquine (CQ). We found that TUBO cell death induced by CUR was increased in vitro by CQ and slightly in vivo in nude mice. Conversely, CQ counteracted the Cur cytotoxic effect in immune competent mice, as demonstrated by the lack of in vivo tumor regression and the reduction of overall mice survival as compared with CUR-treated mice. Immunohistochemistry analysis revealed the presence of a remarkable FoxP3 T cell infiltrate within the tumors in CUR/CQ treated mice and a reduction of T cytotoxic cells, as compared with single CUR treatment. These findings suggest that autophagy is important to elicit anti-tumor immune response and that autophagy inhibition by CQ reduces such response also by recruiting T regulatory (Treg) cells in the tumor microenvironment that may be pro-tumorigenic and might counteract CUR-mediated anti-cancer effects.
Collapse
Affiliation(s)
- L Masuelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - M Granato
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - M Benvenuto
- Department of Clinical Sciences and Traslational Medicine, "Tor Vergata" University of Rome, Rome, Italy
| | - R Mattera
- Department of Clinical Sciences and Traslational Medicine, "Tor Vergata" University of Rome, Rome, Italy
| | - R Bernardini
- STA, "Tor Vergata" University of Rome, Rome, Italy
| | - M Mattei
- STA, "Tor Vergata" University of Rome, Rome, Italy
| | - G d'Amati
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy
| | - G D'Orazi
- Department of Research, Advanced Diagnostics, and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical, Oral and Biotechnological Sciences, Tumor Biology Section, University 'G. d'Annunzio', Chieti, Italy
| | - A Faggioni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - R Bei
- Department of Clinical Sciences and Traslational Medicine, "Tor Vergata" University of Rome, Rome, Italy
| | - M Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
40
|
Lu JQ, Ringrose J, Gross D, Emery D, Blevins G, Power C. Multifocal inflammatory demyelination in a patient with rheumatoid arthritis and treatment complications. J Neurol Sci 2016; 367:305-7. [DOI: 10.1016/j.jns.2016.06.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/25/2016] [Accepted: 06/15/2016] [Indexed: 10/21/2022]
|
41
|
Thomé R, de Carvalho AC, Alves da Costa T, Ishikawa LLW, Fraga-Silva TFDC, Sartori A, de Oliveira ALR, Verinaud L. Artesunate Ameliorates Experimental Autoimmune Encephalomyelitis by Inhibiting Leukocyte Migration to the Central Nervous System. CNS Neurosci Ther 2016; 22:707-14. [PMID: 27165523 DOI: 10.1111/cns.12561] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/14/2016] [Accepted: 04/17/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND AIMS Experimental autoimmune encephalomyelitis (EAE) is T-cell-dependent disease of the central nervous system (CNS) of mice. This model resembles multiple sclerosis (MS) in many aspects. Therapies that focus in the modulation of the immune response and cellular infiltration in the CNS present best effects in the clinics. Artesunate (Art) is a semi-synthetic sesquiterpene derivative from artemisinin and has been shown to reduce the clinical signs of autoimmune disease models through mechanisms not yet understood. In this study, we aimed to evaluate whether administration of Art would ameliorate EAE. METHODS AND RESULTS C57BL6 mice were immunized with MOG35-55 peptide to induce EAE. At the same time, Art treatment started (3 mg/kg/day via i.p.) for five consecutive days. We found that Art treatment reduced the clinical signs of EAE and that correlated with a reduced infiltration of cells in the CNS. Disease amelioration did not correlate with immunomodulation as recall responses, leukocyte subpopulations, and gene expression analysis were similar among treated and untreated mice. Ultimately, further analysis provided data indicating that a possible mechanism of action for Art is dependent on the cellular migration to the CNS. CONCLUSIONS Artesunate reduces the severity of EAE by inhibiting migration of pathogenic T cells to the CNS.
Collapse
Affiliation(s)
- Rodolfo Thomé
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Ana Carolina de Carvalho
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Thiago Alves da Costa
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Larissa Lumi Watanabe Ishikawa
- Department of Immunology and Microbiology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, Brazil
| | | | - Alexandrina Sartori
- Department of Immunology and Microbiology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, Brazil
| | | | - Liana Verinaud
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| |
Collapse
|
42
|
Time to use a dose of Chloroquine as an adjuvant to anti-cancer chemotherapies. Eur J Pharmacol 2015; 771:139-44. [PMID: 26687632 DOI: 10.1016/j.ejphar.2015.12.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/27/2015] [Accepted: 12/09/2015] [Indexed: 01/05/2023]
Abstract
Chloroquine, a drug used for over 80 years to treat and prevent malaria and, more recently, to treat autoimmune diseases, is very safe but has a plethora of dose-dependent effects. By increasing pH in acidic compartments it inhibits for example lysosomal enzymes. In the context of cancer, Chloroquine was found to have direct effects on different types of malignancies that could potentiate chemotherapies. For example, the anti-malaria drug may inhibit both the multidrug-resistance pump and autophagy (mechanisms that tumor cells may use to resist chemotherapies), intercalate in DNA and enhance the penetration of chemotherapeutic drugs in cells or solid cancer tissues. However, these activities were mostly demonstrated at high doses of Chloroquine (higher than 10mg/kg or 10mg/l i.e. ca. 31μM). Nevertheless, it was reported that daily uptake of clinically acceptable doses (less than 10mg/kg) of Chloroquine in addition to chemo-radio-therapy increases the survival of glioblastoma patients (Sotelo et al., 2006; Briceno et al., 2007). However, the optimal dose and schedule of this multi-active drug with respect to chemotherapy has never been experimentally determined. The present article reviews the several known direct and indirect effects of different doses of Chloroquine on cancer and how those effects may indicate that a fine tuning of the dose/schedule of Chloroquine administration versus chemotherapy may be critical to obtain an adjuvant effect of Chloroquine in anti-cancer treatments. We anticipate that the appropriate (time and dose) addition of Chloroquine to the standard of care may greatly and safely potentiate current anti-cancer treatments.
Collapse
|
43
|
Zanucoli F, Thomé R, Bonfanti AP, de Carvalho AC, Issayama LK, da Costa TA, Di Gangi R, Ferreira IT, Bombeiro AL, de Oliveira ALR, Verinaud L. Primaquine treatment suppresses experimental autoimmune encephalomyelitis severity. CNS Neurosci Ther 2015; 20:1061-4. [PMID: 25417930 DOI: 10.1111/cns.12357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 12/24/2022] Open
Affiliation(s)
- Fábio Zanucoli
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lin NY, Chen CW, Kagwiria R, Liang R, Beyer C, Distler A, Luther J, Engelke K, Schett G, Distler JH. Inactivation of autophagy ameliorates glucocorticoid-induced and ovariectomy-induced bone loss. Ann Rheum Dis 2015; 75:1203-10. [PMID: 26113650 DOI: 10.1136/annrheumdis-2015-207240] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 06/09/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Autophagy has recently been shown to regulate osteoclast activity and osteoclast differentiation. Here, we aim to investigate the impact of autophagy inhibition as a potential therapeutic approach for the treatment of osteoporosis in preclinical models. METHODS Systemic bone loss was induced in mice by glucocorticoids and by ovariectomy (OVX). Autophagy was targeted by conditional inactivation of autophagy-related gene 7 (Atg7) and by treatment with chloroquine (CQ). Bone density was evaluated by microCT. The role of autophagy on osteoclastogenesis was analysed by osteoclastogenesis and bone resorption assays. The quantification of receptor activator of nuclear factor κ B ligand and osteoprotegerin proteins in cocultures was performed using ELISA whereas that of osteoclast and osteoblast differentiation markers was by qPCR. RESULTS Selective deletion of Atg7 in monocytes from Atg7(fl/fl)_x_LysM-Cre mice mitigated glucocorticoid-induced and OVX-induced osteoclast differentiation and bone loss compared with Atg7(fl/fl) littermates. Pharmacological inhibition of autophagy by treatment with CQ suppressed glucocorticoid-induced osteoclastogenesis and protected mice from bone loss. Similarly, inactivation of autophagy shielded mice from OVX-induced bone loss. Inhibition of autophagy led to decreased osteoclast differentiation with lower expression of osteoclast markers such as NFATc1, tartrate-resistant acid phosphatase, OSCAR and cathepsin K and attenuated bone resorption in vitro. In contrast, osteoblast differentiation was not affected by inhibition of autophagy. CONCLUSIONS Pharmacological or genetic inactivation of autophagy ameliorated glucocorticoid-induced and OVX-induced bone loss by inhibiting osteoclastogenesis. These findings may have direct translational implications for the treatment of osteoporosis, since inhibitors of autophagy such as CQ are already in clinical use.
Collapse
Affiliation(s)
- Neng-Yu Lin
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Chih-Wei Chen
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Rosebeth Kagwiria
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ruifang Liang
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Christian Beyer
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alfiya Distler
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Julia Luther
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Klaus Engelke
- Institute of Medical Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Jörg Hw Distler
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
45
|
Violacein Treatment Modulates Acute and Chronic Inflammation through the Suppression of Cytokine Production and Induction of Regulatory T Cells. PLoS One 2015; 10:e0125409. [PMID: 25938431 PMCID: PMC4418714 DOI: 10.1371/journal.pone.0125409] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/23/2015] [Indexed: 12/20/2022] Open
Abstract
Inflammation is a necessary process to control infection. However, exacerbated inflammation, acute or chronic, promotes deleterious effects in the organism. Violacein (viola), a quorum sensing metabolite from the Gram-negative bacterium Chromobacterium violaceum, has been shown to protect mice from malaria and to have beneficial effects on tumors. However, it is not known whether this drug possesses anti-inflammatory activity. In this study, we investigated whether viola administration is able to reduce acute and chronic autoimmune inflammation. For that purpose, C57BL/6 mice were intraperitoneally injected with 1 μg of LPS and were treated with viola (3.5mg/kg) via i.p. at the same time-point. Three hours later, the levels of inflammatory cytokines in the sera and phenotypical characterization of leukocytes were determined. Mice treated with viola presented a significant reduction in the production of inflammatory cytokines compared with untreated mice. Interestingly, although viola is a compound derived from bacteria, it did not induce inflammation upon administration to naïve mice. To test whether viola would protect mice from an autoimmune inflammation, Experimental Autoimmune Encephalomyelitis (EAE)-inflicted mice were given viola i.p. at disease onset, at the 10th day from immunization. Viola-treated mice developed mild EAE disease in contrast with placebo-treated mice. The frequencies of dendritic cells and macrophages were unaltered in EAE mice treated with viola. However, the sole administration of viola augmented the levels of splenic regulatory T cells (CD4+Foxp3+). We also found that adoptive transfer of viola-elicited regulatory T cells significantly reduced EAE. Our study shows, for the first time, that violacein is able to modulate acute and chronic inflammation. Amelioration relied in suppression of cytokine production (in acute inflammation) and stimulation of regulatory T cells (in chronic inflammation). New studies must be conducted in order to assess the possible use of viola in therapeutic approaches in human autoimmune diseases.
Collapse
|
46
|
Dalhoff A. Antiviral, antifungal, and antiparasitic activities of fluoroquinolones optimized for treatment of bacterial infections: a puzzling paradox or a logical consequence of their mode of action? Eur J Clin Microbiol Infect Dis 2015; 34:661-8. [PMID: 25515946 PMCID: PMC7087824 DOI: 10.1007/s10096-014-2296-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/02/2014] [Indexed: 12/19/2022]
Abstract
This review summarizes evidence that commercially available fluoroquinolones used for the treatment of bacterial infections are active against other non-bacterial infectious agents as well. Any of these fluoroquinolones exerts, in parallel to its antibacterial action, antiviral, antifungal, and antiparasitic actions at clinically achievable concentrations. This broad range of anti-infective activities is due to one common mode of action, i.e., the inhibition of type II topoisomerases or inhibition of viral helicases, thus maintaining the selective toxicity of fluoroquinolones inhibiting microbial topoisomerases at low concentrations but mammalian topoisomerases at much higher concentrations. Evidence suggests that standard doses of the fluoroquinolones studied are clinically effective against viral and parasitic infections, whereas higher doses administered topically were active against Candida spp. causing ophthalmological infections. Well-designed clinical studies should be performed to substantiate these findings.
Collapse
Affiliation(s)
- A Dalhoff
- Institute for Infection Medicine, University Medical Center Schleswig-Holstein, Brunswiker Str. 4, 24105, Kiel, Germany,
| |
Collapse
|
47
|
Medrano-Campillo P, Sarmiento-Soto H, Álvarez-Sánchez N, Álvarez-Ríos AI, Guerrero JM, Rodríguez-Prieto I, Castillo-Palma MJ, Lardone PJ, Carrillo-Vico A. Evaluation of the immunomodulatory effect of melatonin on the T-cell response in peripheral blood from systemic lupus erythematosus patients. J Pineal Res 2015; 58:219-26. [PMID: 25612066 DOI: 10.1111/jpi.12208] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/15/2015] [Indexed: 12/23/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by the production of antinuclear autoantibodies. In addition, the involvement of CD4+ T-helper (Th) cells in SLE has become increasingly evident. Although the role of melatonin has been tested in some experimental models of lupus with inconclusive results, there are no studies evaluating the melatonin effect on cells from patients with SLE. Therefore, the aim of this study was to analyse the role of in vitro administered melatonin in the immune response of peripheral leukocytes from treated patients with SLE (n = 20) and age- and sex-matched healthy controls. Melatonin was tested for its effect on the production of key Th1, Th2, Th9, Th17 and innate cytokines. The frequency of T regulatory (Treg) cells and the expression of FOXP3 and BAFF were also explored. Our results are the first to show that melatonin decreased the production of IL-5 and to describe the novel role of melatonin in IL-9 production by human circulating cells. Additionally, we highlighted a two-faceted melatonin effect. Although it acted as a prototypical anti-inflammatory compound, reducing exacerbated Th1 and innate responses in PHA-stimulated cells from healthy subjects, it caused the opposite actions in immune-depressed cells from patients with SLE. Melatonin also increased the number of Treg cells expressing FOXP3 and offset BAFF overexpression in SLE patient cells. These findings open a new field of research in lupus that could lead to the use of melatonin as treatment or cotreatment for SLE.
Collapse
Affiliation(s)
- Pablo Medrano-Campillo
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital/CSIC/University of Seville, Seville, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Dey S, Bishayi B. Killing of Staphylococcus aureus in murine macrophages by chloroquine used alone and in combination with ciprofloxacin or azithromycin. J Inflamm Res 2015; 8:29-47. [PMID: 25653549 PMCID: PMC4309780 DOI: 10.2147/jir.s76045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This study aimed to determine any alteration in the killing of Staphylococcus aureus in murine peritoneal macrophages when chloroquine (CQ) is used alone compared with when it is used in combination with ciprofloxacin (CIP) or azithromycin (AZM). The study also aimed to find out the implication of reactive oxygen species (ROS) production and cytokine release in the intracellular killing of S. aureus in macrophages. We present here data obtained with a model of S. aureus-infected mouse peritoneal macrophages in which the intracellular growth of the bacteria and the influence of antibiotics was monitored for 30, 60, and 90 minutes in the presence or absence of CQ along with the production of ROS and alteration in levels of antioxidant enzymes and cytokines. It was observed that S. aureus-triggered cytokine response was regulated when macrophages were co-cultured with CQ and AZM as compared with CQ stimulation only. It can be suggested that action of AZM in mediating bacterial killing is enhanced by the presence of CQ, indicating enhanced uptake of AZM during early infection that may be essential for bacteria killing by AZM. Reduction of oxidative stress burden on the S. aureus-infected macrophages may pave the way for better killing of internalized S. aureus by CQ plus ciprofloxacin (CIP) or CQ plus AZM. Based on these observations, one may speculate that in an inflammatory milieu, CQ loaded with AZM elicits a stronger proinflammatory response by increasing the intracellular uptake of AZM or CIP, thus enabling the immune system to mount a more robust and prolonged response against intracellular pathogens.
Collapse
Affiliation(s)
- Somrita Dey
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, India
| |
Collapse
|
49
|
Thomé R, Issayama LK, Alves da Costa T, Gangi RD, Ferreira IT, Rapôso C, Lopes SCP, da Cruz Höfling MA, Costa FTM, Verinaud L. Dendritic cells treated with crude Plasmodium berghei extracts acquire immune-modulatory properties and suppress the development of autoimmune neuroinflammation. Immunology 2014; 143:164-73. [PMID: 24689455 DOI: 10.1111/imm.12298] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 01/02/2023] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells specifically targeted during Plasmodium infection. Upon infection, DCs show impaired antigen presentation and T-cell activation abilities. In this study, we aimed to evaluate whether cellular extracts obtained from Plasmodium berghei-infected erythrocytes (PbX) modulate DCs phenotypically and functionally and the potential therapeutic usage of PbX-modulated DCs in the control of experimental autoimmune encephalomyelitis (EAE, the mouse model for human multiple sclerosis). We found that PbX-treated DCs have impaired maturation and stimulated the generation of regulatory T cells when cultured with naive T lymphocytes in vitro. When adoptively transferred to C57BL/6 mice the EAE severity was reduced. Disease amelioration correlated with a diminished infiltration of cytokine-producing T cells in the central nervous system as well as the suppression of encephalitogenic T cells. Our study shows that extracts obtained from P. berghei-infected erythrocytes modulate DCs towards an immunosuppressive phenotype. In addition, the adoptive transfer of PbX-modulated DCs was able to ameliorate EAE development through the suppression of specific cellular immune responses towards neuro-antigens. To our knowledge, this is the first study to present evidence that DCs treated with P. berghei extracts are able to control autoimmune neuroinflammation.
Collapse
Affiliation(s)
- Rodolfo Thomé
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Thomé R, Bombeiro AL, Issayama LK, Rapôso C, Lopes SCP, da Costa TA, Di Gangi R, Ferreira IT, Longhini ALF, Oliveira ALR, da Cruz Höfling MA, Costa FTM, Verinaud L. Exacerbation of autoimmune neuro-inflammation in mice cured from blood-stage Plasmodium berghei infection. PLoS One 2014; 9:e110739. [PMID: 25329161 PMCID: PMC4201583 DOI: 10.1371/journal.pone.0110739] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 09/16/2014] [Indexed: 01/24/2023] Open
Abstract
The thymus plays an important role shaping the T cell repertoire in the periphery, partly, through the elimination of inflammatory auto-reactive cells. It has been shown that, during Plasmodium berghei infection, the thymus is rendered atrophic by the premature egress of CD4+CD8+ double-positive (DP) T cells to the periphery. To investigate whether autoimmune diseases are affected after Plasmodium berghei NK65 infection, we immunized C57BL/6 mice, which was previously infected with P. berghei NK65 and treated with chloroquine (CQ), with MOG35-55 peptide and the clinical course of Experimental Autoimmune Encephalomyelitis (EAE) was evaluated. Our results showed that NK65+CQ+EAE mice developed a more severe disease than control EAE mice. The same pattern of disease severity was observed in MOG35-55-immunized mice after adoptive transfer of P. berghei-elicited splenic DP-T cells. The higher frequency of IL-17+- and IFN-γ+-producing DP lymphocytes in the Central Nervous System of these mice suggests that immature lymphocytes contribute to disease worsening. To our knowledge, this is the first study to integrate the possible relationship between malaria and multiple sclerosis through the contribution of the thymus. Notwithstanding, further studies must be conducted to assert the relevance of malaria-induced thymic atrophy in the susceptibility and clinical course of other inflammatory autoimmune diseases.
Collapse
Affiliation(s)
- Rodolfo Thomé
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - André Luis Bombeiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Luidy Kazuo Issayama
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Catarina Rapôso
- Department of Histology and Embryology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Stefanie Costa Pinto Lopes
- Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Thiago Alves da Costa
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Rosária Di Gangi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Isadora Tassinari Ferreira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | | | | | | | | | - Liana Verinaud
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| |
Collapse
|