1
|
Gilfillan MA, Kiladejo A, Bhandari V. Current and Emerging Therapies for Prevention and Treatment of Bronchopulmonary Dysplasia in Preterm Infants. Paediatr Drugs 2025:10.1007/s40272-025-00697-3. [PMID: 40374983 DOI: 10.1007/s40272-025-00697-3] [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] [Accepted: 04/13/2025] [Indexed: 05/18/2025]
Abstract
Although advances in the care of extremely preterm born infants have yielded improvements in survival and reductions in important morbidities, rates of bronchopulmonary dysplasia (BPD) have remained relatively unchanged. As BPD can have a long-lasting impact on the quality of life for survivors of prematurity and their families, this remains a continuing challenge. Treatments that have consistently shown efficacy in preventing either BPD or the composite outcome of BPD and death prior to 36 weeks post menstrual age (PMA) in large-scale randomized clinical trials (RCTs) include caffeine [adjusted odds ratio aOR for BPD, 0.63; 95% confidence interval (95% CI) 0.52-0.76; p < 0.001)], vitamin A [relative risk (RR) for death or BPD 0.89; 95% CI 0.80-0.99], low-dose hydrocortisone in the first week of life [OR for survival without BPD, 1.45; 95% CI 1.11-1.90; p = 0.007], and post-natal dexamethasone [RR for BPD or mortality; 0.76; 95% CI 0.66-0.87]. Although early caffeine therapy is now a widely used strategy to prevent BPD, the potentially severe side effects of post-natal glucocorticoids and the concerns regarding the cost-benefit of vitamin A have led to inconsistent use of these drugs in clinical practice. Inhaled bronchodilators and diuretics provide differing degrees of symptomatic relief for patients according to their phenotypic pattern of lung injury; however, these medications do not prevent BPD. Currently available pharmaceuticals do not sufficiently address the degree of structural immaturity and immune dysregulation that is present in the growing population of survivors born prior to 25 weeks gestational age. In this article, we provide both an evidence-based summary of pharmacological treatments currently available to prevent and manage BPD and a discussion of emerging therapies that could help preserve normal lung development in infants born preterm.
Collapse
Affiliation(s)
- Margaret A Gilfillan
- Department of Pediatrics, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, PA, USA
| | - Adedapo Kiladejo
- Department of Pediatrics, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, PA, USA
| | - Vineet Bhandari
- Division of Neonatology, Department of Pediatrics, The Children's Regional Hospital at Cooper/Cooper Medical School of Rowan University, One Cooper Plaza, Camden, NJ, 08103, USA.
| |
Collapse
|
2
|
Huang YJ, Wang LC, Wang CP, Yu KH, Kuo CF. Non-inflammatory macrophages phagocytose and hydrolyse monosodium urate crystals in different stages of gout. Scand J Rheumatol 2025:1-10. [PMID: 40338022 DOI: 10.1080/03009742.2025.2491176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/07/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVE Macrophages play a crucial role in gouty arthritis; however, the relationship between non-inflammatory macrophages (M0) and different stages of gout remains unclear. This study aimed to investigate the phagocytosis, hydrolysis, and subsequent cytokine secretion of monosodium urate (MSU) by non-inflammatory macrophages in patients in different stages of gout. METHOD Non-inflammatory macrophages were derived from monocytes through stimulation with macrophage colony-stimulating factor (M-CSF) for a duration of 10 days. The study included patients with asymptomatic hyperuricaemia, intercritical gout, tophaceous gout, and a normal control group. The phagocytic and hydrolytic capabilities of non-inflammatory macrophages were measured using flow cytometry based on the increase in side-scatter area. In addition, to evaluate the relationship between the hydrolysis capability of non-inflammatory macrophages and subsequent inflammation, we cultured them with lipopolysaccharide (LPS) and/or MSU. RESULTS We discovered that M0 macrophages were capable of phagocytosing and hydrolysing MSU crystals in various stages of gout, including the control group. Patients with asymptomatic hyperuricaemia exhibited the most pronounced phagocytic and hydrolytic capabilities, surpassing even those of the normal control group. The presence of MSU alone did not induce the secretion of pro-inflammatory cytokines. However, in experiments where M0 macrophages were stimulated with LPS and/or MSU, the phagocytic and hydrolytic abilities of M0 macrophages were correlated with inflammatory cytokine elevation. CONCLUSION The efficient phagocytosis and hydrolysis of MSU crystals by M0 macrophages suggest their role in maintaining the non-inflammatory stage of gout. Our findings suggest that non-inflammatory macrophages play a role in gout.
Collapse
Affiliation(s)
- Y-J Huang
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - L-C Wang
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - C-P Wang
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - K-H Yu
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - C-F Kuo
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
3
|
Talukdar D, Haldar AK, Kumar S, Dastidar R, Basu A, Roy A, Sarkar S, Dey S, Sikder K. Leukocyte infiltration and cross-talk with cardiomyocytes exploit intracellular stress pathways in dilated cardiomyopathy of idiopathic origin. Mol Biol Rep 2024; 51:1090. [PMID: 39446238 DOI: 10.1007/s11033-024-10028-3] [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: 07/20/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND OBJECTIVE Dilated cardiomyopathy (DCM) is a prevalent form of heart failure results in dilation and disruption of heart. Most strikingly a majority of the DCM cases do not have any identified etiology, hence known as idiopathic DCM (IDCM). Our study aimed to investigate the cross-talk between leukocytes and cardiomyocytes in terms of cardiac inflammation and stress response in IDCM. METHODS 60 IDCM patients and 60 age and sex matched healthy volunteers were recruited in this study based on the New York Heart Association (NYHA) guidelines. Their echocardiographic and biochemical markers were assessed and PBMCs were analyzed for leukocyte migration and inflammation. Also C2C12 myocyte cells were cultured with LPS-activated RAW264.7 monocytes to investigate the cross-talk between them. RESULTS Left ventricular (LV) dysfunction was evident in the IDCM patients which were correlated with their physical discomfort level according to NYHA classification. Their serum levels of IL-1β and TNF-α (≈ 20 pg/ml) were found to be very high along with hs-CRP and IL-2. Elevated levels of ROCK, SMA and ICAM-1 proteins indicated activation and migration of the leukocytes. During monocyte-myocyte co-culture, robust diapedesis was observed in the cultured macrophage cells towards myocytes through the transwell pores (8 µM) in presence of IL-1β and TNF-α causing ER stress and cell death in the myocytes. Inhibition of this migration or by alleviating ER stress inhibits leukocyte recruitment and ensures protection to the myocytes. CONCLUSION The present study showed that alleviating cellular stress and managing leukocyte migration promotes protection to the heart.
Collapse
Affiliation(s)
- Debdatta Talukdar
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), 99 Sarat Bose Road, Kolkata, West Bengal, 700026, India
| | - Akash Kumar Haldar
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), 99 Sarat Bose Road, Kolkata, West Bengal, 700026, India
| | - Soumitra Kumar
- Department of Cardiology, Ramakrishna Mission Seva Pratishthan Vivekananda Institute of Medical Sciences (RKMSP VIMS), 99 Sarat Bose Road, Kolkata, West Bengal, 700026, India
| | - Rinini Dastidar
- Department of Clinical Biochemistry and Immunology, Nirnayan Health Care Private Limited, 145, Rajarhat Main Rd, Zarda Bagan, Jyangra, Rajarhat, Kolkata, West Bengal, 700059, India
| | - Arnab Basu
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), 99 Sarat Bose Road, Kolkata, West Bengal, 700026, India
| | - Ajitesh Roy
- Department of Endocrinology and Metabolism, Ramakrishna Mission Seva Pratishthan Vivekananda Institute of Medical Sciences (RKMSP VIMS), 99 Sarat Bose Road, Kolkata, West Bengal, 700026, India
| | - Sankalita Sarkar
- Department of Physiology, Department of Sports Sciences, UGC -Centre for Research in Nano sciences and Nanotechnology (CRNN) UGC - Centre with Potential for Excellence in Particular Area (CPEPA), University of Calcutta, 92 A.P.C. Road, Kolkata, West Bengal, 700009, India
| | - Sanjit Dey
- Department of Physiology, Department of Sports Sciences, UGC -Centre for Research in Nano sciences and Nanotechnology (CRNN) UGC - Centre with Potential for Excellence in Particular Area (CPEPA), University of Calcutta, 92 A.P.C. Road, Kolkata, West Bengal, 700009, India
| | - Kunal Sikder
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), 99 Sarat Bose Road, Kolkata, West Bengal, 700026, India.
- Department of Sports Science and Yoga, School of Indian Heritage, Ramakrishna Mission Vivekananda Educational and Research Institute, Belur Math, Howrah, West Bengal, 711202, India.
| |
Collapse
|
4
|
Dayoub AS, Acharya E, Dibas A, Jones HP, Acharya S. Novel Small Molecules with Anti-Inflammatory and Anti-Angiogenic Activity in a Mouse Model of Oxygen-Induced Retinopathy. Cells 2024; 13:1371. [PMID: 39195259 PMCID: PMC11353024 DOI: 10.3390/cells13161371] [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: 07/17/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Retinopathy of prematurity (ROP) has a dual-phase disease pathology; in phase 1, hyperoxia-induced vaso-obliteration occurs in the retinal vasculature due to increased oxidative stress (OS) and inflammation, followed by phase 2, where hypoxia increases the overproduction of growth factors, inducing retinal neovascularization. Toll-like receptor 2 and -4 (TLR2 and TLR4) overactivation, hyper-inflammation, macrophages, and neutrophil infiltration contribute to the developing ROP. AVR-121 and AVR-123 are novel classes of small-molecule dual inhibitors of TLR2/4 tested in a human leukemia monocytic cell line (THP-1) and cord-blood-derived mononuclear cells (CBMCs). Both compounds inhibited TLR2/4 signaling-related inflammatory cytokines in THP-1 cells and inhibited VEGF-induced neovascularization in human retinal endothelial cells (HRECs), which are hallmarks of ROP. In an oxygen-induced retinopathy (OIR) murine model, the intraperitoneal injection of AVR-123 in the hyperoxia phase (P7-P12) or a nanosuspension eyedrop of AVR-123 in the hypoxic phase (P12-P17) significantly reduced vaso-obliteration, angiogenesis, and inflammatory cytokine profiles while not inhibiting the necessary growth factor VEGF in the juvenile mouse eyes. The results are consistent with our hypothesis that targeting the dual TLR2/4 pathway will reduce inflammation, angiogenesis, and vaso-obliteration in vitro and in vivo and reduce cytotoxic immune cells. AVR-123 has the potential to be developed as a therapy for ROP.
Collapse
Affiliation(s)
- Adam S. Dayoub
- AyuVis Research Inc., Fort Worth, TX 76107, USA; (A.S.D.)
| | - Eesha Acharya
- AyuVis Research Inc., Fort Worth, TX 76107, USA; (A.S.D.)
| | - Adnan Dibas
- The North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Harlan P. Jones
- The North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Suchismita Acharya
- AyuVis Research Inc., Fort Worth, TX 76107, USA; (A.S.D.)
- The North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
5
|
Jabeen K, Khlaid M, Mansoor S, Zalan A, Ejaz M, Mansoor A, Javed A. Host immune players and their response to Hepatitis C therapies. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003110. [PMID: 38865413 PMCID: PMC11168669 DOI: 10.1371/journal.pgph.0003110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/22/2024] [Indexed: 06/14/2024]
Abstract
This study aimed to investigate alterations in the expression of four key cytokines (IL-7, IL-11, IL-15, and IL-27) and assess differential FAM26F expression in response to Hepatitis C virus (HCV) infection. Additionally, it sought to analyze changes in these cytokines after treatment in 244 chronic HCV patients and 28 controls undergoing various treatments, including standard interferon, pegylated interferon, and Direct Acting Antivirals (DAAs). The objective was to compare immune system regulation between treatment groups. The expression levels of FAM26F and the cytokines (IL-7, IL-11, IL-15, and IL-27) were evaluated using Real-time qPCR in PBMCs of treatment groups. Results revealed significant downregulation of IL-7 and IL-27 in infected individuals compared to healthy controls, persisting even after treatment. This suggests the crucial roles of these immune modulators in facilitating the necessary T-cell response for viral clearance. IL-11 expression also remained suppressed post-treatment, supporting viral clearance by restoring the Th1 response. The decrease in IL-11 levels during treatment indicates the restoration of the Th1 response, vital for viral clearance. IL-15, the key cytokine regulating cytotoxic cells (NKT and NK cells), displayed consistent expression across all sample groups, indicating maintained IL-15-induced cytotoxicity in both control and infected individuals. Additionally, FAM26F expression was reduced in the HCV-infected group compared to controls, but higher in HCV-recovered cases, potentially due to reduced infection and enhanced immunity. In conclusion, this research unveils the relationship between FAM26F and HCV infection, highlighting the virus's tendency to suppress cytokine and FAM26F expression. An effective treatment strategy for establishing an ideal host immune response may involve restoring FAM26F and cytokine expression to their normal levels.
Collapse
Affiliation(s)
- Kehkshan Jabeen
- Genomics Research Lab, Department of Biological Sciences, International Islamic University Islamabad, Islamabad, Pakistan
- Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Madiha Khlaid
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Sajid Mansoor
- University of Central Punjab (UCP), Lahore, Punjab, Pakistan
| | - Ali Zalan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Momina Ejaz
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Atika Mansoor
- Institute of Biomedical Genetic Engineering (IBGE), Islamabad, Pakistan
| | - Aneela Javed
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| |
Collapse
|
6
|
Valdes-Fernandez BN, Ruiz-Jimenez C, Armina-Rodriguez A, Mendez LB, Espino AM. Fasciola hepatica GST mu-class suppresses the cytokine storm induced by E. coli-lipopolysaccharide, whereas it modulates the dynamic of peritoneal macrophages in a mouse model and suppresses the classical activation of macrophages. Microbiol Spectr 2024; 12:e0347523. [PMID: 38018982 PMCID: PMC10782955 DOI: 10.1128/spectrum.03475-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 10/12/2023] [Indexed: 11/30/2023] Open
Abstract
IMPORTANCE Sepsis is the consequence of a systemic bacterial infection that exacerbates the immune cell's activation via bacterial products, resulting in the augmented release of inflammatory mediators. A critical factor in the pathogenesis of sepsis is the primary component of the outer membrane of Gram-negative bacteria known as lipopolysaccharide (LPS), which is sensed by TLR4. For this reason, scientists have aimed to develop antagonists able to block TLR4 and, thereby the cytokine storm. We report here that a mixture of mu-class isoforms from the F. hepatica GST protein family administered intraperitoneally 1 h prior to a lethal LPS injection can modulate the dynamics and abundance of large peritoneal macrophages in the peritoneal cavity of septic mice while significantly suppressing the LPS-induced cytokine storm in a mouse model of septic shock. These results suggest that native F. hepatica glutathione S-transferase is a promising candidate for drug development against endotoxemia and other inflammatory diseases.
Collapse
Affiliation(s)
| | | | | | - Loyda B. Mendez
- School of Sciences and Technologies, University Ana G. Mendez, Carolina, Puerto Rico
| | - Ana M. Espino
- Department of Microbiology, University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
7
|
Andriamboavonjy L, MacDonald A, Hamilton LK, Labrecque M, Boivin MN, Karamchandani J, Stratton JA, Tetreault M. Comparative analysis of methods to reduce activation signature gene expression in PBMCs. Sci Rep 2023; 13:23086. [PMID: 38155174 PMCID: PMC10754832 DOI: 10.1038/s41598-023-49611-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/10/2023] [Indexed: 12/30/2023] Open
Abstract
Preserving the in vivo cell transcriptome is essential for accurate profiling, yet factors during cell isolation including time ex vivo and temperature induce artifactual gene expression, particularly in stress-responsive immune cells. In this study, we investigated two methods to mitigate ex vivo activation signature gene (ASG) expression in peripheral blood mononuclear cells (PBMCs): transcription and translation inhibitors (TTis) and cold temperatures during isolation. Comparative analysis of PBMCs isolated with TTis revealed reduced ASG expression. However, TTi treatment impaired responsiveness to LPS stimulation in subsequent in vitro experiments. In contrast, cold isolation methods also prevented ASG expression; up to a point where the addition of TTis during cold isolation offered minimal additional advantage. These findings highlight the importance of considering the advantages and drawbacks of different isolation methods to ensure accurate interpretation of PBMC transcriptomic profiles.
Collapse
Affiliation(s)
- Lovatiana Andriamboavonjy
- Research Center of the University of Montreal Hospital (CRCHUM), Université de Montréal, Montreal, Canada
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Adam MacDonald
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Laura K Hamilton
- Research Center of the University of Montreal Hospital (CRCHUM), Université de Montréal, Montreal, Canada
| | - Marjorie Labrecque
- Research Center of the University of Montreal Hospital (CRCHUM), Université de Montréal, Montreal, Canada
| | - Marie-Noёlle Boivin
- C-BIG Repository (C-BIG), Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
| | - Jason Karamchandani
- C-BIG Repository (C-BIG), Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
- Department of Pathology, Montreal Neurological Institute, Montreal, QC, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| | - Martine Tetreault
- Research Center of the University of Montreal Hospital (CRCHUM), Université de Montréal, Montreal, Canada.
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Canada.
| |
Collapse
|
8
|
Hyun GH, Cho IH, Yang YY, Jeong DH, Kang YP, Kim YS, Lee SJ, Kwon SW. Mechanisms of interactions in pattern-recognition of common glycostructures across pectin-derived heteropolysaccharides by Toll-like receptor 4. Carbohydr Polym 2023; 314:120921. [PMID: 37173020 DOI: 10.1016/j.carbpol.2023.120921] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 05/15/2023]
Abstract
Complex pectin, originating from terrestrial plant cell walls has been attracting research attention as a promising source of a new innate immune modulator. Numerous bioactive polysaccharides associated with pectin are newly reported every year, but the general mechanism of their immunological action remains unclear owing to the complexity and heterogeneity of pectin. Herein, we systematically investigated the interactions in pattern-recognition for common glycostructures of pectic heteropolysaccharides (HPSs) by Toll-like receptors (TLRs). The compositional similarity of glycosyl residues derived from pectic HPS was confirmed by conducting systematic reviews, leading to molecular modeling of representative pectic segments. Via structural investigation, the inner concavity of leucine-rich repeats of TLR4 was predicted to act as a binding motif for carbohydrate recognition, and subsequent simulations predicted the binding modes and conformations. We experimentally demonstrated that pectic HPS exhibits the non-canonical and multivalent binding aspects for TLR4 resulting in receptor activation. Furthermore, we showed that pectic HPSs were selectively clustered with TLR4 during endocytosis, inducing downstream signals to cause phenotypic activation of macrophages. Overall, we have presented a better explanation for the pattern recognition of pectic HPS and further proposed an approach to understand the interaction between complex carbohydrates and proteins.
Collapse
Affiliation(s)
- Gyu Hwan Hyun
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - In Ho Cho
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoon Young Yang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Da-Hye Jeong
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Yun Pyo Kang
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Seul Ji Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Sung Won Kwon
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
9
|
Valdes-Fernandez BN, Ruiz-Jimenez C, Armina-Rodriguez A, Mendez LB, Espino AM. Fasciola hepatica GST mu-class suppresses the cytokine storm induced by E. coli -lipopolysaccharide whereas modulates the dynamic of peritoneal macrophages in a mouse model and suppresses the classical activation of macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552847. [PMID: 37609327 PMCID: PMC10441391 DOI: 10.1101/2023.08.10.552847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The helminth Fasciola hepatica is known as a master of immunomodulation. It suppresses antigen specific Th1 responses in concurrent bacterial infections while promoting the Th2/Treg regulatory responses, thus demonstrating its anti-inflammatory ability in vivo . We have recently demonstrated that a single intraperitoneal injection with native F. hepatica Glutathione S -Transferase (nFhGST), mostly comprised of mu-class isoforms, can suppresses the cytokine storm and increasing the survival rate in a mouse model of septic shock (1). Knowing that the peritoneal macrophages in response to microbial stimuli play essential roles in the defense, tissue repairment, and maintenance of homeostasis, the present study aimed to determine whether nFhGST could modulate the amount and dynamic of these cells concurrently to the suppression of pro-inflammatory cytokines. The remarkable findings described in this article are, (i) nFhGST suppresses serum IL-12, TNF-α, and IFN-γ in BALB/c mice challenged with a lethal dose of LPS, (ii) Although nFhGST does not elicit IL-10, it was able to significantly suppress the high levels of LPS-induced IL-10, which is considered a key cytokine in the pathophysiology of sepsis (2). iii) nFhGST prevent the disappearance of large peritoneal macrophages (LPM) whereas significantly increasing this population in the peritoneal cavity (PerC) of LPS treated animals, (iv) nFhGST promotes the alternative activation of macrophages whereas suppress the classical activation of macrophages in vitro by expressing high levels of Ym-1, a typical M2-type marker, secreting the production of IL-37, and preventing the production of TNF-α, iNOS2 and nitric oxide, which are typical markers of M1-type macrophages, (v) nFhGST suppress the bacterial phagocytosis of macrophages, a role that plays both, M1-and M2-macrophages, thus partially affecting the capacity of macrophages in destroying microbial pathogens. These findings present the first evidence that nFhGST is an excellent modulator of the PerC content in vivo, reinforcing the capacity of nFhGST as an anti-inflammatory drug against sepsis in animal models. Importance Sepsis is an infection that can lead to a life-threatening complication. Sepsis is the consequence of a systemic bacterial infection that exacerbates the immune cells' activation by bacterial products, resulting in the augmented release of inflammatory mediators. A critical factor in the pathogenesis of sepsis is the primary component of the outer membrane of Gram-negative bacteria known as lipopolysaccharide (LPS), which is sensed by toll-like receptor 4 (TLR4). For this reason, scientists aimed to develop antagonists able to block the cytokine storm by blocking TLR4. We report here that a mixture of mu-class isoforms from the F. hepatica glutathione S-transferase (nFhGST) protein family administered intraperitoneally 1 h after a lethal LPS injection, is capable of significantly suppressing the LPS-induced cytokine storm in a mouse model of septic shock whereas modulate the dynamic and abundance of large peritoneal macrophages in the peritoneal cavity of septic mice. These results suggest that nFhGST is a prominent candidate for drug development against endotoxemia and other inflammatory diseases.
Collapse
|
10
|
Lee TH, Maruthai Y, Abd Aziz NH, Chua KH, Hamdan N, Lee CH, Azmi NA. Chemopreventive and immunoadjuvant properties of standardised edible bird’s nest extract on human breast cancer cell line. INTERNATIONAL FOOD RESEARCH JOURNAL 2023; 30:472-486. [DOI: 10.47836/ifrj.30.2.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
The present work investigated the chemopreventive and immunoadjuvant properties of edible bird’s nest (EBN) extract on breast cancer cell line (MCF-7). Specifically, the cytotoxicity level of EBN extracts (HMG, EHMG, pHMG) against MCF-7, human immune cells of cytotoxic T cells, and monocytes (CD8+ and CD14+) were evaluated by measuring the production of pro-apoptotic and anti-apoptotic molecules released in single and co-culture of MCF-7, CD8+, and CD14+ cells, before and after EBN treatment. The highest cytotoxic effect towards MCF-7 using IC50 of 15 µg/mL was demonstrated by HMG but no effects on CD8+ and CD14+, with cell viability of more than 90%. At the mRNA level, activated CD8+ and CD14+ depicted increased pro-apoptotic gene expression after HMG treatment in co-culture. Additionally, HMG treatment increased apoptosis by down-regulating the regulation of anti-apoptotic genes and up-regulating the pro-apoptotic genes in MCF-7. ELISA and multiplex assay reflected increased pro-apoptotic factors, and decreased anti-apoptotic soluble factors, by non-activated and activated CD8+ and CD14+, in a single or co-culture with MCF-7 after HMG treatment. In conclusion, HMG extract possesses immunoadjuvant properties that can be a potential anticancer agent without causing any deleterious effects on the human immune cells.
Collapse
|
11
|
Chakraborty P, Aravindhan V, Mukherjee S. Helminth-derived biomacromolecules as therapeutic agents for treating inflammatory and infectious diseases: What lessons do we get from recent findings? Int J Biol Macromol 2023; 241:124649. [PMID: 37119907 DOI: 10.1016/j.ijbiomac.2023.124649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Despite the tremendous progress in healthcare sectors, a number of life-threatening infectious, inflammatory, and autoimmune diseases are continuously challenging mankind throughout the globe. In this context, recent successes in utilizing helminth parasite-derived bioactive macromolecules viz. glycoproteins, enzymes, polysaccharides, lipids/lipoproteins, nucleic acids/nucleotides, and small organic molecules for treating various disorders primarily resulted from inflammation. Among the several parasites that infect humans, helminths (cestodes, nematodes, and trematodes) are known as efficient immune manipulators owing to their explicit ability to modulate and modify the innate and adaptive immune responses of humans. These molecules selectively bind to immune receptors on innate and adaptive immune cells and trigger multiple signaling pathways to elicit anti-inflammatory cytokines, expansion of alternatively activated macrophages, T-helper 2, and immunoregulatory T regulatory cell types to induce an anti-inflammatory milieu. Reduction of pro-inflammatory responses and repair of tissue damage by these anti-inflammatory mediators have been exploited for treating a number of autoimmune, allergic, and metabolic diseases. Herein, the potential and promises of different helminths/helminth-derived products as therapeutic agents in ameliorating immunopathology of different human diseases and their mechanistic insights of function at cell and molecular level alongside the molecular signaling cross-talks have been reviewed by incorporating up-to-date findings achieved in the field.
Collapse
Affiliation(s)
- Pritha Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India
| | | | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India.
| |
Collapse
|
12
|
Peng Y, Li Y, Zhang W, ShangGuan Y, Xie T, Wang K, Qiu J, Pu W, Hu B, Zhang X, Yin L, Tang D, Dai Y. The characteristics of extrachromosomal circular DNA in patients with end-stage renal disease. Eur J Med Res 2023; 28:134. [PMID: 36967395 PMCID: PMC10041755 DOI: 10.1186/s40001-023-01064-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 02/15/2023] [Indexed: 03/28/2023] Open
Abstract
BACKGROUND End-stage renal disease (ESRD) is the final stage of chronic kidney disease (CKD). In addition to the structurally intact chromosome genomic DNA, there is a double-stranded circular DNA called extrachromosomal circular DNA (eccDNA), which is thought to be involved in the epigenetic regulation of human disease. However, the features of eccDNA in ESRD patients are barely known. In this study, we identified eccDNA from ESRD patients and healthy people, as well as revealed the characteristics of eccDNA in patients with ESRD. METHODS Using the high-throughput Circle-Sequencing technique, we examined the eccDNA in peripheral blood mononuclear cells (PBMCs) from healthy people (NC) (n = 12) and ESRD patients (n = 16). We analyzed the length distribution, genome elements, and motifs feature of eccDNA in ESRD patients. Then, after identifying the specific eccDNA in ESRD patients, we explored the potential functions of the target genes of the specific eccDNA. Finally, we investigated the probable hub eccDNA using algorithms. RESULTS In total, 14,431 and 11,324 eccDNAs were found in the ESRD and NC groups, respectively, with sizes ranging from 0.01 kb to 60 kb at most. Additionally, the ESRD group had a greater distribution of eccDNA on chromosomes 4, 11, 13, and 20. In two groups, we also discovered several motifs of specific eccDNAs. Furthermore, we identified 13,715 specific eccDNAs in the ESRD group and 10,585 specific eccDNAs in the NC group, both of which were largely annotated as mRNA catalog. Pathway studies using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) showed that the specific eccDNA in ESRD was markedly enriched in cell junction and communication pathways. Furthermore, we identified potentially 20 hub eccDNA-targeting genes from all ESRD-specific eccDNA-targeting genes. Also, we found that 39 eccDNA-targeting genes were associated with ESRD, and some of these eccDNAs may be related to the pathogenesis of ESRD. CONCLUSIONS Our findings revealed the characteristics of eccDNA in ESRD patients and discovered potentially hub and ESRD-relevant eccDNA-targeting genes, suggesting a novel probable mechanism of ESRD.
Collapse
Affiliation(s)
- Yue Peng
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Yixi Li
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Wei Zhang
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
| | - Yu ShangGuan
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
| | - Ting Xie
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Kang Wang
- Key Renal Laboratory of Shenzhen, Department of Nephrology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, Guangdong, China
| | - Jing Qiu
- Key Renal Laboratory of Shenzhen, Department of Nephrology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, Guangdong, China
| | - Wenjun Pu
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
| | - Biying Hu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xinzhou Zhang
- Key Renal Laboratory of Shenzhen, Department of Nephrology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, Guangdong, China
| | - Lianghong Yin
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China.
- Guangzhou Enttxs Medical Products Co., Ltd. P.R. Guangzhou, Guangzhou, China.
| | - Donge Tang
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China.
| | - Yong Dai
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China.
- Department of Pathology, The 924th Hospital of the Chinese People's Liberation Army Joint Logistic Support Force, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin, 541002, Guangxi, China.
| |
Collapse
|
13
|
Weyer R, Hellmann MJ, Hamer-Timmermann SN, Singh R, Moerschbacher BM. Customized chitooligosaccharide production-controlling their length via engineering of rhizobial chitin synthases and the choice of expression system. Front Bioeng Biotechnol 2022; 10:1073447. [PMID: 36588959 PMCID: PMC9795070 DOI: 10.3389/fbioe.2022.1073447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Chitooligosaccharides (COS) have attracted attention from industry and academia in various fields due to their diverse bioactivities. However, their conventional chemical production is environmentally unfriendly and in addition, defined and pure molecules are both scarce and expensive. A promising alternative is the in vivo synthesis of desired COS in microbial platforms with specific chitin synthases enabling a more sustainable production. Hence, we examined the whole cell factory approach with two well-established microorganisms-Escherichia coli and Corynebacterium glutamicum-to produce defined COS with the chitin synthase NodC from Rhizobium sp. GRH2. Moreover, based on an in silico model of the synthase, two amino acids potentially relevant for COS length were identified and mutated to direct the production. Experimental validation showed the influence of the expression system, the mutations, and their combination on COS length, steering the production from originally pentamers towards tetramers or hexamers, the latter virtually pure. Possible explanations are given by molecular dynamics simulations. These findings pave the way for a better understanding of chitin synthases, thus allowing a more targeted production of defined COS. This will, in turn, at first allow better research of COS' bioactivities, and subsequently enable sustainable large-scale production of oligomers.
Collapse
|
14
|
Ariyaratne A, Kim SY, Pollo SMJ, Perera S, Liu H, Nguyen WNT, Coria AL, Luzzi MDC, Bowron J, Szabo EK, Patel KD, Wasmuth JD, Nair MG, Finney CAM. Trickle infection with Heligmosomoides polygyrus results in decreased worm burdens but increased intestinal inflammation and scarring. Front Immunol 2022; 13:1020056. [PMID: 36569914 PMCID: PMC9773095 DOI: 10.3389/fimmu.2022.1020056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/03/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Intestinal roundworms cause chronic debilitating disease in animals, including humans. Traditional experimental models of these types of infection use a large single-dose infection. However, in natural settings, hosts are exposed to parasites on a regular basis and when mice are exposed to frequent, smaller doses of Heligmosomoides polygyrus, the parasites are cleared more quickly. Whether this more effective host response has any negative consequences for the host is not known. Results Using a trickle model of infection, we found that worm clearance was associated with known resistance-related host responses: increased granuloma and tuft cell numbers, increased levels of granuloma IgG and decreased intestinal transit time, as well as higher serum IgE levels. However, we found that the improved worm clearance was also associated with an inflammatory phenotype in and around the granuloma, increased smooth muscle hypertrophy/hyperplasia, and elevated levels of Adamts gene expression. Discussion To our knowledge, we are the first to identify the involvement of this protein family of matrix metalloproteinases (MMPs) in host responses to helminth infections. Our results highlight the delicate balance between parasite clearance and host tissue damage, which both contribute to host pathology. When continually exposed to parasitic worms, improved clearance comes at a cost.
Collapse
Affiliation(s)
- Anupama Ariyaratne
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Sang Yong Kim
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Stephen M. J. Pollo
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Shashini Perera
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Hongrui Liu
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - William N. T. Nguyen
- Departments of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - Aralia Leon Coria
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Mayara de Cassia Luzzi
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Joel Bowron
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Edina K. Szabo
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| | - Kamala D. Patel
- Departments of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, AB, Canada
| | - James D. Wasmuth
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Meera G. Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, United States
| | - Constance A. M. Finney
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, AB, Canada
- Host Parasite Interactions Training Network, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
15
|
Chitin Derived Small Molecule AVR-48 Reprograms the Resting Macrophages to an Intermediate Phenotype and Decrease Pseudomonas aeruginosa Mouse Lung Infection. IMMUNO 2022. [DOI: 10.3390/immuno2040040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
AVR-48 is a structural derivative of chitin previously shown by our laboratory to significantly decrease lung injury parameters in LPS, hyperoxia and sepsis-induced rodent models. The current study objectives are to determine the cellular mechanism of action and demonstrate efficacy in a mouse bacterial lung infection model. For in vitro receptor binding and macrophage polarization studies, C57Bl/6J mouse derived spleens and human peripheral blood mononuclear cells (hPBMCs) were treated with AVR-48 ± LPS or biotin conjugated AVR-48. Different macrophage types were determined using flow cytometry and secreted cytokines were measured using ELISA. In vivo, a CD-1 mouse Pseudomonas aeruginosa lung infection was treated with AVR-48, assessing bacterial colony forming unit (CFU), IL-10 and IL-17A levels in lung and blood samples. AVR-48 binds to both the toll-like receptor 4 (TLR4) and the CD163 receptor on mouse monocytes. In hPBMCs, frequency of intermediate macrophages increased upon AVR-48 treatment for 72 h. Increased bacterial phagocytosis/intracellular killing were observed in THP-1 cells and reduction in CFU in CD-1 mouse lungs. Binding of AVR-48 to both TLR4 and CD163 receptors bring the macrophages to an intermediary stage, resulting in increased phagocytosis and decreased inflammation, altogether providing an optimal immune balance for treating lung injury and infection.
Collapse
|
16
|
Wen Z, Zhang Y, Feng J, Aimulajiang K, Aleem MT, Lu M, Xu L, Song X, Li X, Yan R. Excretory/secretory proteins inhibit host immune responses by downregulating the TLR4/NF-κB/MAPKs signaling pathway: A possible mechanism of immune evasion in parasitic nematode Haemonchus contortus. Front Immunol 2022; 13:1013159. [PMID: 36238295 PMCID: PMC9551057 DOI: 10.3389/fimmu.2022.1013159] [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: 08/06/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Haemonchus contortus is an important parasitic nematode of ruminants. Previous studies showed that H. contortus escape the immunity through complex mechanisms, including releasing excretory/secretory proteins (ESPs) to modulate the host immune response. However, the detailed mechanism through which H. contortus excretory/secretory proteins (HcESPs) promote immune evasion remains unknown. In the present study, we demonstrated that HcESPs inhibit the adaptive immune response of goats including downregulation of immune cell antigen presentation, upregulation of immune checkpoint molecules, activation of the STAT3/PD-L1 pathway, and activation of immunosuppressive regulatory T (Treg) cells. Furthermore, HcESPs reversed the LPS-induced upregulation of pro-inflammatory mediators in PBMCs by inhibiting the TLR4/NF-κB/MAPKs/NLRP3 signaling pathway. Our study provides a better understanding of the evasion mechanisms for H. contortus, which could be helpful in providing an alternative way to prevent the infection of this parasite.
Collapse
Affiliation(s)
- Zhaohai Wen
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yue Zhang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jiajun Feng
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Kalibixiati Aimulajiang
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Muhammad Tahir Aleem
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Mingmin Lu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Lixin Xu
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaokai Song
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiangrui Li
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ruofeng Yan
- Ministry of Education Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Ruofeng Yan,
| |
Collapse
|
17
|
Amelogenin as a regenerative endodontic molecule for immature teeth with apical periodontitis. An experimental study. J Oral Biol Craniofac Res 2022; 12:721-726. [DOI: 10.1016/j.jobcr.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/28/2022] [Accepted: 08/10/2022] [Indexed: 11/21/2022] Open
|
18
|
Zakeri A, Everts B, Williams AR, Nejsum P. Antigens from the parasitic nematode Trichuris suis induce metabolic reprogramming and trained immunity to constrain inflammatory responses in macrophages. Cytokine 2022; 156:155919. [DOI: 10.1016/j.cyto.2022.155919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/27/2022]
|
19
|
Serrano-Maciá M, Lachiondo-Ortega S, Iruzubieta P, Goikoetxea-Usandizaga N, Bosch A, Egia-Mendikute L, Jiménez-Lasheras B, Azkargorta M, Elortza F, Martinez-Redondo D, Castro B, Lozano JJ, Nogueiras R, Irure-Ventura J, Crespo J, Palazón A, Fariñas MC, Delgado TC, López-Hoyos M, Martínez-Chantar ML. Neddylation tunes peripheral blood mononuclear cells immune response in COVID-19 patients. Cell Death Discov 2022; 8:316. [PMID: 35831294 PMCID: PMC9277603 DOI: 10.1038/s41420-022-01115-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 has reached 5.5 million deaths worldwide, generating a huge impact globally. This highly contagious viral infection produces a severe acute respiratory syndrome that includes cough, mucus, fever and pneumonia. Likewise, many hospitalized patients develop severe pneumonia associated with acute respiratory distress syndrome (ARDS), along an exacerbated and uncontrolled systemic inflammation that in some cases induces a fatal cytokine storm. Although vaccines clearly have had a beneficial effect, there is still a high percentage of unprotected patients that develop the pathology, due to an ineffective immune response. Therefore, a thorough understanding of the modulatory mechanisms that regulate the response to SARS-CoV-2 is crucial to find effective therapeutic alternatives. Previous studies describe the relevance of Neddylation in the activation of the immune system and its implications in viral infection. In this context, the present study postulates Neddylation, a reversible ubiquitin-like post-translational modification of proteins that control their stability, localization and activity, as a key regulator in the immune response against SARS-CoV-2. For the first time, we describe an increase in global neddylation levels in COVID-19 in the serum of patients, which is particularly associated with the early response to infection. In addition, the results showed that overactivation of neddylation controls activation, proliferation, and response of peripheral blood mononuclear cells (PBMCs) isolated from COVID-19 patients. Inhibition of neddylation, and the subsequent avoidance of activated PBMCs, reduces cytokine production, mainly IL-6 and MCP-1 and induce proteome modulation, being a critical mechanism and a potential approach to immunomodulate COVID-19 patients.
Collapse
Affiliation(s)
- Marina Serrano-Maciá
- Liver Disease Laboratory, CIC bioGUNE-BRTA (Basque Research & Technology Alliance), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain
| | - Sofia Lachiondo-Ortega
- Liver Disease Laboratory, CIC bioGUNE-BRTA (Basque Research & Technology Alliance), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain
| | - Paula Iruzubieta
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital, Clinical and Translational Digestive Research Group, IDIVAL, Santander, Spain
| | - Naroa Goikoetxea-Usandizaga
- Liver Disease Laboratory, CIC bioGUNE-BRTA (Basque Research & Technology Alliance), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain
| | - Alexandre Bosch
- Cancer Immunology and Immunotherapy Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia, Spain
| | - Leire Egia-Mendikute
- Cancer Immunology and Immunotherapy Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia, Spain
| | - Borja Jiménez-Lasheras
- Cancer Immunology and Immunotherapy Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia, Spain
| | - Mikel Azkargorta
- Proteomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Carlos IIINetworked Proteomics Platform (ProteoRed-ISCIII), 48160, Derio, Bizkaia, Spain
| | - Félix Elortza
- Proteomics Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Carlos IIINetworked Proteomics Platform (ProteoRed-ISCIII), 48160, Derio, Bizkaia, Spain
| | | | - Begoña Castro
- Histocell S.L., Bizkaia Science and Technology Park, Derio, Bizkaia, Spain
| | - Juan J Lozano
- Bioinformatics Platform, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Catalonia, Spain
| | - Ruben Nogueiras
- Department of Physiology, Research Centre of Molecular Medicine and Chronic Diseases, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- Centro de Fisiopatología de la Obesidad y Nutrición, Centro de Investigación Biomédica en Red, Santiago de Compostela, Spain
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Juan Irure-Ventura
- Servicio Inmunología, Hospital Universitario Marqués de Valdecilla-IDIVAL, Facultad de Medicina, Universidad de Cantabria, Barcelona, Cantabria, Spain
| | - Javier Crespo
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital, Clinical and Translational Digestive Research Group, IDIVAL, Santander, Spain
| | - Asís Palazón
- Cancer Immunology and Immunotherapy Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Bizkaia, Spain
| | - María Carmen Fariñas
- Servicio Enfermedades Infecciosas, Hospital Universitario Marqués de Valdecilla-IDIVAL, Facultad de Medicina, Universidad de Cantabria, Barcelona, Cantabria, Spain
| | - Teresa C Delgado
- Liver Disease Laboratory, CIC bioGUNE-BRTA (Basque Research & Technology Alliance), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain
| | - Marcos López-Hoyos
- Servicio Inmunología, Hospital Universitario Marqués de Valdecilla-IDIVAL, Facultad de Medicina, Universidad de Cantabria, Barcelona, Cantabria, Spain
| | - Maria L Martínez-Chantar
- Liver Disease Laboratory, CIC bioGUNE-BRTA (Basque Research & Technology Alliance), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain.
| |
Collapse
|
20
|
Wang J, Zhang L, Shi Q, Yang B, He Q, Wang J, Weng Q. Targeting innate immune responses to attenuate acetaminophen-induced hepatotoxicity. Biochem Pharmacol 2022; 202:115142. [PMID: 35700755 DOI: 10.1016/j.bcp.2022.115142] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 11/02/2022]
Abstract
Acetaminophen (APAP) hepatotoxicity is an important cause of acute liver failure, resulting in massive deaths in many developed countries. Currently, the metabolic process of APAP in the body has been well studied. However, the underlying mechanism of APAP-induced liver injury remains elusive. Increasing clinical and experimental evidences indicate that the innate immune responses are involved in the pathogenesis of APAP-induced acute liver injury (AILI), in which immune cells have dual roles of inducing inflammation to exacerbate hepatotoxicity and removing dead cells and debris to help liver regeneration. In this review, we summarize the latest findings of innate immune cells involved in AILI, particularly emphasizing the activation of innate immune cells and their different roles during the injury and repair phases. Moreover, current available treatments are discussed according to the different roles of innate immune cells in the development of AILI. This review aims to update the knowledge about innate immune responses in the pathogenesis of AILI, and provide potential therapeutic interventions for AILI.
Collapse
Affiliation(s)
- Jincheng Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lulu Zhang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qi Shi
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
21
|
Designing efficient multi-epitope peptide-based vaccine by targeting the antioxidant thioredoxin of bancroftian filarial parasite. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 98:105237. [PMID: 35131521 DOI: 10.1016/j.meegid.2022.105237] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/22/2022] [Accepted: 02/02/2022] [Indexed: 12/24/2022]
Abstract
Thioredoxin is a low molecular weight redox-active protein of filarial parasite that plays a crucial role in downregulating the host immune response to prolong the survival of the parasite within the host body. It has the ability to cope up with the oxidative challenges posed by the host. Hence, the antioxidant protein of the filarial parasite has been suggested to be a useful target for immunotherapeutic intervention of human filariasis. In this study, we have designed a multi-epitope peptide-based vaccine using thioredoxin of Wuchereria bancrofti. Different MHC-I and MHC-II epitopes were predicted using various web servers to construct the vaccine model as MHC-I and MHC-II epitopes are crucial for the development of both humoral and cellular immune responses. Moreover, TLRs specific adjuvants were also incorporated into the vaccine candidates as TLRs are the key immunomodulator to execute innate immunity. Protein-protein molecular docking and simulation analysis between the vaccine and human TLR was performed. TLR5 is the most potent receptor to convey the vaccine-mediated inductive signal for eliciting an innate immune response. A satisfactory immunogenic report from an in-silico immune simulation experiment directed us to propose our vaccine model for experimental and clinical validation. The reverse translated vaccine sequence was also cloned in pET28a(+) to apply the concept in a wet lab experiment in near future. Taken together, this in-silico study on the design of a vaccine construct to target W. bancrofti thioredoxin is predicted to be a future hope in saving human-being from the threat of filariasis.
Collapse
|
22
|
Joardar N, Bhattacharya R, Halder S, Sen A, Biswas SR, Jana K, Babu SPS. Filarial thioredoxin reductase exerts anti-inflammatory effects upon lipopolysaccharide induced inflammation in macrophages. Int J Biol Macromol 2021; 193:1379-1390. [PMID: 34774593 DOI: 10.1016/j.ijbiomac.2021.10.200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/19/2021] [Accepted: 10/27/2021] [Indexed: 01/12/2023]
Abstract
Lymphatic filariasis and its associated health hazards have taken enormous tolls especially in the tropical and sub-tropical countries round the globe. Our present work contemplates the immunomodulatory role of filarial Thioredoxin reductase (TrxR) for the survival of the parasite inside the human host. For this, the protein TrxR was purified from the filarial parasite Setaria cervi and further substantiated through specific anti-TrxR antibody raised in mice. Both commercially available anti-TrxR antibody and laboratory raised antibody produced a single band with a molecular mass of ~80 kDa on western blot. The protein is optimally active at pH 7.0 and at temperature 37 °C. This protein contains both alpha helix and beta pleated sheet with selenocysteine at its active site. The Km was found to be 2.75 ± 0.49 mM. TrxR was found to downregulate lipopolysaccharide (LPS)-induced inflammation in macrophages due to inhibition of TLR4-NF-κB pathway. The result was further supported by the downregulation of inflammasome pathway and activation of alternatively activated macrophages upon TrxR treatment. Hence this study projects insights into the importance of filarial TrxR in host-parasite interface as well as it illustrates novel therapeutic strategy towards anti-filarial drug development.
Collapse
Affiliation(s)
- Nikhilesh Joardar
- Parasitology Laboratory, Department of Zoology, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India
| | - Rajarshi Bhattacharya
- Molecular Food Microbiology Laboratory, Department of Botany, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India
| | - Satyajit Halder
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India
| | - Animesh Sen
- Applied Phycology Laboratory, Department of Botany, Siksha-Bhavana, Visva-Bharati, Santiniketan 731235, West Bengal, India
| | - Swadesh Ranjan Biswas
- Molecular Food Microbiology Laboratory, Department of Botany, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Road, Scheme VIIM, Kankurgachi, Kolkata 700054, India.
| | - Santi Prasad Sinha Babu
- Parasitology Laboratory, Department of Zoology, Siksha Bhavana, Visva-Bharati, Santiniketan 731235, India.
| |
Collapse
|
23
|
Sibi JM, Mohan V, Munisankar S, Babu S, Aravindhan V. Augmented Innate and Adaptive Immune Responses Under Conditions of Diabetes-Filariasis Comorbidity. Front Immunol 2021; 12:716515. [PMID: 34566972 PMCID: PMC8462934 DOI: 10.3389/fimmu.2021.716515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Metainflammation, as seen in chronic diabetes subjects, impairs immunity and increases the susceptibility to infections. In the present study, the effect of diabetes on immune response against filariasis was studied. Both toll-like receptor (TLR)-mediated and crude antigen-induced immune responses were quantified, in whole blood cultures from filariasis-infected subjects (LF+), with and without diabetes. Blood cultures were stimulated with TLR ligands (TLR2 and TLR4) or filarial antigen or were left unstimulated (control) for 18 h. Cytokine, chemokine, and defensin secretion was quantified by ELISA. Expression of HLA-DR, B7-1, B7-2, activation marker (CD69), and Th (Th1, Th2, Th17, and Th9) phenotypes was quantified by flow cytometry. Expression of immunomodulatory effectors (Cox-2, HO-1, IDO-1, and p47Phox) and Th-polarizing transcription factors (T-bet, GATA3, and ROR-γt) was quantified by quantitative PCR. Secretion of IL-27, IL-1Ra, IL-12, IL-33, IL-9, and SDF-1 was increased under diabetes conditions with increased Th9 polarization and increased expression of Cox-2 and IDO. Overall, diabetes was found to augment both TLR-mediated and antigen-induced inflammation, which can promote chronic pathology in LF+ subjects.
Collapse
Affiliation(s)
- Joy Manohar Sibi
- Department of Genetics, Dr A. L. Mudaliar Post Graduate Institute of Basic Medical Sciences (ALM PG IBMS), University of Madras, Chennai, India
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialties Centre, Chennai, India
| | - Saravanan Munisankar
- National Institute of Health-International Centre for Excellence in Research, National Institute for Research in Tuberculosis, Chennai, India
| | - Subash Babu
- National Institute of Health-International Centre for Excellence in Research, National Institute for Research in Tuberculosis, Chennai, India
| | - Vivekanandhan Aravindhan
- Department of Genetics, Dr A. L. Mudaliar Post Graduate Institute of Basic Medical Sciences (ALM PG IBMS), University of Madras, Chennai, India
| |
Collapse
|
24
|
Chitin-Derived AVR-48 Prevents Experimental Bronchopulmonary Dysplasia (BPD) and BPD-Associated Pulmonary Hypertension in Newborn Mice. Int J Mol Sci 2021; 22:ijms22168547. [PMID: 34445253 PMCID: PMC8395179 DOI: 10.3390/ijms22168547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 01/03/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of prematurity and a key contributor to the large health care burden associated with prematurity, longer hospital stays, higher hospital costs, and frequent re-hospitalizations of affected patients through the first year of life and increased resource utilization throughout childhood. This disease is associated with abnormal pulmonary function that may lead to BPD-associated pulmonary hypertension (PH), a major contributor to neonatal mortality and morbidity. In the absence of any definitive treatment options, this life-threatening disease is associated with high resource utilization during and after neonatal intensive care unit (NICU) stay. The goal of this study was to test the safety and efficacy of a small molecule derivative of chitin, AVR-48, as prophylactic therapy for preventing experimental BPD in a mouse model. Two doses of AVR-48 were delivered either intranasally (0.11 mg/kg), intraperitoneally (10 mg/kg), or intravenously (IV) (10 mg/kg) to newborn mouse pups on postnatal day (P)2 and P4. The outcomes were assessed by measuring total inflammatory cells in the broncho-alveolar lavage fluid (BALF), chord length, septal thickness, and radial alveolar counts of the alveoli, Fulton’s Index (for PH), cell proliferation and cell death by immunostaining, and markers of inflammation by Western blotting and ELISA. The bioavailability and safety of the drug were assessed by pharmacokinetic and toxicity studies in both neonatal mice and rat pups (P3-P5). Following AVR-48 treatment, alveolar simplification was improved, as evident from chord length, septal thickness, and radial alveolar counts; total inflammatory cells were decreased in the BALF; Fulton’s Index was decreased and lung inflammation and cell death were decreased, while angiogenesis and cell proliferation were increased. AVR-48 was found to be safe and the no-observed-adverse-effect level (NOAEL) in rat pups was determined to be 100 mg/kg when delivered via IV dosing with a 20-fold safety margin. With no reported toxicity and with a shorter half-life, AVR-48 is able to reverse the worsening cardiopulmonary phenotype of experimental BPD and BPD-PH, compared to controls, thus positioning it as a future drug candidate.
Collapse
|
25
|
Al-Rahim AM, AlChalabi R, Al-Saffar AZ, Sulaiman GM, Albukhaty S, Belali T, Ahmed EM, Khalil KAA. Folate-methotrexate loaded bovine serum albumin nanoparticles preparation: an in vitro drug targeting cytokines overwhelming expressed immune cells from rheumatoid arthritis patients. Anim Biotechnol 2021; 34:166-182. [PMID: 34319853 DOI: 10.1080/10495398.2021.1951282] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The study planned to estimate biological parameters linked to rheumatoid arthritis (RA) patients, detecting the influence of MTX and biotherapy treatments on these parameters and synthesizing methotrexate bovine serum albumin nanoparticles linked to folate (FA-MTX-BSA NPs) to reduce the overwhelming expression of inflammatory cytokines. Inflammatory parameters showed significant increases in newly diagnosed and MTX-receiving groups while no changes were observed in the biotherapy-maintained group. MTX-loaded BSA nanoparticles were fabricated by the desolvation method and further linked to activated folic acid to obtain FA-MTX-BSA NPs. FA-MTX-BSA NPs were successfully characterized within the nanoscale range using different screening techniques. FA-MTX-BSA NPs showed an in vitro release in a sustained manner. The potential of MTX, MTX-BSA NPs, and FA-MTX-BSA NPs in inducing cytokine level reduction was detected. Significant decreases in interleukin- 1 beta (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) levels were obtained in cultures treated with FA-MTX-BSA NPs compared to the untreated culture in a dose-dependent pattern. Furthermore, FA-MTX-BSA NPs comparing with MTX and MTX-BSA NPs exhibited a significant advanced effect in decreasing cytokines levels. Accordingly, the conjunction of BSA NPs and MTX linked to folate potentially reduced cytokines manifestation in RA.
Collapse
Affiliation(s)
- Aya M Al-Rahim
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | - Rawaa AlChalabi
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | - Ali Z Al-Saffar
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | | | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan, Iraq
| | - Tareg Belali
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Elsadig M Ahmed
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia.,Department of Clinical Chemistry, Faculty of Medical Laboratory Sciences, University of El Imam El Mahdi, Kosti, Sudan
| | - Khalil A A Khalil
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Medicine and Health Sciences, University of Hodeidah, Hodeidah, Yemen
| |
Collapse
|
26
|
Das NC, Sen Gupta PS, Biswal S, Patra R, Rana MK, Mukherjee S. In-silico evidences on filarial cystatin as a putative ligand of human TLR4. J Biomol Struct Dyn 2021; 40:8808-8824. [PMID: 33955317 DOI: 10.1080/07391102.2021.1918252] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cystatin is a small molecular weight immunomodulatory protein of filarial parasite that plays a pivotal role in downregulating the host immune response to prolong the survival of the parasite inside the host body. Hitherto, this protein is familiar as an inhibitor of human proteases. However, growing evidences on the role of cystatin in regulating inflammatory homeostasis prompted us to investigate the molecular reasons behind the explicit anti-inflammatory trait of this protein. We have explored molecular docking and molecular dynamics simulation approaches to explore the interaction of cystatin of Wuchereria bancrofti (causative parasite of human filariasis) with human Toll-like receptors (TLRs). TLRs are the most crucial component of frontline host defence against pathogenic infections including filarial infection. Our in-silico data clearly revealed that cystatin strongly interacts with the extracellular domain of TLR4 (binding energy=-93.5 ± 10 kJ/mol) and this biophysical interaction is mediated by hydrogen bonding and hydrophobic interaction. Molecular dynamics simulation analysis revealed excellent stability of the cystatin-TLR4 complex. Taken together, our data indicated that cystatin appears to be a ligand of TLR4 and we hypothesize that cystatin-TLR4 interaction most likely to play a key role in activating the alternative activation pathways to establish an anti-inflammatory milieu. Thus, the study provokes the development of chemotherapeutics and/or vaccines for targeting the cystatin-TLR4 interaction to disrupt the pathological attributes of human lymphatic filariasis. Our findings are expected to provide a novel dimension to the existing knowledge on filarial immunopathogenesis and it will encourage the scientific communities for experimental validation of the present investigation. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nabarun Chandra Das
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Parth Sarthi Sen Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Berhampur, Odisha, India
| | - Satyaranjan Biswal
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Berhampur, Odisha, India
| | - Ritwik Patra
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Malay Kumar Rana
- Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Berhampur, Odisha, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| |
Collapse
|
27
|
Tu HF, Ko CJ, Lee CT, Lee CF, Lan SW, Lin HH, Lin HY, Ku CC, Lee DY, Chen IC, Chuang YH, Del Caño-Ochoa F, Ramón-Maiques S, Ho CC, Lee MS, Chang GD. Afatinib Exerts Immunomodulatory Effects by Targeting the Pyrimidine Biosynthesis Enzyme CAD. Cancer Res 2021; 81:3270-3282. [PMID: 33771897 DOI: 10.1158/0008-5472.can-20-3436] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/24/2021] [Accepted: 03/18/2021] [Indexed: 11/16/2022]
Abstract
Current clinical trials of combined EGFR-tyrosine kinase inhibitors (TKI) and immune checkpoint blockade (ICB) therapies show no additional effect. This raises questions regarding whether EGFR-TKIs attenuate ICB-enhanced CD8+ T lymphocyte function. Here we show that the EGFR-TKI afatinib suppresses CD8+ T lymphocyte proliferation, and we identify CAD, a key enzyme of de novo pyrimidine biosynthesis, to be a novel afatinib target. Afatinib reduced tumor-infiltrating lymphocyte numbers in Lewis lung carcinoma (LLC)-bearing mice. Early afatinib treatment inhibited CD8+ T lymphocyte proliferation in patients with non-small cell lung cancer, but their proliferation unexpectedly rebounded following long-term treatment. This suggests a transient immunomodulatory effect of afatinib on CD8+ T lymphocytes. Sequential treatment of afatinib with anti-PD1 immunotherapy substantially enhanced therapeutic efficacy in MC38 and LLC-bearing mice, while simultaneous combination therapy showed only marginal improvement over each single treatment. These results suggest that afatinib can suppress CD8+ T lymphocyte proliferation by targeting CAD, proposing a timing window for combined therapy that may prevent the dampening of ICB efficacy by EGFR-TKIs. SIGNIFICANCE: This study elucidates a mechanism of afatinib-mediated immunosuppression and provides new insights into treatment timing for combined targeted therapy and immunotherapy. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3270/F1.large.jpg.
Collapse
Affiliation(s)
- Hsin-Fang Tu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Jung Ko
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ching-Tai Lee
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Fan Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shao-Wei Lan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Hsien Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Ying Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Chi Ku
- Graduate Institute of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Der-Yen Lee
- Graduate Institute of Integrated Medicine/Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - I-Chun Chen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Hui Chuang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Francisco Del Caño-Ochoa
- Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain.,Group 739, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-Instituto de Salud Carlos III, Valencia, Spain
| | - Santiago Ramón-Maiques
- Instituto de Biomedicina de Valencia (IBV-CSIC), Valencia, Spain.,Group 739, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-Instituto de Salud Carlos III, Valencia, Spain
| | - Chao-Chi Ho
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Ming-Shyue Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Geen-Dong Chang
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
28
|
Shah D, Das P, Acharya S, Agarwal B, Christensen DJ, Robertson SM, Bhandari V. Small Immunomodulatory Molecules as Potential Therapeutics in Experimental Murine Models of Acute Lung Injury (ALI)/Acute Respiratory Distress Syndrome (ARDS). Int J Mol Sci 2021; 22:ijms22052573. [PMID: 33806560 PMCID: PMC7961996 DOI: 10.3390/ijms22052573] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 12/23/2022] Open
Abstract
Background: Acute lung injury (ALI) or its most advanced form, acute respiratory distress syndrome (ARDS) is a severe inflammatory pulmonary process triggered by a variety of insults including sepsis, viral or bacterial pneumonia, and mechanical ventilator-induced trauma. Currently, there are no effective therapies available for ARDS. We have recently reported that a novel small molecule AVR-25 derived from chitin molecule (a long-chain polymer of N-acetylglucosamine) showed anti-inflammatory effects in the lungs. The goal of this study was to determine the efficacy of two chitin-derived compounds, AVR-25 and AVR-48, in multiple mouse models of ALI/ARDS. We further determined the safety and pharmacokinetic (PK) profile of the lead compound AVR-48 in rats. Methods: ALI in mice was induced by intratracheal instillation of a single dose of lipopolysaccharide (LPS; 100 µg) for 24 h or exposed to hyperoxia (100% oxygen) for 48 h or undergoing cecal ligation and puncture (CLP) procedure and observation for 10 days. Results: Both chitin derivatives, AVR-25 and AVR-48, showed decreased neutrophil recruitment and reduced inflammation in the lungs of ALI mice. Further, AVR-25 and AVR-48 mediated diminished lung inflammation was associated with reduced expression of lung adhesion molecules with improvement in pulmonary endothelial barrier function, pulmonary edema, and lung injury. Consistent with these results, CLP-induced sepsis mice treated with AVR-48 showed a significant increase in survival of the mice (80%) and improved lung histopathology in the treated CLP group. AVR-48, the lead chitin derivative compound, demonstrated a good safety profile. Conclusion: Both AVR-25 and AVR-48 demonstrate the potential to be developed as therapeutic agents to treat ALI/ARDS.
Collapse
Affiliation(s)
- Dilip Shah
- Division of Neonatology, Department of Pediatrics, Drexel University, Philadelphia, PA 19197, USA; (D.S.); (P.D.)
| | - Pragnya Das
- Division of Neonatology, Department of Pediatrics, Drexel University, Philadelphia, PA 19197, USA; (D.S.); (P.D.)
| | - Suchismita Acharya
- AyuVis Research, Inc., 1120 South Freeway, Fort Worth, TX 76104, USA; (S.A.); (S.M.R.)
- Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76104, USA
| | | | - Dale J. Christensen
- Dale J. Christensen Consulting LLC, Cary, NC 27511, USA;
- Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC 27722, USA
| | - Stella M. Robertson
- AyuVis Research, Inc., 1120 South Freeway, Fort Worth, TX 76104, USA; (S.A.); (S.M.R.)
- Arrochar Consulting LLC, Fort Worth, TX 76104, USA
| | - Vineet Bhandari
- Division of Neonatology, Department of Pediatrics, Drexel University, Philadelphia, PA 19197, USA; (D.S.); (P.D.)
- Correspondence:
| |
Collapse
|
29
|
Jabeen K, Malik U, Mansoor S, Shahzad S, Zahid S, Javed A. Effect of oxidative stress and calcium deregulation on FAM26F (CALHM6) expression during hepatitis B virus infection. BMC Infect Dis 2021; 21:228. [PMID: 33639860 PMCID: PMC7913464 DOI: 10.1186/s12879-021-05888-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 02/10/2021] [Indexed: 11/23/2022] Open
Abstract
Background Family with sequence similarity 26, member F (FAM26F) is an important innate immunity modulator playing a significant role in diverse immune responses, however, the association of FAM26F expression with HBV infection is not yet known. Thus, the current study aims to explore the differential expression of FAM26F in vitro in HepAD38 and HepG2 cell lines upon HBV infection, and in vivo in HBV infected individuals. The effects of antioxidant and calcium inhibitors on the regulation of FAM26F expression were also evaluated. The expression of FAM26F was simultaneously determined with well-established HBV infection markers: IRF3, and IFN-β. Methods The expression of FAM26F and marker genes was analyzed through Real-time qPCR and western blot. Results Our results indicate that the differential expression of FAM26F followed the same trend as that of IRF3 and IFN-β. The in vitro study revealed that, in both HBV infected cell lines, FAM26F expression was significantly down-regulated as compared to uninfected control cells. Treatment of cells with N-acetyl-L-cysteine (NAC), EGTA-AM, BAPTA-AM, and Ru360 significantly upregulated the expression of FAM26F in both the cell lines. Moreover, in in vivo study, FAM26F expression was significantly downregulated in all HBV infected groups as compared to controls (p = 0.0007). The expression was higher in the HBV recovered cases, probably due to the decrease in infection and increase in the immunity of these individuals. Conclusion Our study is the first to show the association of FAM26F with HBV infection. It is proposed that FAM26F expression could be an early predictive marker for HBV infection, and thus is worthy of further investigation. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-05888-0.
Collapse
Affiliation(s)
- Kehkshan Jabeen
- Genomics Research Lab, Department of Biological Sciences, International Islamic University Islamabad, Islamabad, 44000, Pakistan
| | - Uzma Malik
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan
| | - Sajid Mansoor
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan.,Department of Microbiology, Faculty of Life Sciences, University of Central Punjab, Lahore, 54000, Pakistan
| | - Shaheen Shahzad
- Genomics Research Lab, Department of Biological Sciences, International Islamic University Islamabad, Islamabad, 44000, Pakistan
| | - Saadia Zahid
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan
| | - Aneela Javed
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan.
| |
Collapse
|
30
|
Steinhagen F, Hilbert T, Cramer N, Senzig S, Parcina M, Bode C, Boehm O, Frede S, Klaschik S. Development of a minimal invasive and controllable murine model to study polymicrobial abdominal sepsis. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1909663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Folkert Steinhagen
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Tobias Hilbert
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Nina Cramer
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Sebastian Senzig
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Marijo Parcina
- Department of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Olaf Boehm
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Stilla Frede
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Sven Klaschik
- Department of Anesthesiology and Critical Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
31
|
Das P, Acharya S, Shah D, Agarwal B, Prahaladan V, Bhandari V. Chitin Analog AVR-25 Prevents Experimental Bronchopulmonary Dysplasia. J Pediatr Intensive Care 2020; 9:225-232. [PMID: 32685255 DOI: 10.1055/s-0040-1709994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 03/27/2020] [Indexed: 12/12/2022] Open
Abstract
Infants born extremely preterm are at a high risk of developing bronchopulmonary dysplasia (BPD) which is characterized by large, simplified alveoli, increased inflammation, disrupted and dysregulated vasculogenesis, decreased cell proliferation, and increased cell death in the lungs. Due to lack of specific drug treatments to combat this condition, BPD and its long-term complications have taken a significant toll of healthcare resources. AVR-25, a novel immune modulator experimental compound, was able to partially recover the pulmonary phenotype in the hyperoxia-induced experimental mouse model of BPD. We anticipate that AVR-25 will have therapeutic potential for managing human BPD.
Collapse
Affiliation(s)
- Pragnya Das
- Department of Pediatrics, Division of Neonatology, Drexel University, Philadelphia, Pennsylvania, United States
| | - Suchismita Acharya
- Acceleration Laboratory, University of North Texas Health Science Center, Fort Worth, Texas, United States.,AyuVis Research Inc, Fort Worth, Texas, United States
| | - Dilip Shah
- Department of Pediatrics, Division of Neonatology, Drexel University, Philadelphia, Pennsylvania, United States
| | | | - Varsha Prahaladan
- Department of Pediatrics, Division of Neonatology, Drexel University, Philadelphia, Pennsylvania, United States
| | - Vineet Bhandari
- Department of Pediatrics, Division of Neonatology, Drexel University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
32
|
Self-reactive IgG4 antibodies are associated with blocking of pathology in human lymphatic filariasis. Cell Immunol 2019; 341:103927. [DOI: 10.1016/j.cellimm.2019.103927] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/11/2019] [Accepted: 05/21/2019] [Indexed: 01/07/2023]
|
33
|
Udgata A, Dolasia K, Ghosh S, Mukhopadhyay S. Dribbling through the host defence: targeting the TLRs by pathogens. Crit Rev Microbiol 2019; 45:354-368. [PMID: 31241370 DOI: 10.1080/1040841x.2019.1608904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Atul Udgata
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Manipal Academy of Higher Education, Manipal, India
| | - Komal Dolasia
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Manipal Academy of Higher Education, Manipal, India
| | - Sudip Ghosh
- Molecular Biology Division, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| |
Collapse
|
34
|
Martínez-Barricarte R, Kong XF, Casanova JL. Measurement of CD74 N-terminal Fragment Accumulation in Cells Treated with SPPL2a Inhibitor. Bio Protoc 2019; 9:e3254. [PMID: 33654779 DOI: 10.21769/bioprotoc.3254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/29/2019] [Accepted: 05/27/2019] [Indexed: 11/02/2022] Open
Abstract
The recent discovery of human signal peptide peptidase-like 2a (SPPL2a) deficiency in humans revealed the toxicity associated with the accumulation of one of its substrates, CD74 N-terminal fragment (CD74-NTF), for certain type of dendritic cells (cDC2). We developed a two-step protocol for monitoring the accumulation of this molecule in different subsets of PBMCs and immortalized B cells, in which SPPL2a is chemically inhibited and CD74-NTF levels are then assessed by flow cytometry or western blotting. The chemical inhibition of SPPL2a has been described elsewhere, but this is the first time that this inhibition has been reported as a protocol.
Collapse
Affiliation(s)
- Rubén Martínez-Barricarte
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York City, NY, USA
| | - Xiao-Fei Kong
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York City, NY, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York City, NY, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France.,Howard Hughes Medical Institute, New York, USA.,Pediatric Hematology and Immunology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| |
Collapse
|
35
|
Chang SH, Wu GJ, Wu CH, Huang CH, Tsai GJ. Oral administration with chitosan hydrolytic products modulates mitogen-induced and antigen-specific immune responses in BALB/c mice. Int J Biol Macromol 2019; 131:158-166. [DOI: 10.1016/j.ijbiomac.2019.02.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/22/2019] [Accepted: 02/11/2019] [Indexed: 01/15/2023]
|
36
|
Das P, Panda SK, Agarwal B, Behera S, Ali SM, Pulse ME, Solomkin JS, Opal SM, Bhandari V, Acharya S. Novel Chitohexaose Analog Protects Young and Aged mice from CLP Induced Polymicrobial Sepsis. Sci Rep 2019; 9:2904. [PMID: 30814582 PMCID: PMC6393422 DOI: 10.1038/s41598-019-38731-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/31/2018] [Indexed: 02/06/2023] Open
Abstract
In Gram-negative bacterial sepsis, production of excess pro-inflammatory cytokines results in hyperinflammation and tissue injury. Anti-inflammatory cytokines such as IL-10 inhibit inflammation and enhance tissue healing. Here, we report a novel approach to treat septicemia associated with intra-abdominal infection in a murine model by delicately balancing pro- and anti-inflammatory cytokines. A novel oligosaccharide compound AVR-25 selectively binds to the TLR4 protein (IC50 = 0.15 µM) in human peripheral blood monocytes and stimulates IL-10 production. Following the cecal ligation and puncture (CLP) procedure, intravenous dosing of AVR-25 (10 mg/kg, 6-12 h post-CLP) alone and in combination with antibiotic imipenem protected both young adult (10-12 week old) and aged (16-18 month old) mice against polymicrobial infection, organ dysfunction, and death. Proinflammatory cytokines (TNF-α, MIP-1, i-NOS) were decreased significantly and restoration of tissue damage was observed in all organs. A decrease in serum C-reactive protein (CRP) and bacterial colony forming unit (CFU) confirmed improved bacterial clearance. Together, these findings demonstrate the therapeutic ability of AVR-25 to mitigate the storm of inflammation and minimize tissue injury with high potential for adjunctive therapy in intra-abdominal sepsis.
Collapse
Affiliation(s)
- Pragnya Das
- Department of Pediatrics, Division of Neonatology, Drexel University School of Medicine, Philadelphia, PA, 19102, USA
| | - Santosh K Panda
- School of Medicine, Washington University, St. Louis, MO, 63110, USA
| | | | - Sumita Behera
- AyuVis Research Inc, 1120 South Freeway, Fort Worth, TX, 76104, USA.,University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Syed M Ali
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, 110025, India
| | - Mark E Pulse
- Preclinical Service, University of North Texas Health Science Center, Fort Worth, Texas, 76107, USA
| | - Joseph S Solomkin
- University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Steven M Opal
- The Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Vineet Bhandari
- Department of Pediatrics, Division of Neonatology, Drexel University School of Medicine, Philadelphia, PA, 19102, USA
| | - Suchismita Acharya
- AyuVis Research Inc, 1120 South Freeway, Fort Worth, TX, 76104, USA. .,Acceleration laboratory, University of North Texas Health Science Center, Fort Worth, Texas, 76107, USA.
| |
Collapse
|
37
|
Buerfent BC, Gölz L, Hofmann A, Rühl H, Stamminger W, Fricker N, Hess T, Oldenburg J, Nöthen MM, Schumacher J, Hübner MP, Hoerauf A. Transcriptome-wide analysis of filarial extract-primed human monocytes reveal changes in LPS-induced PTX3 expression levels. Sci Rep 2019; 9:2562. [PMID: 30796272 PMCID: PMC6385373 DOI: 10.1038/s41598-019-38985-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 01/15/2019] [Indexed: 12/24/2022] Open
Abstract
Filarial nematodes modulate immune responses in their host to enable their survival and mediate protective effects against autoimmunity and allergies. In this study, we examined the immunomodulatory capacity of extracts from the human pathogenic filaria Brugia malayi (BmA) on human monocyte responses in a transcriptome-wide manner to identify associated pathways and diseases. As previous transcriptome studies often observed quiescent responses of innate cells to filariae, the potential of BmA to alter LPS driven responses was investigated by analyzing >47.000 transcripts of monocytes from healthy male volunteers stimulated with BmA, Escherichia coli LPS or a sequential stimulation of both. In comparison to ~2200 differentially expressed genes in LPS-only stimulated monocytes, only a limited number of differentially expressed genes were identified upon BmA priming before LPS re-stimulation with only PTX3↓ reaching statistical significance after correcting for multiple testing. Nominal significant differences were reached for metallothioneins↑, MMP9↑, CXCL5/ENA-78↑, CXCL6/GCP-2↑, TNFRSF21↓, and CCL20/MIP3α↓ and were confirmed by qPCR or ELISA. Flow cytometric analysis of activation markers revealed a reduced LPS-induced expression of HLA-DR and CD86 on BmA-primed monocytes as well as a reduced apoptosis of BmA-stimulated monocytes. While our experimental design does not allow a stringent extrapolation of our results to the development of filarial pathology, several genes that were identified in BmA-primed monocytes had previously been associated with filarial pathology, supporting the need for further research.
Collapse
Affiliation(s)
- B C Buerfent
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - L Gölz
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Bonn, Germany
- Department of Orthodontics and Orofacial Orthopedics, University Hospital of Erlangen, Erlangen, Germany
| | - A Hofmann
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - H Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital of Bonn, Bonn, Germany
| | - W Stamminger
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| | - N Fricker
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - T Hess
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - J Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital of Bonn, Bonn, Germany
| | - M M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - J Schumacher
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - M P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany.
| | - A Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
38
|
Aguayo V, Valdés Fernandez BN, Rodríguez-Valentín M, Ruiz-Jiménez C, Ramos-Benítez MJ, Méndez LB, Espino AM. Fasciola hepatica GST downregulates NF-κB pathway effectors and inflammatory cytokines while promoting survival in a mouse septic shock model. Sci Rep 2019; 9:2275. [PMID: 30783117 PMCID: PMC6381083 DOI: 10.1038/s41598-018-37652-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/28/2018] [Indexed: 12/14/2022] Open
Abstract
Parasitic helminths and helminth-derived molecules have demonstrated to possess powerful anti-inflammatory properties and confirmed therapeutic effects on inflammatory diseases. The helminth Fasciola hepatica has been reported to suppress specific Th1 specific immune responses induced by concurrent bacterial infections, thus demonstrating its anti-inflammatory ability in vivo. In this study, we demonstrate that native F. hepatica glutathione S-transferase (nFhGST), a major parasite excretory-secretory antigen, majorly comprised of Mu-class GST isoforms, significantly suppresses the LPS-induced TNFα and IL1β of mouse bone-marrow derived macrophages in vitro and the pro-inflammatory cytokine/chemokine storm within C57BL/6 mice exposed to lethal doses of LPS increasing their survival rate by more than 85%. Using THP1-Blue CD14 cells, a human monocyte cell line, we also demonstrate that nFhGST suppresses NF-κB activation in response to multiple TLR-ligands, including whole bacteria clinical isolates and this suppression was found to be dose-dependent and independent of the timing of exposure. Moreover, the suppressive effect of nFhGST on NF-κB activation was shown to be independent of enzyme activity or secondary structure of protein. As part of its anti-inflammatory effect nFhGST target multiple proteins of the canonic and non-canonic NF-κB signaling pathway as well as also JAK/STAT pathway. Overall, our results demonstrate the potent anti-inflammatory properties of nFhGST and its therapeutic potential as an anti-inflammatory agent.
Collapse
Affiliation(s)
- Vasti Aguayo
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | | | | | - Caleb Ruiz-Jiménez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Marcos J Ramos-Benítez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Loyda B Méndez
- School of Science & Technology Universidad del Este, Carolina, Puerto Rico
| | - Ana M Espino
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico.
| |
Collapse
|
39
|
Ramos-Benitez MJ, Ruiz-Jimenez C, Rosado-Franco JJ, Ramos-Pérez WD, Mendez LB, Osuna A, Espino AM. Fh15 Blocks the Lipopolysaccharide-Induced Cytokine Storm While Modulating Peritoneal Macrophage Migration and CD38 Expression within Spleen Macrophages in a Mouse Model of Septic Shock. mSphere 2018; 3:e00548-18. [PMID: 30567900 PMCID: PMC6300687 DOI: 10.1128/msphere.00548-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/05/2018] [Indexed: 12/22/2022] Open
Abstract
Sepsis caused by Gram-negative bacteria is the consequence of an unrestrained infection that continuously releases lipopolysaccharide (LPS) into the bloodstream, which triggers an uncontrolled systemic inflammatory response leading to multiorgan failure and death. After scrutinizing the immune modulation exerted by a recombinant Fasciola hepatica fatty acid binding protein termed Fh15, our group demonstrated that addition of Fh15 to murine macrophages 1 h prior to LPS stimulation significantly suppresses the expression of proinflammatory cytokines tumor necrosis factor alpha (TNF-α) and interleukin-1β (IL1-β). The present study aimed to demonstrate that Fh15 could exert a similar anti-inflammatory effect in vivo using a mouse model of septic shock. Among the novel findings reported in this article, (i) Fh15 suppressed numerous serum proinflammatory cytokines/chemokines when injected intraperitoneally 1 h after exposure of animals to lethal doses of LPS, (ii) concurrently, Fh15 increased the population of large peritoneal macrophages (LPMs) in the peritoneal cavity (PerC) of LPS-injected animals, and (iii) Fh15 downregulated the expression on spleen macrophages of CD38, a cell surface ectoenzyme with a critical role during inflammation. These findings present the first evidence that the recombinant parasitic antigen Fh15 is an excellent modulator of the PerC cell content and in vivo macrophage activation, endorsing Fh15's potential as a drug candidate against sepsis-related inflammatory response.IMPORTANCE Sepsis is a potentially life-threatening complication of an infection. Sepsis is mostly the consequence of systemic bacterial infections leading to exacerbated activation of immune cells by bacterial products, resulting in enhanced release of inflammatory mediators. Lipopolysaccharide (LPS), the major component of the outer membrane of Gram-negative bacteria, is a critical factor in the pathogenesis of sepsis, which is sensed by Toll-like receptor 4 (TLR4). The scientific community highly pursues the development of antagonists capable of blocking the cytokine storm by blocking TLR4. We report here that a recombinant molecule of 14.5 kDa belonging to the Fasciola hepatica fatty acid binding protein (Fh15) is capable of significantly suppressing the LPS-induced cytokine storm in a mouse model of septic shock when administered by the intraperitoneal route 1 h after a lethal LPS injection. These results suggest that Fh15 is an excellent candidate for drug development against endotoxemia.
Collapse
Affiliation(s)
- Marcos J Ramos-Benitez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Caleb Ruiz-Jimenez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Jose J Rosado-Franco
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Willy D Ramos-Pérez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Loyda B Mendez
- School of Science & Technology Universidad del Este, Carolina, Puerto Rico
| | - Antonio Osuna
- Instituto de Biotecnologia, Grupo de Bioquimica y Parasitología Molecular, Departamento de Parasitologia, Universidad de Granada, Granada, Spain
| | - Ana M Espino
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| |
Collapse
|
40
|
Sahoo PK, Panda SK, Satapathy AK, Pati S. Anti-filarial immunity blocks parasite development and plays a protective role. PLoS One 2018; 13:e0199090. [PMID: 29927974 PMCID: PMC6013016 DOI: 10.1371/journal.pone.0199090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/31/2018] [Indexed: 11/25/2022] Open
Abstract
Lymphatic filariasis is a complex parasitic disease having a spectrum of clinical parameters which are critical in deciding the severity of the pathogenesis. Individuals residing in the endemic areas are categorized into different clinical groups such as: EC (endemic controls-free of disease and infection), AS (asymptomatic carriers- free of disease but carries both antigens and microfilaria (Mf) in circulation), CR (cryptic-free of disease and Mf but having circulatory antigen) and CH (chronic-having manifestations of elephantiasis and hydrocele). The immune response to the parasitic infection is well studied, whereas the protective mechanism explaining the fate of antigenemia and filaremia between AS and CR group remains unexplained. Increased anti-Mf antibodies have been implicated for Mf clearance in experimental infection models whereas its role in clinical filariasis is not known. Here, we followed up two groups of 24 and 33 CR cases for 18 and 36 months respectively and analyzed both the clinical parameters and the anti-filarial antibody response. The humoral response to both whole filarial antigen and Mf antigens as well as recombinant active parasitic antigens was significantly higher in CR cases than AS individuals, whereas the clinical parameters remain unchanged. This study made insights into the protective immune mechanism responsible for the clearance of Mf from circulation in CR individuals.
Collapse
Affiliation(s)
- Prakash Kumar Sahoo
- Division of Immunology, ICMR-Regional Medical Research Centre, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - Santosh K. Panda
- Division of Immunology, ICMR-Regional Medical Research Centre, Chandrasekharpur, Bhubaneswar, Odisha, India
- Medimmune, One Medimmune Way, Gaithersburg, MD, United States of America
| | - Ashok Kumar Satapathy
- Division of Immunology, ICMR-Regional Medical Research Centre, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - Sanghamitra Pati
- Division of Immunology, ICMR-Regional Medical Research Centre, Chandrasekharpur, Bhubaneswar, Odisha, India
| |
Collapse
|
41
|
Ahmed A, Dolasia K, Mukhopadhyay S. Mycobacterium tuberculosisPPE18 Protein Reduces Inflammation and Increases Survival in Animal Model of Sepsis. THE JOURNAL OF IMMUNOLOGY 2018; 200:3587-3598. [DOI: 10.4049/jimmunol.1602065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/14/2018] [Indexed: 12/16/2022]
|
42
|
Yang T, Fang F, Chen Y, Ma J, Xiao Z, Zou S, Zheng N, Yan D, Liao S, Chen S, Fang H, Yu C, Liu J, Dong M. Elevated plasma interleukin-37 playing an important role in acute coronary syndrome through suppression of ROCK activation. Oncotarget 2018; 8:9686-9695. [PMID: 28039466 PMCID: PMC5354763 DOI: 10.18632/oncotarget.14195] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 11/24/2016] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE The plasma level of interleukin-37 is elevated in patients with acute coronary syndrome, however, its function during the onset and progress of the disease remains unclear. This study aimed to investigate the clinical significance of IL-37 in acute coronary syndrome and its underlying mechanism. METHODS 124 patients with acute coronary syndrome and 40 healthy controls were recruited in this study. Plasma interleukin-37 levels were measured in 41 patients with ST elevation myocardial infarction (STEMI), 41 patients with non-STEMI, 42 patients with unstable angina, and 40 controls. Mortality was defined as an event. RESULTS In this study, the mean follow-up period was 824±306 days (2-1077 days). 22% (n=27) of patients died. The mortality rate was significantly lower in patients with interleukin-37 serum levels below the median (6.4 pg/mL) than those with interleukin-37 serum levels above 6.4 pg/mL at 36-month follow-up (16% vs. 24%, p=0.02, log rank X2=5.39). Highly concentration of the anti-inflammatory interleukin-37 exerted a protective effect by suppressing the activated Rho Kinase (ROCK) activity in the peripheral blood mononuclear cells in vivo and in vitro after ischemia/reperfusion injury and stimulation of the Rho activator, calpeptin. CONCLUSIONS The interleukin-37 level is significantly increased in acute coronary syndrome. Elevated baseline interleukin-37 levels in patients on admission are associated with poor outcomes. Thus, we propose that interleukin-37 could be a biomarker predictive of mortality in acute coronary syndrome. Moreover, this study reveals that the protective effect of interleukin-37 against atherosclerosis may involve the inhibition of ROCK activity.
Collapse
Affiliation(s)
- Tengyu Yang
- Division of Pathophysiology, Medical College, Shenzhen University, Shenzhen, Guangdong, China
| | - Fang Fang
- Division of cardiology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Li Ka Shing Institute of Health and Sciences, Institute of Vascular Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yawen Chen
- Division of Pathophysiology, Medical College, Shenzhen University, Shenzhen, Guangdong, China
| | - Jing Ma
- Division of Pathophysiology, Medical College, Shenzhen University, Shenzhen, Guangdong, China
| | - Zhaowen Xiao
- Division of Pathophysiology, Medical College, Shenzhen University, Shenzhen, Guangdong, China
| | - Songfeng Zou
- Division of Pathophysiology, Medical College, Shenzhen University, Shenzhen, Guangdong, China
| | - Na Zheng
- Division of Pathophysiology, Medical College, Shenzhen University, Shenzhen, Guangdong, China
| | - Dewen Yan
- Department of Endocrinology, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Songyan Liao
- Division of Pathophysiology, Medical College, Shenzhen University, Shenzhen, Guangdong, China
| | - Shaoyuan Chen
- Cardiology Division, Department of Medicine, The Nanshan Hostipal, Shenzhen, Guangdong, China
| | - Hongchen Fang
- Cardiology Division, Department of Medicine, The Nanshan Hostipal, Shenzhen, Guangdong, China
| | - Chekmen Yu
- Division of cardiology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Li Ka Shing Institute of Health and Sciences, Institute of Vascular Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jie Liu
- Division of Pathophysiology, Medical College, Shenzhen University, Shenzhen, Guangdong, China
| | - Ming Dong
- Division of Pathophysiology, Medical College, Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
43
|
Dolasia K, Bisht MK, Pradhan G, Udgata A, Mukhopadhyay S. TLRs/NLRs: Shaping the landscape of host immunity. Int Rev Immunol 2017; 37:3-19. [PMID: 29193992 DOI: 10.1080/08830185.2017.1397656] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Innate immune system provides the first line of defense against pathogenic organisms. It has a varied and large collection of molecules known as pattern recognition receptors (PRRs) which can tackle the pathogens promptly and effectively. Toll-like receptors (TLRs) and NOD-like receptors (NLRs) are members of the PRR family that recognize pathogen associated molecular patterns (PAMPs) and play pivotal roles to mediate defense against infections from bacteria, fungi, virus and various other pathogens. In this review, we discuss the critical roles of TLRs and NLRs in the regulation of host immune-effector functions such as cytokine production, phagosome-lysosome fusion, inflammasome activation, autophagy, antigen presentation, and B and T cell immune responses that are known to be essential for mounting a protective immune response against the pathogens. This review may be helpful to design TLRs/NLRs based immunotherapeutics to control various infections and pathophysiological disorders.
Collapse
Affiliation(s)
- Komal Dolasia
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| | - Manoj K Bisht
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| | - Gourango Pradhan
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| | - Atul Udgata
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| | - Sangita Mukhopadhyay
- a Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD) , Tuljaguda Complex, Nampally, Hyderabad , India
| |
Collapse
|
44
|
Pellefigues C, Tang SC, Schmidt A, White RF, Lamiable O, Connor LM, Ruedl C, Dobrucki J, Le Gros G, Ronchese F. Toll-Like Receptor 4, but Not Neutrophil Extracellular Traps, Promote IFN Type I Expression to Enhance Th2 Responses to Nippostrongylus brasiliensis. Front Immunol 2017; 8:1575. [PMID: 29201030 PMCID: PMC5696323 DOI: 10.3389/fimmu.2017.01575] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/02/2017] [Indexed: 12/20/2022] Open
Abstract
The induction of Th2 responses is thought to be multifactorial, and emerge from specific pathways distinct from those associated with antagonistic antibacterial or antiviral Th1 responses. Here, we show that the recognition of non-viable Nippostrongylus brasiliensis (Nb) in the skin induces a strong recruitment of monocytes and neutrophils and the release of neutrophil extracellular traps (NETs). Nb also activates toll-like receptor 4 (TLR4) signaling with expression of Ifnb transcripts in the skin and the development of an IFN type I signature on helminth antigen-bearing dendritic cells in draining lymph nodes. Co-injection of Nb together with about 10,000 Gram-negative bacteria amplified this TLR4-dependent but NET-independent IFN type I response and enhanced the development of Th2 responses. Thus, a limited activation of antibacterial signaling pathways is able to boost antihelminthic responses, suggesting a role for bacterial sensing in the optimal induction of Th2 immunity.
Collapse
Affiliation(s)
| | | | - Alfonso Schmidt
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Ruby F White
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | - Lisa M Connor
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Jurek Dobrucki
- Faculty of Biochemistry, Biophysics and Biotechnology, Department of Cell Biophysics, Jagiellonian University, Kraków, Poland
| | - Graham Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
45
|
Asalla S, Mohareer K, Banerjee S. Small Molecule Mediated Restoration of Mitochondrial Function Augments Anti-Mycobacterial Activity of Human Macrophages Subjected to Cholesterol Induced Asymptomatic Dyslipidemia. Front Cell Infect Microbiol 2017; 7:439. [PMID: 29067283 PMCID: PMC5641336 DOI: 10.3389/fcimb.2017.00439] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/25/2017] [Indexed: 12/15/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb) infection manifests into tuberculosis (TB) in a small fraction of the infected population that comprises the TB susceptible group. Identifying the factors potentiating susceptibility to TB persistence is one of the prime agenda of TB control programs. Recently, WHO recognized diabetes as a risk factor for TB disease progression. The closely related pathological state of metabolic imbalance, dyslipidemia, is yet another emerging risk factor involving deregulation in host immune responses. While high cholesterol levels are clinically proven condition for perturbations in cardiac health, a significant fraction of population these days suffer from borderline risk cholesterol profiles. This apparently healthy population is susceptible to various health risks placing them in the "pre-disease" range. Our study focuses on determining the role of such asymptomatic dyslipidemia as a potential risk factor for susceptibility to TB persistence. Macrophages exposed to sub-pathological levels of cholesterol for chronic period, besides impaired release of TNF-α, could not clear intracellular pathogenic mycobacteria effectively as compared to the unexposed cells. These cells also allowed persistence of opportunistic mycobacterial infection by M. avium and M. bovis BCG, indicating highly compromised immune response. The cholesterol-treated macrophages developed a foamy phenotype with a significant increase in intracellular lipid-bodies prior to M.tb infection, potentially contributing to pre-disease state for tuberculosis infection. The foamy phenotype, known to support M.tb infection, increased several fold upon infection in these cells. Additionally, mitochondrial morphology and function were perturbed, more so during infection in cholesterol treated cells. Pharmacological supplementation with small molecule M1 that restored mitochondrial structural and functional integrity limited M.tb survival more effectively in cholesterol exposed macrophages. Mechanistically, M1 molecule promoted clearance of mycobacteria by reducing total cellular lipid content and restoring mitochondrial morphology and function to its steady state. We further supported our observations by infection assays in PBMC-derived macrophages from clinically healthy volunteers with borderline risk cholesterol profiles. With these observations, we propose that prolonged exposure to sub-pathological cholesterol can lead to asymptomatic susceptibility to M.tb persistence. Use of small molecules like M1 sets yet another strategy for host-directed therapy where re-functioning of mitochondria in cholesterol abused macrophages can improve M.tb clearance.
Collapse
Affiliation(s)
- Suman Asalla
- Molecular Pathogenesis Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Krishnaveni Mohareer
- Molecular Pathogenesis Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Sharmistha Banerjee
- Molecular Pathogenesis Laboratory, Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
46
|
Mukherjee S, Mukherjee S, Bhattacharya S, Sinha Babu SP. Surface proteins of Setaria cervi induce inflammation in macrophage through Toll-like receptor 4 (TLR4)-mediated signalling pathway. Parasite Immunol 2017; 39. [PMID: 27659561 DOI: 10.1111/pim.12389] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 09/19/2016] [Indexed: 12/25/2022]
Abstract
Lymphatic filariasis is a vectorborne parasitic disease that results in morbidities, disabilities and socio-economic loss each year globally. Inflammatory consequences associated with any form of filariasis have drawn special attention. However, the molecular insight behind the inflammation of host macrophage (MФ) is considered as one of the shaded areas in filarial research. Herein, major emphasis was given to study the signalling pathway of MФ inflammation induced by surface proteins (SPs) of filarial parasite through in vitro and in vivo approaches. Twenty-four hours of in vitro stimulation of Raw MФs with endotoxin-free SPs of Setaria cervi resulted in the secretion of pro-inflammatory cytokines (TNF-α and IL-1β) that revealed induction of inflammation, which was found to be elicited from classical NF-кB activation. Moreover, this NF-кB activation was found to be signalled from TLR4 and mediated by the downstream signalling intermediates, viz. MyD88, pTAK1 and NEMO. In vivo studies in adult Wistar rats, experimentally injected with SPs, clearly supported the outcomes of in vitro experiments by showing higher degree of inflammation rather classical activation of the peritoneal MФs. Therefore, SPs from S. cervi cuticle could be responsible for the induction of pro-inflammatory response in MФ, which appears to be propagated through TLR4-NF-кB route.
Collapse
Affiliation(s)
- Su Mukherjee
- Department of Zoology, Centre for Advanced Studies, Visva-Bharati University, Santiniketan, West Bengal, India
| | - Sa Mukherjee
- Department of Zoology, Centre for Advanced Studies, Visva-Bharati University, Santiniketan, West Bengal, India
| | - S Bhattacharya
- Department of Zoology, Centre for Advanced Studies, Visva-Bharati University, Santiniketan, West Bengal, India
| | - S P Sinha Babu
- Department of Zoology, Centre for Advanced Studies, Visva-Bharati University, Santiniketan, West Bengal, India
| |
Collapse
|
47
|
Mukherjee S, Mukherjee S, Maiti TK, Bhattacharya S, Sinha Babu SP. A Novel Ligand of Toll-like Receptor 4 From the Sheath of Wuchereria bancrofti Microfilaria Induces Proinflammatory Response in Macrophages. J Infect Dis 2017; 215:954-965. [PMID: 28453850 DOI: 10.1093/infdis/jix067] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/28/2017] [Indexed: 11/13/2022] Open
Abstract
Background Lymphatic filariasis, frequently caused from Wuchereria bancrofti infection, is endemic in several parts of the globe and responsible for human health problems and socioeconomic loss to a large extent. Inflammatory consequences originating from host-parasite interaction play a major role in the disease pathology and allied complications. The identity of the key mediator of this process is yet unknown in filarial research. Methods Microfilarial protein (MfP) was isolated from the sheath of W. bancrofti microfilariae through ultrafiltration, followed by chromatographic separation. Expression of signaling molecules was studied by enzyme-linked immunosorbent assay and immunoblotting. Binding of MfP to Toll-like receptor 4 (TLR4) was determined by co-immunoprecipitation, fluorescein isothiocyanate-probing, and surface plasmon resonance analysis. Results We found that MfP (approximately 70 kDa) binds to macrophage-TLR4 and triggers nuclear factor kappa beta activation that upregulates secretion of proinflammatory cytokines. Microfilarial protein failed to induce inflammation in either TLRKO macrophage or macrophage treated with TLR4 inhibitor, indicating that MfP acts through TLR4. We have also detected phenotypic transformation of macrophages from anti-inflammatory (M2) to proinflammatory (M1) subtype after incubation with MfP. Conclusions Microfilarial protein appears to be a new ligand of TLR4 from W. bancrofti. Determination of its functional attributions in the host-parasite relationship, especially immunopathogenesis of filarial infection, may improve our understanding.
Collapse
Affiliation(s)
- Suprabhat Mukherjee
- Department of Zoology (Centre for Advanced Studies), Visva-Bharati University, West Bengal, India
| | - Sandip Mukherjee
- Department of Zoology (Centre for Advanced Studies), Visva-Bharati University, West Bengal, India
| | - Tushar K Maiti
- Regional Centre for Biotechnology, Haryana (NCR Delhi), India
| | - Samir Bhattacharya
- Department of Zoology (Centre for Advanced Studies), Visva-Bharati University, West Bengal, India
| | - Santi P Sinha Babu
- Department of Zoology (Centre for Advanced Studies), Visva-Bharati University, West Bengal, India
| |
Collapse
|
48
|
Chen G, Xu Y, Jing J, Mackie B, Zheng X, Zhang X, Wang J, Li X. The anti-sepsis activity of the components of Huanglian Jiedu Decoction with high lipid A-binding affinity. Int Immunopharmacol 2017; 46:87-96. [PMID: 28278436 DOI: 10.1016/j.intimp.2017.02.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/09/2017] [Accepted: 02/27/2017] [Indexed: 12/11/2022]
Abstract
Huanglian Jiedu Decoction (HJD), one of the classic recipes for relieving toxicity and fever, is a common method for treating sepsis in China. However, the effective components of HJD have not yet been identified. This experiment was carried out to elucidate the effective components of HJD against sepsis. Thus, seven fractions from HJD were tested using a biosensor to test their affinity for lipid A. The components obtained that had high lipid A-binding fractions were further separated, and their affinities to lipid A were assessed with the aid of a biosensor. The levels of LPS in the blood were measured, and pathology experiments were conducted. The LPS levels and mRNA expression analysis of TNF-α and IL-6 of the cell supernatant and animal tissue were evaluated to investigate the molecular mechanisms. Palmatine showed the highest affinity to lipid A and was evaluated by in vitro and in vivo experiments. The results of the in vitro and in vivo experiments indicated that the levels of LPS, TNF-α and IL-6 of the palmatine group were significantly lower than those of the sepsis model group (p<0.01). The group treated with palmatine showed strong neutralizing LPS activity in vivo. The palmatine group exhibited stronger protective activity on vital organs compared to the LPS-induced animal model. This verifies that HJD is a viable treatment option for sepsis given that there are multiple components in HJD that neutralize LPS, decrease the release of IL-6 and TNF-α induced by LPS, and protect vital organs.
Collapse
Affiliation(s)
- Guirong Chen
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD port, Dalian 116600, China
| | - Yubin Xu
- Key Laboratory of Biological Invasions and Global Changes, College of Biological Science and Technology, Shenyang Agricultural University, Shenyang 110161, China.
| | - Jing Jing
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD port, Dalian 116600, China
| | - Brianna Mackie
- Department of Medicinal Chemistry, Virginia Commonwealth University, 23219, USA
| | - Xinchuan Zheng
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xu Zhang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD port, Dalian 116600, China
| | - Jing Wang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD port, Dalian 116600, China
| | - Xuetao Li
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD port, Dalian 116600, China
| |
Collapse
|
49
|
Davydova VN, Kalitnik AA, Markov PA, Volod’ko AV, Popov SV, Ermak IM. Cytokine-inducing and anti-inflammatory activity of chitosan and its low-molecular derivative. APPL BIOCHEM MICRO+ 2016. [DOI: 10.1134/s0003683816050070] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
Sheela B, George G, Mandagini G, Appukuttan PS. Plasma anti-α-galactoside antibody mediates lipoprotein(a) binding to macrophages. Glycoconj J 2016; 33:953-961. [DOI: 10.1007/s10719-016-9713-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/01/2016] [Accepted: 07/12/2016] [Indexed: 01/27/2023]
|