1
|
Kumbhojkar N, Mitragotri S. Activated neutrophils: A next generation cellular immunotherapy. Bioeng Transl Med 2025; 10:e10704. [PMID: 39801751 PMCID: PMC11711228 DOI: 10.1002/btm2.10704] [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: 03/02/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 01/16/2025] Open
Abstract
Cell therapies are at the forefront of novel therapeutics. Neutrophils, despite being the most populous immune cells in human blood circulation, are not considered a viable option for cellular therapies because of their short lifespan and poor understanding of their role in the pathophysiology of various diseases. In inflammatory conditions, neutrophils exhibit an activated phenotype. Activation brings about significant changes to neutrophil biology such as increased lifespan, inflammatory cytokine secretion, and enhanced effector functions. Activated neutrophils also possess the potential to stimulate the downstream immune response and are described as essential effectors in the immune response to tumors. This makes activated neutrophils an interesting candidate for cell therapies. Here, we review the biology of activated neutrophils in detail. We discuss the different ways neutrophils can be activated and the effect they have on other immune cells for stimulation of downstream immune response. We review the conditions where activated neutrophil therapy can be therapeutically beneficial and discuss the challenges associated with their eventual translation. Overall, this review summarizes the current state of understanding of neutrophil-based immunotherapies and their clinical potential.
Collapse
Affiliation(s)
- Ninad Kumbhojkar
- Harvard John A. Paulson School of Engineering and Applied SciencesAllstonMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Samir Mitragotri
- Harvard John A. Paulson School of Engineering and Applied SciencesAllstonMassachusettsUSA
- Wyss Institute for Biologically Inspired EngineeringBostonMassachusettsUSA
| |
Collapse
|
2
|
Wang T, Zhang Y. Mechanisms and therapeutic targets of carbon monoxide poisoning: A focus on reactive oxygen species. Chem Biol Interact 2024; 403:111223. [PMID: 39237073 DOI: 10.1016/j.cbi.2024.111223] [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: 04/28/2024] [Revised: 08/08/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Carbon monoxide (CO) poisoning presents a substantial public health challenge that necessitates the identification of its pathological mechanisms and therapeutic targets. CO toxicity arises from tissue hypoxia-ischemia secondary to carboxyhemoglobin formation, and cellular damage mediated by CO at the cellular level. The mitochondria are the major targets of neuronal damage caused by CO. Under normal physiological conditions, mitochondria produce reactive oxygen species (ROS), which are byproducts of aerobic metabolism. While low ROS levels are crucial for essential cellular functions, including signal transduction, differentiation, responses to hypoxia and immunity, transcriptional regulation, and autophagy, excess ROS become pathological and exacerbate CO poisoning. This review presents the evidence of elevated ROS being associated with the progression of CO poisoning. Antioxidant treatments targeting ROS removal have been proven effective in mitigating CO poisoning, underscoring their therapeutic potential. In this review, we highlight the latest advances in the understanding of the role and the clinical implications of ROS in CO poisoning. We focus on cellular sources of ROS, the molecular mechanisms underlying mitochondrial oxidative stress, and potential therapeutic strategies for targeting ROS in CO poisoning.
Collapse
Affiliation(s)
- Tianhong Wang
- Department of Neurology, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Yanli Zhang
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
3
|
Choi YJ, Kim Y, Hwang S. Role of Neutrophils in the Development of Steatotic Liver Disease. Semin Liver Dis 2024; 44:300-318. [PMID: 39117322 DOI: 10.1055/s-0044-1789207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
This review explores the biological aspects of neutrophils, their contributions to the development of steatotic liver disease, and their potential as therapeutic targets for the disease. Although alcohol-associated and metabolic dysfunction-associated liver diseases originate from distinct etiological factors, the two diseases frequently share excessive lipid accumulation as a common contributor to their pathogenesis, thereby classifying them as types of steatotic liver disease. Dysregulated lipid deposition in the liver induces hepatic injury, triggering the activation of the innate immunity, partially through neutrophil recruitment. Traditionally recognized for their role in microbial clearance, neutrophils have recently garnered attention for their involvement in sterile inflammation, a pivotal component of steatotic liver disease pathogenesis. In conclusion, technological innovations, including single-cell RNA sequencing, have gradually disclosed the existence of various neutrophil subsets; however, how the distinct subsets of neutrophil population contribute differentially to the development of steatotic liver disease remains unclear.
Collapse
Affiliation(s)
- You-Jin Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Republic of Korea
| | - Yeonsoo Kim
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
4
|
Chen SH, Chen CH, Lin HC, Yeh SA, Hwang TL, Chen PJ. Drug repurposing of cyclin-dependent kinase inhibitors for neutrophilic acute respiratory distress syndrome and psoriasis. J Adv Res 2024:S2090-1232(24)00310-2. [PMID: 39089617 DOI: 10.1016/j.jare.2024.07.026] [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: 05/05/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Neutrophilic inflammation, characterized by dysregulated neutrophil activation, triggers a variety of inflammatory responses such as chemotactic infiltration, oxidative bursts, degranulation, neutrophil extracellular traps (NETs) formation, and delayed turnover. This type of inflammation is pivotal in the pathogenesis of acute respiratory distress syndrome (ARDS) and psoriasis. Despite current treatments, managing neutrophil-associated inflammatory symptoms remains a significant challenge. AIM OF REVIEW This review emphasizes the role of cyclin-dependent kinases (CDKs) in neutrophil activation and inflammation. It aims to highlight the therapeutic potential of repurposing CDK inhibitors to manage neutrophilic inflammation, particularly in ARDS and psoriasis. Additionally, it discusses the necessary precautions for the clinical application of these inhibitors due to potential off-target effects and the need for dose optimization. KEY SCIENTIFIC CONCEPTS OF REVIEW CDKs regulate key neutrophilic functions, including chemotactic responses, degranulation, NET formation, and apoptosis. Repurposing CDK inhibitors, originally developed for cancer treatment, shows promise in controlling neutrophilic inflammation. Clinical anticancer drugs, palbociclib and ribociclib, have demonstrated efficacy in treating neutrophilic ARDS and psoriasis by targeting off-label pathways, phosphoinositide 3-kinase (PI3K) and phosphodiesterase 4 (PDE4), respectively. While CDK inhibitors offer promising therapeutic benefits, their clinical repurposing requires careful consideration of off-target effects and dose optimization. Further exploration and clinical trials are necessary to ensure their safety and efficacy in treating inflammatory conditions.
Collapse
Affiliation(s)
- Shun-Hua Chen
- School of Nursing, Fooyin University, Kaohsiung 831301, Taiwan.
| | - Chun-Hong Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Hsin-Chieh Lin
- Department of Chinese Medicine, E-Da Cancer Hospital, I-Shou University, Kaohsiung 824410, Taiwan; School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Shyh-An Yeh
- Medical Physics and Informatics Laboratory of Electronic Engineering and Department of Electronic Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 80778, Taiwan; Department of Medical Imaging and Radiological Sciences, I-Shou University, Kaohsiung 824410, Taiwan; Department of Radiation Oncology, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan.
| | - Tsong-Long Hwang
- Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333324, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333324, Taiwan.
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung 824410, Taiwan; Graduate Institute of Medicine, College of Medicine, I-Shou University, Kaohsiung 824410, Taiwan.
| |
Collapse
|
5
|
Quinn M, Zhang RYK, Bello I, Rye KA, Thomas SR. Myeloperoxidase as a Promising Therapeutic Target after Myocardial Infarction. Antioxidants (Basel) 2024; 13:788. [PMID: 39061857 PMCID: PMC11274265 DOI: 10.3390/antiox13070788] [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: 03/30/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Coronary artery disease (CAD) and myocardial infarction (MI) remain leading causes of death and disability worldwide. CAD begins with the formation of atherosclerotic plaques within the intimal layer of the coronary arteries, a process driven by persistent arterial inflammation and oxidation. Myeloperoxidase (MPO), a mammalian haem peroxidase enzyme primarily expressed within neutrophils and monocytes, has been increasingly recognised as a key pro-inflammatory and oxidative enzyme promoting the development of vulnerable coronary atherosclerotic plaques that are prone to rupture, and can precipitate a MI. Mounting evidence also implicates a pathogenic role for MPO in the inflammatory process that follows a MI, which is characterised by the rapid infiltration of activated neutrophils into the damaged myocardium and the release of MPO. Excessive and persistent cardiac inflammation impairs normal cardiac healing post-MI, resulting in adverse cardiac outcomes and poorer long-term cardiac function, and eventually heart failure. This review summarises the evidence for MPO as a significant oxidative enzyme contributing to the inappropriate inflammatory responses driving the progression of CAD and poor cardiac healing after a MI. It also details the proposed mechanisms underlying MPO's pathogenic actions and explores MPO as a novel therapeutic target for the treatment of unstable CAD and cardiac damage post-MI.
Collapse
Affiliation(s)
| | | | | | | | - Shane R. Thomas
- Cardiometabolic Disease Research Group, School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
6
|
Cartwright IM, Zhou L, Koch SD, Welch N, Zakharov D, Callahan R, Steiner CA, Gerich ME, Onyiah JC, Colgan SP. Chlorination of epithelial tight junction proteins by neutrophil myeloperoxidase promotes barrier dysfunction and mucosal inflammation. JCI Insight 2024; 9:e178525. [PMID: 39133648 PMCID: PMC11383587 DOI: 10.1172/jci.insight.178525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/31/2024] [Indexed: 08/16/2024] Open
Abstract
Neutrophils (polymorphonuclear leukocytes, PMNs) comprise a major component of the immune cell infiltrate during acute mucosal inflammation and have an important role in molding the inflammatory tissue environment. While PMNs are essential to clearance of invading microbes, the major PMN antimicrobial enzyme myeloperoxidase (MPO) can also promote bystander tissue damage. We hypothesized that blocking MPO would attenuate acute colitis and prevent the development of chronic colitis by limiting bystander tissue damage. Using the acute and chronic dextran sodium sulfate model of murine colitis, we demonstrated that MPO-deficient mice experienced less inflammation and more rapidly resolved colitis relative to wild-type controls. Mechanistic studies demonstrated that activated MPO disrupted intestinal epithelial barrier function through the dysregulation of the epithelial tight junction proteins. Our findings revealed that activated MPO chlorinates tyrosine within several tight junction proteins, thereby promoting tight junction mislocalization and dysfunction. These observations in cell models and in murine colitis were validated in human intestinal biopsies from individuals with ulcerative colitis and revealed a strong correlation between disease severity (Mayo score) and tissue chlorinated tyrosine levels. In summary, these findings implicate MPO as a viable therapeutic target to limit bystander tissue damage and preserve mucosal barrier function during inflammation.
Collapse
Affiliation(s)
- Ian M Cartwright
- Mucosal Inflammation Program and
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| | - Liheng Zhou
- Mucosal Inflammation Program and
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Samuel D Koch
- Mucosal Inflammation Program and
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Nichole Welch
- Mucosal Inflammation Program and
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Daniel Zakharov
- School of Medicine, University College Dublin, Dublin, United Kingdom
| | - Rosemary Callahan
- Mucosal Inflammation Program and
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Calen A Steiner
- Mucosal Inflammation Program and
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mark E Gerich
- Mucosal Inflammation Program and
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Joseph C Onyiah
- Mucosal Inflammation Program and
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| | - Sean P Colgan
- Mucosal Inflammation Program and
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| |
Collapse
|
7
|
Giotakis AI, Dudas J, Glueckert R, Buechel E, Riechelmann H. Identification of neutrophils and eosinophils in upper airway mucosa with immunofluorescence multiplex image cytometry. Histochem Cell Biol 2024:10.1007/s00418-024-02284-y. [PMID: 38600336 DOI: 10.1007/s00418-024-02284-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2024] [Indexed: 04/12/2024]
Abstract
Characterization of inflammation in chronic rhinosinusitis with (CRSwNP) and without nasal polyps (CRSsNP) is an ongoing research process. To overcome limitations of current cytologic techniques, we investigated whether immunofluorescence multiplex image cytometry could quantify intact neutrophils, eosinophils, and other immune cells in solid upper airway mucosa. We used a four-channel immunofluorescence-microscopy technique for the simultaneous detection of the leukocyte marker CD45, the neutrophil marker myeloperoxidase, two eosinophil markers, i.e., major basic protein and eosinophil peroxidase, and DAPI (4',6-diamidin-2-phenylindole), in formalin-fixed paraffin-embedded upper airway tissue samples of patients with CRSwNP and CRSsNP, as well as of patients free of CRS with inferior turbinate hypertrophy (controls). Image acquisition and analysis were performed with TissueFAXS and StrataQuest (TissueGnostics, Vienna, Austria), respectively. Positive and negative immunostaining were differentiated with a specific fluorescence signal/background signal ratio. Isotype controls were used as negative controls. In six controls, nine patients with CRSsNP, and 11 patients with CRSwNP, the median area scanned and median cell count per patient were 14.2 mm2 and 34,356, respectively. In CRSwNP, the number of eosinophils was three times higher (23%) than that of neutrophils (7%). Three times more immune cells were encountered in CRSwNP (33%) compared to CRSsNP (11%). In controls, inflammation was balanced between the epithelial layer and lamina propria, in contrast to CRS (three times more pronounced inflammation in the lamina propria). The quantification of intact neutrophils, eosinophils, and other immune cells in solid tissue with undisrupted architecture seems feasible with immunofluorescence multiplex image cytometry.
Collapse
Affiliation(s)
- Aris I Giotakis
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - József Dudas
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Rudolf Glueckert
- University Clinics Innsbruck, Tirol Kliniken, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Elias Buechel
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Herbert Riechelmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| |
Collapse
|
8
|
Zafar N, Khan MA. Effects of Dietary Zinc on Growth, Haematological Indices, Digestive Enzyme Activity, Tissue Mineralization, Antioxidant and Immune Status of Fingerling Heteropneustes fossilis. Biol Trace Elem Res 2024; 202:1249-1263. [PMID: 37392362 DOI: 10.1007/s12011-023-03749-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/24/2023] [Indexed: 07/03/2023]
Abstract
A 12 week feeding trial was conducted to evaluate the effects of dietary zinc levels on Heteropneustes fossilis. Triplicate groups of fish were fed isoproteic (CP; 400 g/kg) and isocaloric (GE; 17.89 kJ/g) diets increasing levels of zinc (0, 5, 10, 15, 20, 25, 30 mg/kg) achieved by supplementing zinc sulphate heptahydrate to basal diet. Analysed concentrations of zinc in diets were 10.68, 15.83, 21.34, 26.74, 30.61, 34.91 and 41.34 mg/kg. Growth indices increased linearly (P<0.05) up to 26.74 mg/kg Zn. The protein and ash content of whole body also improved significantly up to 26.74 mg/kg Zn. Whole body fat content showed inverse pattern. Haematological parameters also showed an improving trend with the increase in dietary zinc up to 26.74 mg/kg and then levelled off. Activities of antioxidant enzymes were improved with the increase in dietary zinc level up to 26.74 mg/kg followed by no significant change (P>0.05). Serum lysozyme activity also exhibited the similar pattern. Immune response in terms of the activities of lysozyme, alkaline phosphatase and myeloperoxidase was also improved with the increase in dietary zinc levels up to 26.74 mg/kg. Dietary zinc levels affected significantly the whole body as well as vertebrae mineralization. Broken-line regression analysis of weight gain, vertebrae zinc activity, serum superoxide dismutase and protease activity against increasing amounts of dietary zinc revealed that the inclusion of zinc in diet in the range of 26.82-29.84 mg/kg is optimum for growth, haematological indices, antioxidant status, immune response and tissue mineralization in fingerling H. fossilis. The information obtained from present study would be helpful in formulating the zinc-balanced commercial feeds to improve the growth and health status of this important fish, thus contributing to aquaculture production and strengthening the food security.
Collapse
Affiliation(s)
- Noorin Zafar
- Fish Nutrition Research Laboratory, Aligarh Muslim University, Aligarh, India
- Department of Zoology, Aligarh Muslim University, Aligarh, 202 002, India
| | - Mukhtar A Khan
- Fish Nutrition Research Laboratory, Aligarh Muslim University, Aligarh, India.
- Department of Zoology, Aligarh Muslim University, Aligarh, 202 002, India.
| |
Collapse
|
9
|
Rizo-Téllez SA, Filep JG. Beyond host defense and tissue injury: the emerging role of neutrophils in tissue repair. Am J Physiol Cell Physiol 2024; 326:C661-C683. [PMID: 38189129 PMCID: PMC11193466 DOI: 10.1152/ajpcell.00652.2023] [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: 11/29/2023] [Revised: 12/31/2023] [Accepted: 12/31/2023] [Indexed: 01/09/2024]
Abstract
Neutrophils, the most abundant immune cells in human blood, play a fundamental role in host defense against invading pathogens and tissue injury. Neutrophils carry potentially lethal weaponry to the affected site. Inadvertent and perpetual neutrophil activation could lead to nonresolving inflammation and tissue damage, a unifying mechanism of many common diseases. The prevailing view emphasizes the dichotomy of their function, host defense versus tissue damage. However, tissue injury may also persist during neutropenia, which is associated with disease severity and poor outcome. Numerous studies highlight neutrophil phenotypic heterogeneity and functional versatility, indicating that neutrophils play more complex roles than previously thought. Emerging evidence indicates that neutrophils actively orchestrate resolution of inflammation and tissue repair and facilitate return to homeostasis. Thus, neutrophils mobilize multiple mechanisms to limit the inflammatory reaction, assure debris removal, matrix remodeling, cytokine scavenging, macrophage reprogramming, and angiogenesis. In this review, we will summarize the homeostatic and tissue-reparative functions and mechanisms of neutrophils across organs. We will also discuss how the healing power of neutrophils might be harnessed to develop novel resolution and repair-promoting therapies while maintaining their defense functions.
Collapse
Affiliation(s)
- Salma A Rizo-Téllez
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - János G Filep
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| |
Collapse
|
10
|
Li G, Park HJ, Suh JH, Choi HS. 7-Ketocholesterol plays a key role in cholesterol-induced hepatitis via macrophage and neutrophil infiltration. J Nutr Biochem 2024; 125:109552. [PMID: 38134972 DOI: 10.1016/j.jnutbio.2023.109552] [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: 07/23/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
This study sought to explore the role of 7-ketocholesterol (7-KC) in liver damage caused by high cholesterol intake and its potential pathological mechanism in mice. Our in vivo findings indicated that mice fed a high-cholesterol diet had elevated serum levels of 7-KC, accompanied by liver injury and inflammation, similar to human nonalcoholic steatohepatitis. Furthermore, the high-cholesterol diet induced neutrophil infiltration, which played a critical role in liver damage through myeloperoxidase (MPO) activity. Upon stimulation with 7-KC, macrophages exhibited increased expression of C-X-C motif chemokine ligand 1 (CXCL1) and CXCL2, as well as ATP-binding cassette transporter A1 (ABCA1) and ABCG1. Hepatocytes, on the other hand, exhibited increased expression of CXCL2 and ABCG1. The infiltration of neutrophils in the liver was primarily caused by CXCL1 and CXCL2, resulting in hepatocyte cell death due to elevated MPO activity. Our data also revealed that the activation of macrophages by 7-KC via ABCA1 or ABCG1 was not associated with lipid accumulation. Collectively, these findings suggest that high cholesterol-induced hepatitis in mice involves, at least partially, the recruitment of neutrophils to the liver by 7-KC-activated macrophages. This is mediated by increased expression of CXCL1 and CXCL2 through ABCA1 or ABCG1, which act as 7-KC efflux transporters. Additionally, hepatocytes contribute to this process by increased expression of CXCL2 through ABCG1. Therefore, our findings suggest that 7-KC may play a role in high cholesterol-induced hepatitis in mice by activating macrophages and hepatocytes, ultimately leading to neutrophil infiltration.
Collapse
Affiliation(s)
- Guoen Li
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Hyun-Jung Park
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Jae-Hee Suh
- Department of Pathology, Ulsan University Hospital, Ulsan, Korea
| | - Hye-Seon Choi
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea.
| |
Collapse
|
11
|
Lassnig S, Hennig-Pauka I, Bonilla MC, Mörgelin M, Imker R, von Köckritz-Blickwede M, de Buhr N. Impact of bronchoalveolar lavage from influenza A virus diseased pigs on neutrophil functions and growth of co-infecting pathogenic bacteria. Front Immunol 2024; 15:1325269. [PMID: 38449874 PMCID: PMC10914936 DOI: 10.3389/fimmu.2024.1325269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/26/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Influenza A viruses (IAVs) infect the respiratory tract of mainly humans, poultry, and pigs. Co-infections with pathogenic lung bacteria are a common event and contribute to the severity of disease progression. Neutrophils are a major cell type of the innate immune system and are rapidly recruited to the site of infection. They have several effector functions to fight invading pathogens such as the secretion of reactive oxygen species (ROS) or the release of neutrophil extracellular traps (NETs). NETs are known to promote the growth of Pasteurellaceae bacteria, especially if degraded by nucleases. Methods In this study, bronchoalveolar lavage fluid (BALF) from 45 field-infected pigs was analyzed for 1) NET markers, 2) influence on growth of lung bacteria, and 3) impact on neutrophil functions. BALF samples from 21 IAV-positive pigs and 24 lung diseased but IAV-negative pigs were compared. Results Here, we show that neutrophils in the lungs of IAV-positive pigs release vesicular NETs. Several NET markers were increased in the BALF of IAV-positive pigs compared with the BALF from IAV-negative pigs. The amount of NET markers positively correlated with the viral load of the IAV infection. Interestingly, the BALF of IAV-positive pigs enhanced the growth of bacteria belonging to the family of Pasteurellaceae as potential coinfecting bacteria. These effects were weaker with the BALF derived from IAV-negative pigs with other lung infections. The intensity of oxidative burst in neutrophils was significantly decreased by BALF from IAVpositive pigs, indicating impaired antimicrobial activity of neutrophils. Finally, the lung milieu reflected by IAV-positive BALF does not enable neutrophils to kill Actinobacillus pleuropneumoniae but rather enhances its growth. Discussion In summary, our data show that an IAV infection is affecting neutrophil functions, in particular the release of NETs and ROS. Furthermore, IAV infection seems to provide growth-enhancing factors for especially coinfecting Pasteurellaceae and reduces the killing efficiency of neutrophils.
Collapse
Affiliation(s)
- Simon Lassnig
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Isabel Hennig-Pauka
- Clinic for Swine, Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine Hannover, Hannover, Germany
- Field Station for Epidemiology, University of Veterinary Medicine Hannover, Bakum, Germany
| | - Marta C. Bonilla
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Rabea Imker
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicole de Buhr
- Institute of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
12
|
Basnet A, Landreth KM, Nohoesu R, Santiago SP, Geldenhuys WJ, Boone BA, Liu TW. Targeting myeloperoxidase limits myeloid cell immunosuppression enhancing immune checkpoint therapy for pancreatic cancer. Cancer Immunol Immunother 2024; 73:57. [PMID: 38367056 PMCID: PMC10874341 DOI: 10.1007/s00262-024-03647-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/29/2024] [Indexed: 02/19/2024]
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease characterized by an extreme resistance to current therapies, including immune checkpoint therapy. The limited success of immunotherapies can be attributed to a highly immunosuppressive pancreatic cancer microenvironment characterized by an extensive infiltration of immune suppressing myeloid cells. While there are several pathways through which myeloid cells contribute to immunosuppression, one important mechanism is the increased production of reactive oxygen species. Here, we evaluated the contribution of myeloperoxidase, a myeloid-lineage restricted enzyme and primary source of reactive oxygen species, to regulate immune checkpoint therapy response in preclinical pancreatic cancer models. We compared treatment outcome, immune composition and characterized myeloid cells using wild-type, myeloperoxidase-deficient, and myeloperoxidase inhibitor treated wild-type mice using established subcutaneous pancreatic cancer models. Loss of host myeloperoxidase and pharmacological inhibition of myeloperoxidase in combination with immune checkpoint therapy significantly delayed tumor growth. The tumor microenvironment and systemic immune landscape demonstrated significant decreases in myeloid cells, exhausted T cells and T regulatory cell subsets when myeloperoxidase was deficient. Loss of myeloperoxidase in isolated myeloid cell subsets from tumor-bearing mice resulted in decreased reactive oxygen species production and T cell suppression. These data suggest that myeloperoxidase contributes to an immunosuppressive microenvironment and immune checkpoint therapy resistance where myeloperoxidase inhibitors have the potential to enhance immunotherapy response. Repurposing myeloperoxidase specific inhibitors may provide a promising therapeutic strategy to expand therapeutic options for pancreatic cancer patients to include immunotherapies.
Collapse
Affiliation(s)
- Angisha Basnet
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Kaitlyn M Landreth
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Remi Nohoesu
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Stell P Santiago
- Department of Pathology, Anatomy and Laboratory Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Werner J Geldenhuys
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, 26506, USA
| | - Brian A Boone
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA
- Division of Surgical Oncology, Department of Surgery, West Virginia University, Morgantown, WV, 26506, USA
| | - Tracy W Liu
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA.
- WVU Cancer Institute, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
13
|
Gao X, Zhang Y, Zhu Q, Han Y, Jia R, Zhang W. Effects of myeloperoxidase on inflammatory responses with hypoxia in Citrobacter rodentium-infectious mice. Immun Inflamm Dis 2024; 12:e1157. [PMID: 38415976 PMCID: PMC10836036 DOI: 10.1002/iid3.1157] [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: 07/17/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 02/29/2024] Open
Abstract
PURPOSE Myeloperoxidase (MPO) has been identified as a mediator in various inflammatory diseases. Bacterial infection of the intestine and hypoxia can both lead to inflammatory responses, but the role of MPO in these phenomena remains unclear. METHODS By building the MPO-/- mice, we evaluated relevant inflammatory factors and tissue damage in mice with intestinal Citrobacter rodentium infection and hypoxia. The body weight and excreted microorganisms were monitored. Intestinal tissues were collected 7 days after bacterial infection under hypoxia to undergo haematoxylin-eosin staining and assess the degree of pathological damage. ELISA assays were performed to quantify the serum levels of TNF-α, IFN-γ, IL-6, and IL-1β inflammatory cytokines. PCR, WB, and IF assays were conducted to determine the expression of chemokines MCP1, MIP2, and KC in the colon and spleen. RESULTS The C. rodentium infection and hypoxia caused weight loss, intestinal colitis, and splenic inflammatory cells active proliferation in wild-type mice. MPO deficiency alleviated this phenomenon. MPO-/- mice also displayed a significant decline in bacteria clearing ability. The level of TNF-α in the serum and spleen was both lower in MPO-/- hypoxia C. rodentium-infected mice than that in wild-type mice. The chemokines expression levels of MIP2, KC, and MCP1 in the spleen and colon of each bacterial infected group were significantly increased (p < .05), while in hypoxia, the factors in the spleen and colon were decreased (p < .05). MPO deficiency was found to lower the levels of these chemokines compared with wild-type mice. CONCLUSION MPO plays an important role of the inflammatory responses in infectious enteritis and hypoxia in mice, and the loss of MPO may greatly reduce the body's inflammatory responses to fight diseases.
Collapse
Affiliation(s)
- Xiang Gao
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| | - Yu Zhang
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
| | - Qinfang Zhu
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| | - Ying Han
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| | - Ruhan Jia
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| | - Wei Zhang
- Department of Basic Medical Sciences, Medical CollegeQinghai UniversityXiningQinghaiChina
- Research Centre for High Altitude Medicine, Research Centre for High Altitude MedicineQinghai UniversityXiningQinghaiChina
- The Key Laboratory of High‐Altitude Medical Application of Qinghai ProvinceXiningQinghaiChina
| |
Collapse
|
14
|
Kim SM, Hong SA, Kim JM. Association of immunologic findings of atheromatous plaques with subsequent cardiovascular events in patients with peripheral artery disease. Sci Rep 2024; 14:469. [PMID: 38172197 PMCID: PMC10764821 DOI: 10.1038/s41598-023-50751-8] [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: 03/28/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024] Open
Abstract
Patients with peripheral artery disease (PAD) have a higher risk of cardiovascular events. We examined the histology of atheromatous plaques in the femoral artery and investigated their association with subsequent cardiovascular events in patients with PAD. Patients who underwent femoral artery endarterectomy between March 2010 and January 2021 were included. We analyzed the expression of myeloperoxidase (MPO), citrullinated histone, and programmed cell death ligand 1 (PD-L1) in femoral artery plaques by immunohistochemistry. Data on the subsequent occurrence of major adverse cardiovascular events (MACEs), major adverse limb events (MALEs), and all-cause mortality were retrospectively collected. A total of 37 patients were included. The median age was 71 (range, 42-90) years, and 25 patients (67.6%) were male. During the median follow-up of 24 months, 10 patients experienced MACEs and 16 patients had MALEs. Patients with MACEs had a higher number of MPO-stained cells (p = 0.044) and lower PD-L1 staining intensity (p = 0.021) in atheromatous plaques compared with those of patients with a stable prognosis. When the patients were grouped according to the immunologic score based on the MPO-stained cell number and PD-L1 staining intensity, those with a higher score had a significantly higher cumulative risk of MACEs (p = 0.014). The immunologic profile of excised peripheral artery plaques may be associated with future cardiovascular events in patients with PAD.
Collapse
Affiliation(s)
- Suh Min Kim
- Department of Surgery, Chung-Ang University College of Medicine, Chung-Ang University Hospital, Seoul, South Korea
| | - Soon Auck Hong
- Department of Pathology, Chung-Ang University College of Medicine, 102 Heukseok-Ro, Dongjak-Gu, Seoul, 06973, Republic of Korea.
| | - Jeong-Min Kim
- Department of Neurology, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
15
|
Casado-Bedmar M, Roy M, Berthet L, Hugot JP, Yang C, Manceau H, Peoc'h K, Chassaing B, Merlin D, Viennois E. Fecal let-7b and miR-21 directly modulate the intestinal microbiota, driving chronic inflammation. Gut Microbes 2024; 16:2394249. [PMID: 39224018 PMCID: PMC11376420 DOI: 10.1080/19490976.2024.2394249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Inflammatory bowel diseases (IBD) etiology is multifactorial. Luminal microRNAs (miRNAs) have been suspected to play a role in the promotion of chronic inflammation, but the extent to which fecal miRNAs are interacting with the intestinal ecosystem in a way that contribute to diseases, including IBD, remains unknown. Here, fecal let-7b and miR-21 were found elevated, associated with inflammation, and correlating with multiple bacteria in IBD patients and IL-10-/- mice, model of spontaneous colitis. Using an in vitro microbiota modeling system, we revealed that these two miRNAs can directly modify the composition and function of complex human microbiota, increasing their proinflammatory potential. In vivo investigations revealed that luminal increase of let-7b drastically alters the intestinal microbiota and enhances macrophages' associated proinflammatory cytokines (TNF, IL-6, and IL-1β). Such proinflammatory effects are resilient and dependent on the bacterial presence. Moreover, we identified that besides impairing the intestinal barrier function, miR-21 increases myeloperoxidase and antimicrobial peptides secretion, causing intestinal dysbiosis. More importantly, in vivo inhibition of let-7b and miR-21 with anti-miRNAs significantly improved the intestinal mucosal barrier function and promoted a healthier host-microbiota interaction in the intestinal lining, which altogether conferred protection against colitis. In summary, we provide evidence of the functional significance of fecal miRNAs in host-microbiota communication, highlighting their therapeutic potential in intestinal inflammation and dysbiosis-related conditions, such as IBD.
Collapse
Affiliation(s)
| | - Maryline Roy
- Center for Research on Inflammation, Université Paris Cité, Paris, France
| | - Louis Berthet
- Center for Research on Inflammation, Université Paris Cité, Paris, France
| | - Jean-Pierre Hugot
- Center for Research on Inflammation, Université Paris Cité, Paris, France
- Department of Pediatric Gastroenterology, Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Chunhua Yang
- Institute for Biomedical Sciences, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Hana Manceau
- Center for Research on Inflammation, Université Paris Cité, Paris, France
- Laboratory of Clinical Biochemistry, Beaujon Hospital, APHP, Clichy, France
| | - Katell Peoc'h
- Center for Research on Inflammation, Université Paris Cité, Paris, France
- Laboratory of Clinical Biochemistry, Beaujon Hospital, APHP, Clichy, France
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris, France
- CHRU Nancy, IHU Infiny, Nancy, France
| | - Didier Merlin
- Institute for Biomedical Sciences, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
- Veterans Affairs Medical Center, Decatur, GA, USA
| | - Emilie Viennois
- Center for Research on Inflammation, Université Paris Cité, Paris, France
- CHRU Nancy, IHU Infiny, Nancy, France
| |
Collapse
|
16
|
Mihalic ZN, Kloimböck T, Cosic-Mujkanovic N, Valadez-Cosmes P, Maitz K, Kindler O, Wadsack C, Heinemann A, Marsche G, Gauster M, Pollheimer J, Kargl J. Myeloperoxidase enhances the migration and invasion of human choriocarcinoma JEG-3 cells. Redox Biol 2023; 67:102885. [PMID: 37776707 PMCID: PMC10556814 DOI: 10.1016/j.redox.2023.102885] [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: 04/13/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 10/02/2023] Open
Abstract
Myeloperoxidase (MPO) is one of the most abundant proteins in neutrophil granules. It catalyzes the production of reactive oxygen species, which are important in inflammation and immune defense. MPO also binds to several proteins, lipids, and DNA to alter their function. MPO is present at the feto-maternal interface during pregnancy, where neutrophils are abundant. In this study, we determined the effect of MPO on JEG-3 human choriocarcinoma cells as a model of extravillous trophoblasts (EVTs) during early pregnancy. We found that MPO was internalized by JEG-3 cells and localized to the cytoplasm and nuclei. MPO internalization and activity enhanced JEG-3 cell migration and invasion, whereas this effect was impaired by pre-treating cells with heparin, to block cellular uptake, and MPO-activity inhibitor 4-ABAH. This study identifies a novel mechanism for the effect of MPO on EVT function during normal pregnancy and suggests a potential role of MPO in abnormal pregnancies.
Collapse
Affiliation(s)
- Z N Mihalic
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - T Kloimböck
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - N Cosic-Mujkanovic
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - P Valadez-Cosmes
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - K Maitz
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - O Kindler
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - C Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria; BioTechMed-Graz, 8010, Graz, Austria
| | - A Heinemann
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria; BioTechMed-Graz, 8010, Graz, Austria
| | - G Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria; BioTechMed-Graz, 8010, Graz, Austria
| | - M Gauster
- Division of Cell Biology, Histology and Embryology, Medical University of Graz, Austria
| | - J Pollheimer
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Maternal-Fetal Immunology Group, Medical University of Vienna, Austria
| | - J Kargl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria; BioTechMed-Graz, 8010, Graz, Austria.
| |
Collapse
|
17
|
Jin L, Wang F, Wang X, Harvey BP, Bi Y, Hu C, Cui B, Darcy AT, Maull JW, Phillips BR, Kim Y, Jenkins GJ, Sornasse TR, Tian Y. Identification of Plasma Biomarkers from Rheumatoid Arthritis Patients Using an Optimized Sequential Window Acquisition of All THeoretical Mass Spectra (SWATH) Proteomics Workflow. Proteomes 2023; 11:32. [PMID: 37873874 PMCID: PMC10594463 DOI: 10.3390/proteomes11040032] [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: 08/16/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/25/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune and inflammatory disease. Plasma biomarkers are critical for understanding disease mechanisms, treatment effects, and diagnosis. Mass spectrometry-based proteomics is a powerful tool for unbiased biomarker discovery. However, plasma proteomics is significantly hampered by signal interference from high-abundance proteins, low overall protein coverage, and high levels of missing data from data-dependent acquisition (DDA). To achieve quantitative proteomics analysis for plasma samples with a balance of throughput, performance, and cost, we developed a workflow incorporating plate-based high abundance protein depletion and sample preparation, comprehensive peptide spectral library building, and data-independent acquisition (DIA) SWATH mass spectrometry-based methodology. In this study, we analyzed plasma samples from both RA patients and healthy donors. The results showed that the new workflow performance exceeded that of the current state-of-the-art depletion-based plasma proteomic platforms in terms of both data quality and proteome coverage. Proteins from biological processes related to the activation of systemic inflammation, suppression of platelet function, and loss of muscle mass were enriched and differentially expressed in RA. Some plasma proteins, particularly acute-phase reactant proteins, showed great power to distinguish between RA patients and healthy donors. Moreover, protein isoforms in the plasma were also analyzed, providing even deeper proteome coverage. This workflow can serve as a basis for further application in discovering plasma biomarkers of other diseases.
Collapse
Affiliation(s)
- Liang Jin
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Fei Wang
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Xue Wang
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Bohdan P. Harvey
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Yingtao Bi
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Chenqi Hu
- DMPK, Takeda Development Center Americas Inc., Cambridge, MA 02142, USA; (C.H.)
| | - Baoliang Cui
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Anhdao T. Darcy
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - John W. Maull
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Ben R. Phillips
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Youngjae Kim
- DMPK, Takeda Development Center Americas Inc., Cambridge, MA 02142, USA; (C.H.)
| | - Gary J. Jenkins
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Thierry R. Sornasse
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| | - Yu Tian
- Research & Development, AbbVie, North Chicago, IL 60064, USA; (L.J.); (B.P.H.); (B.C.); (A.T.D.); (J.W.M.); (T.R.S.)
| |
Collapse
|
18
|
Asejeje FO, Akinola KD, Abiola MA. Sodium benzoate exacerbates hepatic oxidative stress and inflammation in lipopolysaccharide-induced liver injury in rats. Immunopharmacol Immunotoxicol 2023; 45:558-564. [PMID: 36927185 DOI: 10.1080/08923973.2023.2191818] [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: 08/01/2022] [Accepted: 03/12/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Liver damage is a global health concern associated with a high mortality rate. Sodium benzoate (SB) is a widely used preservative in the food industry with a wide range of applications. However, there's a lack of scientific reports on its effect on lipopolysaccharide-induced hepatic dysfunction. OBJECTIVE The present study investigated the influence of SB on lipopolysaccharide (LPS)-induced liver injury. MATERIALS AND METHODS Twenty-eight rats were randomly allocated into four groups: control (received distilled water), SB (received 600 mg/kg), LPS (received 0.25 mg/kg), and LPS + SB (received LPS, 0.25 mg/kg, and SB, 600 mg/kg). SB was administered orally for 14 days while LPS was administered intraperitoneally for 7 days. RESULTS Administration of SB to rats with hepatocyte injury exacerbated liver damage with a significant increase in the activities of serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP). We also observed that SB aggravated LPS-mediated hepatic oxidative stress occasioned by a marked decrease in antioxidant status with a concomitant increase in lipid peroxidation. Furthermore, LPS - mediated increase in inflammatory biomarkers as well as histological deterioration in the liver was exacerbated following the administration of SB to rats. CONCLUSION Taken together, the study provides experimental evidence that SB exacerbates hepatic oxidative stress and inflammation in LPS-mediated liver injury.
Collapse
Affiliation(s)
- Folake Olubukola Asejeje
- Department of Chemical Sciences, Faculty of Natural Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Khalid Damilare Akinola
- Department of Chemical Sciences, Faculty of Natural Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Michael Abayomi Abiola
- Department of Chemical Sciences, Faculty of Natural Sciences, Ajayi Crowther University, Oyo, Nigeria
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
19
|
Bhat TA, Kalathil SG, Goniewicz ML, Hutson A, Thanavala Y. Not all vaping is the same: differential pulmonary effects of vaping cannabidiol versus nicotine. Thorax 2023; 78:922-932. [PMID: 36823163 PMCID: PMC10447384 DOI: 10.1136/thorax-2022-218743] [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: 01/20/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023]
Abstract
RATIONALE Vaping has become a popular method of inhaling various psychoactive substances. While evaluating respiratory effects of vaping have primarily focused on nicotine-containing products, cannabidiol (CBD)-vaping is increasingly becoming popular. It currently remains unknown whether the health effects of vaping nicotine and cannabinoids are similar. OBJECTIVES This study compares side by side the pulmonary effects of acute inhalation of vaporised CBD versus nicotine. METHODS In vivo inhalation study in mice and in vitro cytotoxicity experiments with human cells were performed to assess the pulmonary damage-inducing effects of CBD or nicotine aerosols emitted from vaping devices. MEASUREMENTS AND MAIN RESULTS Pulmonary inflammation in mice was scored by histology, flow cytometry, and quantifying levels of proinflammatory cytokines and chemokines. Lung damage was assessed by histology, measurement of myeloperoxidase activity and neutrophil elastase levels in the bronchoalveolar lavage fluid and lung tissue. Lung epithelial/endothelial integrity was assessed by quantifying BAL protein levels, albumin leak and pulmonary FITC-dextran leak. Oxidative stress was determined by measuring the antioxidant potential in the BAL and lungs. The cytotoxic effects of CBD and nicotine aerosols on human neutrophils and human small airway epithelial cells were evaluated using in vitro air-liquid interface system. Inhalation of CBD aerosol resulted in greater inflammatory changes, more severe lung damage and higher oxidative stress compared with nicotine. CBD aerosol also showed higher toxicity to human cells compared with nicotine. CONCLUSIONS Vaping of CBD induces a potent inflammatory response and leads to more pathological changes associated with lung injury than vaping of nicotine.
Collapse
Affiliation(s)
- Tariq A Bhat
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Suresh G Kalathil
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Maciej L Goniewicz
- Department of Health Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Alan Hutson
- Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Yasmin Thanavala
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
20
|
Liu Z, Al-Quran L, Tong J, Cao X. Analysis of clinical features and inflammatory-related molecules with the disease in acute infectious urticaria. Arch Dermatol Res 2023; 315:1915-1925. [PMID: 36853509 PMCID: PMC10366306 DOI: 10.1007/s00403-023-02564-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/05/2022] [Accepted: 02/01/2023] [Indexed: 03/01/2023]
Abstract
Acute infectious urticaria, a subset of acute urticaria, with severe persistence wheals and systemic symptoms, response well to corticosteroids treatment in combination with antibiotics. The exact pathogenic mechanisms are not fully understood. In this study, we aim to analyze the different clinical features, compare the level of neutrophil activation, and investigate the expression of inflammatory related cytokine in patients with acute urticaria and acute infectious urticaria. Eighteen patients with acute infectious urticaria and eighteen patients with acute urticaria were included in this study. We analyzed the difference between the clinical features and the serum expressions of pro-inflammatory factors in the two groups, then examined the levels of inflammation-associated cytokines before and after treatment of acute infectious urticaria. Hematoxylin & eosin (HE) staining and immunohistochemistry (IHC) were used to further study the relationship between neutrophil and neutrophil-derived Myeloperoxidase (MPO) of lesions in the two groups. The expression levels of C-reactive protein (CRP), D-dimer, interleukin 6 (IL-6), IL-8 and chemokine ligand 8 (CCL8) in serum were significantly higher in acute infectious urticaria than acute urticaria. In acute infectious urticaria, the serum expression levels of CCL8 were significantly decreased after the treatment, a significant correlation observed between CRP levels and IL-6, both CCL8 and CRP were positively correlated with neutrophil granulocytes. Neutrophils infiltration were not observed by HE stains in two groups, but in IHC stains we found a positive expression of MPO in acute infectious urticaria lesions. Elevated neutrophil in the serum, which is associated with the levels of IL-8 & CCL8, and positively expressed MPO in lesions, may be involved in the pathogenic mechanism of acute infectious urticaria.
Collapse
Affiliation(s)
- Zhezhang Liu
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Institute of Dermatology, Jiangxi Academy of Clinical Medical Sciences, No. 17 Yongwaizheng Street, Nanchang City, Jiangxi Province, China
| | - Lina Al-Quran
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Institute of Dermatology, Jiangxi Academy of Clinical Medical Sciences, No. 17 Yongwaizheng Street, Nanchang City, Jiangxi Province, China
| | - Jianbo Tong
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Institute of Dermatology, Jiangxi Academy of Clinical Medical Sciences, No. 17 Yongwaizheng Street, Nanchang City, Jiangxi Province, China.
| | - Xianwei Cao
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
- Institute of Dermatology, Jiangxi Academy of Clinical Medical Sciences, No. 17 Yongwaizheng Street, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
21
|
Langer F, Quick H, Beitzen-Heineke A, Janjetovic S, Mäder J, Lehr C, Bokemeyer C, Kuta P, Renné T, Fiedler W, Beckmann L, Klingler F, Rolling CC. Regulation of coagulation activation in newly diagnosed AML by the heme enzyme myeloperoxidase. Thromb Res 2023; 229:155-163. [PMID: 37473552 DOI: 10.1016/j.thromres.2023.07.006] [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: 02/14/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/22/2023]
Abstract
INTRODUCTION Patients with acute myeloid leukemia (AML) are at increased risk of thrombohemorrhagic complications. Overexpressed tissue factor (TF) on AML blasts contributes to systemic coagulation activation. We have recently shown that the heme enzyme myeloperoxidase (MPO) negatively regulates TF procoagulant activity (PCA) on myelomonocytic cells in vitro. We now aimed to further characterize the functional interaction of MPO and TF in AML in vivo. METHODS We prospectively recruited 66 patients with newly diagnosed AML. TF PCA of isolated peripheral blood mononuclear cells (PBMC) was assessed by single-stage clotting assay in the presence or absence of inhibitors against MPO catalytic activity (ABAH) or against MPO-binding integrins (anti-CD18). MPO in plasma and in AML blasts was measured by ELISA, and plasma D-dimers and prothrombin fragment F1+2 were quantified by automated immunoturbidimetric and chemiluminescence assays, respectively. RESULTS Patients with AML had significantly higher MPO plasma levels compared to healthy controls and exhibited increased levels of D-dimers and F1+2. In vivo thrombin generation was mediated by TF PCA on circulating PBMC. Ex vivo incubation of isolated PBMC with ABAH or anti-CD18 antibody resulted in either increased or decreased TF PCA. The strong and robust correlation of F1+2 with TF PCA of circulating PBMC was abrogated at MPO plasma levels higher than 150 ng/mL, indicating a modulatory role for MPO on TF-mediated in vivo thrombin generation above this threshold. CONCLUSION Our study indicates that catalytically active MPO released by circulating myeloblasts regulates TF-dependent coagulation in patients with newly diagnosed AML in a CD18-dependent manner.
Collapse
Affiliation(s)
- Florian Langer
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hanna Quick
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Beitzen-Heineke
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Snjezana Janjetovic
- Klinik für Hämatologie und Zelltherapie, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Jonathan Mäder
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carina Lehr
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Piotr Kuta
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland; Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Walter Fiedler
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lennart Beckmann
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Klingler
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina C Rolling
- Oncology, Hematology and BMT with section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
22
|
Granai M, Warm V, Vogelsberg A, Milla J, Greif K, Vogel U, Bakchoul T, Rosenberger P, Quintanilla-Martinez L, Schürch CM, Klingel K, Fend F, Bösmüller H. Impact of P-selectin-PSGL-1 Axis on Platelet-Endothelium-Leukocyte Interactions in Fatal COVID-19. J Transl Med 2023; 103:100179. [PMID: 37224922 PMCID: PMC10202465 DOI: 10.1016/j.labinv.2023.100179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023] Open
Abstract
In critically ill patients infected with SARS-CoV-2, early leukocyte recruitment to the respiratory system was found to be orchestrated by leukocyte trafficking molecules accompanied by massive secretion of proinflammatory cytokines and hypercoagulability. Our study aimed to explore the interplay between leukocyte activation and pulmonary endothelium in different disease stages of fatal COVID-19. Our study comprised 10 COVID-19 postmortem lung specimens and 20 control lung samples (5 acute respiratory distress syndrome, 2 viral pneumonia, 3 bacterial pneumonia, and 10 normal), which were stained for antigens representing the different steps of leukocyte migration: E-selectin, P-selectin, PSGL-1, ICAM1, VCAM1, and CD11b. Image analysis software QuPath was used for quantification of positive leukocytes (PSGL-1 and CD11b) and endothelium (E-selectin, P-selectin, ICAM1, VCAM1). Expression of IL-6 and IL-1β was quantified by RT-qPCR. Expression of P-selectin and PSGL-1 was strongly increased in the COVID-19 cohort compared with all control groups (COVID-19:Controls, 17:23, P < .0001; COVID-19:Controls, 2:75, P < .0001, respectively). Importantly, P-selectin was found in endothelial cells and associated with aggregates of activated platelets adherent to the endothelial surface in COVID-19 cases. In addition, PSGL-1 staining disclosed positive perivascular leukocyte cuffs, reflecting capillaritis. Moreover, CD11b showed a strongly increased positivity in COVID-19 compared with all controls (COVID-19:Controls, 2:89; P = .0002), indicating a proinflammatory immune microenvironment. Of note, CD11b exhibited distinct staining patterns at different stages of COVID-19 disease. Only in cases with very short disease course, high levels of IL-1β and IL-6 mRNA were observed in lung tissue. The striking upregulation of PSGL-1 and P-selectin reflects the activation of this receptor-ligand pair in COVID-19, increasing the efficiency of initial leukocyte recruitment, thus promoting tissue damage and immunothrombosis. Our results show that endothelial activation and unbalanced leukocyte migration play a central role in COVID-19 involving the P-selectin-PSGL-1 axis.
Collapse
Affiliation(s)
- Massimo Granai
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Verena Warm
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Antonio Vogelsberg
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Jakob Milla
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Karen Greif
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Ulrich Vogel
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Tamam Bakchoul
- Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen, Tübingen, Germany; Centre for Clinical Transfusion Medicine Tübingen ZKT gGmbH, University of Tübingen, Tübingen, Germany
| | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University of Tübingen, Tübingen, Germany
| | | | - Christian M Schürch
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Karin Klingel
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Falko Fend
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany.
| | - Hans Bösmüller
- Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
23
|
Bojti I, Wang Q, Bojti T, Bojti F, Siegel PM, Heidt T, Moser M, Bode C, Westermann D, Peter K, Diehl P. An Activation-Specific Anti-Mac-1 Designed-Ankyrin-Repeat-Protein Attenuates Colitis in Mice. Life (Basel) 2023; 13:1464. [PMID: 37511839 PMCID: PMC10381548 DOI: 10.3390/life13071464] [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: 04/17/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
(1) Background: Inflammatory bowel diseases are complex and multifactorial disorders of unknown etiology. The extravasation of activated leukocytes is a critical step in the pathogenesis of these diseases. Leukocyte integrin Mac-1 (αMβ2; CD11b/CD18) is crucial for the extravasation of myeloid cells, and a novel activation-specific anti-Mac-1 Designed Ankyrin Repeat protein (DARPin F7) is a promising therapeutic agent for inflammatory diseases. In its activated conformation, Mac-1 expresses the high-affinity binding site I-domain, which the DARPin F7 selectively targets. In our study, we aimed to explore the therapeutic potential of anti-Mac-1 DARPin F7 in murine dextrane sodium sulfate (DSS)-induced colitis. (2) Methods: C57BL/6J mice received 3% DSS drinking water for five days, followed by normal drinking water for one week. The mice were treated with DARPin F7 or a control substance daily via intraperitoneal injections. Disease activity index (DAI), colon length, myeloperoxidase (MPO) activity measurements, H&E staining, and qRT-PCR were conducted after euthanizing the mice on day 12. (3) Results: Treatment with DARPin F7 resulted in less pronounced colon shortening and significantly lower histological scores. The DARPin F7-treated animals experienced substantially less disease and myeloperoxidase (MPO) activity. Animals that received DARPin F7 treatment suffered less weight loss and recovered from the weight loss more efficiently. Treatment with DARPin F7 also led to significantly reduced mRNA expression of inflammatory cytokines. (4) Conclusion: Anti-Mac-1 treatment markedly reduced disease activity and inflammatory reaction accompanying DSS-induced colitis in mice.
Collapse
Affiliation(s)
- Istvan Bojti
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| | - Qianqi Wang
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| | - Tibor Bojti
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| | - Felicitas Bojti
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| | - Patrick Malcolm Siegel
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| | - Timo Heidt
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| | - Martin Moser
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Medicine, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3004, Australia
| | - Philipp Diehl
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Hugstetter Street 55, 79106 Freiburg, Germany
| |
Collapse
|
24
|
Conley HE, Sheats MK. Targeting Neutrophil β 2-Integrins: A Review of Relevant Resources, Tools, and Methods. Biomolecules 2023; 13:892. [PMID: 37371473 DOI: 10.3390/biom13060892] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Neutrophils are important innate immune cells that respond during inflammation and infection. These migratory cells utilize β2-integrin cell surface receptors to move out of the vasculature into inflamed tissues and to perform various anti-inflammatory responses. Although critical for fighting off infection, neutrophil responses can also become dysregulated and contribute to disease pathophysiology. In order to limit neutrophil-mediated damage, investigators have focused on β2-integrins as potential therapeutic targets, but so far these strategies have failed in clinical trials. As the field continues to move forward, a better understanding of β2-integrin function and signaling will aid the design of future therapeutics. Here, we provide a detailed review of resources, tools, experimental methods, and in vivo models that have been and will continue to be utilized to investigate the vitally important cell surface receptors, neutrophil β2-integrins.
Collapse
Affiliation(s)
- Haleigh E Conley
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
| | - M Katie Sheats
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
| |
Collapse
|
25
|
Wang F, Peters R, Jia J, Mudd M, Salemi M, Allers L, Javed R, Duque TLA, Paddar MA, Trosdal ES, Phinney B, Deretic V. ATG5 provides host protection acting as a switch in the atg8ylation cascade between autophagy and secretion. Dev Cell 2023; 58:866-884.e8. [PMID: 37054706 PMCID: PMC10205698 DOI: 10.1016/j.devcel.2023.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/26/2023] [Accepted: 03/20/2023] [Indexed: 04/15/2023]
Abstract
ATG5 is a part of the E3 ligase directing lipidation of ATG8 proteins, a process central to membrane atg8ylation and canonical autophagy. Loss of Atg5 in myeloid cells causes early mortality in murine models of tuberculosis. This in vivo phenotype is specific to ATG5. Here, we show using human cell lines that absence of ATG5, but not of other ATGs directing canonical autophagy, promotes lysosomal exocytosis and secretion of extracellular vesicles and, in murine Atg5fl/fl LysM-Cre neutrophils, their excessive degranulation. This is due to lysosomal disrepair in ATG5 knockout cells and the sequestration by an alternative conjugation complex, ATG12-ATG3, of ESCRT protein ALIX, which acts in membrane repair and exosome secretion. These findings reveal a previously undescribed function of ATG5 in its host-protective role in murine experimental models of tuberculosis and emphasize the significance of the branching aspects of the atg8ylation conjugation cascade beyond the canonical autophagy.
Collapse
Affiliation(s)
- Fulong Wang
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Ryan Peters
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Jingyue Jia
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Michal Mudd
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Michelle Salemi
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA
| | - Lee Allers
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Ruheena Javed
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Thabata L A Duque
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Masroor A Paddar
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Einar S Trosdal
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA
| | - Brett Phinney
- Proteomics Core Facility, UC Davis Genome Center, University of California, Davis, Davis, CA 95616, USA
| | - Vojo Deretic
- Autophagy, Inflammation and Metabolism Center of Biochemical Research Excellence, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, 915 Camino de Salud, NE, Albuquerque, NM 87131, USA.
| |
Collapse
|
26
|
Lemal P, May K, König S, Schroyen M, Gengler N. Invited review: From heat stress to disease-Immune response and candidate genes involved in cattle thermotolerance. J Dairy Sci 2023:S0022-0302(23)00214-X. [PMID: 37164864 DOI: 10.3168/jds.2022-22727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/01/2023] [Indexed: 05/12/2023]
Abstract
Heat stress implies unfavorable effects on primary and functional traits in dairy cattle and, in consequence, on the profitability of the whole production system. The increasing number of days with extreme hot temperatures suggests that it is imperative to detect the heat stress status of animals based on adequate measures. However, confirming the heat stress status of an individual is still challenging, and, in consequence, the identification of novel heat stress biomarkers, including molecular biomarkers, remains a very relevant issue. Currently, it is known that heat stress seems to have unfavorable effects on immune system mechanisms, but this information is of limited use in the context of heat stress phenotyping. In addition, there is a lack of knowledge addressing the molecular mechanisms linking the relevant genes to the observed phenotype. In this review, we explored the potential molecular mechanisms explaining how heat stress affects the immune system and, therefore, increases the occurrence of immune-related diseases in cattle. In this regard, 2 relatively opposite hypotheses are under focus: the immunosuppressive action of cortisol, and the proinflammatory effect of heat stress. In both hypotheses, the modulation of the immune response during heat stress is highlighted. Moreover, it is possible to link candidate genes to these potential mechanisms. In this context, immune markers are very valuable indicators for the detection of heat stress in dairy cattle, broadening the portfolio of potential biomarkers for heat stress.
Collapse
Affiliation(s)
- P Lemal
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium
| | - K May
- Institute of Animal Breeding and Genetics, Justus-Liebig-University of Gießen, Ludwigstraße 21B, 35390 Gießen, Germany
| | - S König
- Institute of Animal Breeding and Genetics, Justus-Liebig-University of Gießen, Ludwigstraße 21B, 35390 Gießen, Germany
| | - M Schroyen
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium
| | - N Gengler
- TERRA Teaching and Research Center, University of Liège, Gembloux Agro-Bio Tech (ULiège-GxABT), 5030 Gembloux, Belgium.
| |
Collapse
|
27
|
Hao X, Shen Y, Chen N, Zhang W, Valverde E, Wu L, Chan HL, Xu Z, Yu L, Gao Y, Bado I, Michie LN, Rivas CH, Dominguez LB, Aguirre S, Pingel BC, Wu YH, Liu F, Ding Y, Edwards DG, Liu J, Alexander A, Ueno NT, Hsueh PR, Tu CY, Liu LC, Chen SH, Hung MC, Lim B, Zhang XHF. Osteoprogenitor-GMP crosstalk underpins solid tumor-induced systemic immunosuppression and persists after tumor removal. Cell Stem Cell 2023; 30:648-664.e8. [PMID: 37146584 PMCID: PMC10165729 DOI: 10.1016/j.stem.2023.04.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/09/2023] [Accepted: 04/06/2023] [Indexed: 05/07/2023]
Abstract
Remote tumors disrupt the bone marrow (BM) ecosystem (BME), eliciting the overproduction of BM-derived immunosuppressive cells. However, the underlying mechanisms remain poorly understood. Herein, we characterized breast and lung cancer-induced BME shifts pre- and post-tumor removal. Remote tumors progressively lead to osteoprogenitor (OP) expansion, hematopoietic stem cell dislocation, and CD41- granulocyte-monocyte progenitor (GMP) aggregation. The tumor-entrained BME is characterized by co-localization between CD41- GMPs and OPs. OP ablation abolishes this effect and diminishes abnormal myeloid overproduction. Mechanistically, HTRA1 carried by tumor-derived small extracellular vesicles upregulates MMP-13 in OPs, which in turn induces the alterations in the hematopoietic program. Importantly, these effects persist post-surgery and continue to impair anti-tumor immunity. Conditional knockout or inhibition of MMP-13 accelerates immune reinstatement and restores the efficacies of immunotherapies. Therefore, tumor-induced systemic effects are initiated by OP-GMP crosstalk that outlasts tumor burden, and additional treatment is required to reverse these effects for optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yichao Shen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Nan Chen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Elizabeth Valverde
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ling Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hilda L Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhan Xu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Liqun Yu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yang Gao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Igor Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Laura Natalee Michie
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Charlotte Helena Rivas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Luis Becerra Dominguez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sergio Aguirre
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Bradley C Pingel
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yi-Hsuan Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yunfeng Ding
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - David G Edwards
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jun Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Angela Alexander
- Department of Breast Medical Oncology and Morgan Welch IBC Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology and Morgan Welch IBC Research Program and Clinic, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; University of Hawai'i Cancer Center (UHCC), 701 Ilalo Street, Honolulu, HI 96813, USA
| | - Po-Ren Hsueh
- Departments of Laboratory Medicine and Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Chih-Yen Tu
- School of Medicine, College of Medicine, China Medical University, Taichung 406, Taiwan; Division of Pulmonary and Critical Care, Department of Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Liang-Chih Liu
- School of Medicine, College of Medicine, China Medical University, Taichung 406, Taiwan; Division of Breast Surgery, Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Shu-Hsia Chen
- Immunomonitoring Core, Center for Immunotherapy Research, Houston Methodist Research Institute (HMRI), Houston, TX, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung 40402, Taiwan
| | - Bora Lim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
28
|
Schoen TJ, Calise DG, Bok JW, Giese MA, Nwagwu CD, Zarnowski R, Andes D, Huttenlocher A, Keller NP. Aspergillus fumigatus transcription factor ZfpA regulates hyphal development and alters susceptibility to antifungals and neutrophil killing during infection. PLoS Pathog 2023; 19:e1011152. [PMID: 37126504 PMCID: PMC10174577 DOI: 10.1371/journal.ppat.1011152] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/11/2023] [Accepted: 04/18/2023] [Indexed: 05/02/2023] Open
Abstract
Hyphal growth is essential for host colonization during Aspergillus infection. The transcription factor ZfpA regulates A. fumigatus hyphal development including branching, septation, and cell wall composition. However, how ZfpA affects fungal growth and susceptibility to host immunity during infection has not been investigated. Here, we use the larval zebrafish-Aspergillus infection model and primary human neutrophils to probe how ZfpA affects A. fumigatus pathogenesis and response to antifungal drugs in vivo. ZfpA deletion promotes fungal clearance and attenuates virulence in wild-type hosts and this virulence defect is abrogated in neutrophil-deficient zebrafish. ZfpA deletion also increases susceptibility to human neutrophils ex vivo while overexpression impairs fungal killing. Overexpression of ZfpA confers protection against the antifungal caspofungin by increasing chitin synthesis during hyphal development, while ZfpA deletion reduces cell wall chitin and increases caspofungin susceptibility in neutrophil-deficient zebrafish. These findings suggest a protective role for ZfpA activity in resistance to the innate immune response and antifungal treatment during A. fumigatus infection.
Collapse
Affiliation(s)
- Taylor J. Schoen
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Dante G. Calise
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jin Woo Bok
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Morgan A. Giese
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Chibueze D. Nwagwu
- Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Robert Zarnowski
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - David Andes
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Nancy P. Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Plant Pathology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
29
|
Influenza Virus Infection Increases Host Susceptibility To Secondary Infection with Pseudomonas aeruginosa, and This Is Attributed To Neutrophil Dysfunction through Reduced Myeloperoxidase Activity. Microbiol Spectr 2023; 11:e0365522. [PMID: 36475755 PMCID: PMC9927171 DOI: 10.1128/spectrum.03655-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Secondary bacterial infection greatly increased the morbidity and mortality of influenza virus infection. To investigate the underlying mechanism by which influenza impairs the pulmonary defense against secondary Pseudomonas aeruginosa (P. aeruginosa) infection, we established a lethal mouse model in which to study secondary P. aeruginosa infection after influenza virus infection. We found a significant increase in host susceptibility to a secondary infection with P. aeruginosa in mice after an influenza virus infection, and this was accompanied by severe immunopathology and pulmonary inflammation. Importantly, we demonstrated that neutrophils were essential for P. aeruginosa clearance in secondarily infected mice. Further, we revealed that influenza impaired the phagocytosis and digestion functions of pulmonary neutrophils for P. aeruginosa clearance. We identified that the activity of reactive oxygen species (ROS) and the myeloperoxidase (MPO) activity of neutrophils in the lungs played an important role in antibacterial host defense in influenza-infected lungs. Hereby, influenza virus infection causes deficient MPO activity in neutrophils, and this contributes to the increased susceptibility to secondary P. aeruginosa infection. Treatment with Bacillus Calmette-Guerin polysaccharide nucleic acid (BCG-PSN) prior to secondary P. aeruginosa infection may improve the function of neutrophils, resulting in significantly reduced lethality during secondary P. aeruginosa infection. We also demonstrated that treatment with anti-influenza immune serum during the early stage of an influenza virus infection could decrease the disease severity of secondary P. aeruginosa infection. Our findings suggest that improving the MPO activity of neutrophils may provide a therapeutic strategy for viral-bacterial coinfection. IMPORTANCE A secondary bacterial infection, such as that of P. aeruginosa, often occurs after a pulmonary virus infection and contributes to severe disease. However, the underlying mechanisms responsible for viral-bacterial synergy in the lung remain largely unknown. In this study, we reported that influenza virus infection increases a host’s susceptibility to secondary infection by P. aeruginosa by reducing the MPO activity of neutrophils. We also demonstrated that treatment with BCG-PSN or anti-influenza immune serum prior to secondary P. aeruginosa infection can reduce the disease severity. Our findings suggest that improving the MPO activity of neutrophils may provide a therapeutic strategy for viral-bacterial coinfection.
Collapse
|
30
|
Singh AK, Awasthi R, Malviya R. Bioinspired microrobots: Opportunities and challenges in targeted cancer therapy. J Control Release 2023; 354:439-452. [PMID: 36669531 DOI: 10.1016/j.jconrel.2023.01.042] [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: 12/08/2022] [Revised: 01/15/2023] [Accepted: 01/15/2023] [Indexed: 01/21/2023]
Abstract
Chemotherapy is still the most effective technique to treat many forms of cancer. However, it also carries a high risk of side effects. Numerous nanomedicines have been developed to avoid unintended consequences and significant negative effects of conventional therapies. Achieving targeted drug delivery also has several challenges. In this context, the development of microrobots is receiving considerable attention of formulation scientists and clinicians to overcome such challenges. Due to their mobility, microrobots can infiltrate tissues and reach tumor sites more quickly. Different types of microrobots, like custom-made moving bacteria, microengines powered by small bubbles, and hybrid spermbots, can be designed with complex features that are best for precise targeting of a wide range of cancers. In this review, we mainly focus on the idea of how microrobots can quickly target cancer cells and discuss specific advantages of microrobots. A brief summary of the microrobots' drug loading and release behavior is provided in this manuscript. This manuscript will assist clinicians and other medical professionals in diagnosing and treating cancer without surgery.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rajendra Awasthi
- Department of Pharmaceutical Sciences, School of Health Sciences & Technology, University of Petroleum and Energy Studies (UPES), Energy Acres, P.O. Bidholi, Via-Prem Nagar, Dehradun 248 007, Uttarakhand, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
31
|
Neurobehavioral and biochemical responses to artemisinin-based drug and aflatoxin B 1 co-exposure in rats. Mycotoxin Res 2023; 39:67-80. [PMID: 36701108 DOI: 10.1007/s12550-023-00474-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/27/2023]
Abstract
Populations in malaria endemic areas are frequently exposed to mycotoxin-contaminated diets. The possible toxicological outcome of co-exposure to dietary aflatoxin B1 (AFB1) and artemisinin-based combination therapy warrants investigation to ascertain amplification or attenuation of cellular injury. Here, we investigated the neurobehavioral and biochemical responses associated with co-exposure to anti-malarial drug coartem, an artemether-lumefantrine combination (5 mg/kg body weight, twice a day and 3 days per week) and AFB1 (35 and 70 µg/kg body weight) in rats. Motor deficits, locomotor incompetence, and anxiogenic-like behavior induced by low AFB1 dose were significantly (p < 0.05) assuaged by coartem but failed to rescue these behavioral abnormalities in high AFB1-dosed group. Coartem administration did not alter exploratory deficits typified by reduced track plot densities and greater heat map intensity in high AFB1-dosed animals. Furthermore, the reduction in cerebral and cerebellar acetylcholinesterase activity, anti-oxidant enzyme activities, and glutathione and thiol levels were markedly assuaged by coartem administration in low AFB1 group but not in high AFB1-dosed animals. The significant attenuation of cerebral and cerebellar oxidative stress indices namely reactive oxygen and nitrogen species, xanthine oxidase activity, and lipid peroxidation by coartem administration was evident in low AFB1 group but not high AFB1 dose. Although coartem administration abated nitric oxide level, activities of myeloperoxidase, caspase-9, and caspase-3 in animals exposed to both doses of AFB1, these indices were significantly higher than the control. Coartem administration ameliorated histopathological and mophometrical changes due to low AFB1 exposure but not in high AFB1 exposure. In conclusion, contrary to AFB1 alone, behavioral and biochemical responses were not altered in animals singly exposed to coartem. Co-exposure to coartem and AFB1 elicited no additional risk but partially lessened neurotoxicity associated with AFB1 exposure.
Collapse
|
32
|
Ngum NH, Fakeh NB, Lem AE, Mahamat O. Prevalence of malaria and associated clinical manifestations and myeloperoxidase amongst populations living in different altitudes of Mezam division, North West Region, Cameroon. Malar J 2023; 22:20. [PMID: 36658587 PMCID: PMC9850770 DOI: 10.1186/s12936-022-04438-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 12/31/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Malaria is a growing problem in Africa, with prevalence varies from areas to areas due to several factors including the altitude. This study aimed to investigate the malaria distribution and its relationship with level of some blood parameters and plasma myeloperoxidase (MPO) in population of three localities with different altitudes. METHODS A total of 150 participants were recruited in each locality and facial body temperature of each was measured using a forehead digital thermometer. Blood samples were collected and used diagnose malaria parasite using the rapid test followed by Giemsa stain microscopy and have the full blood count and MPO level using a colorimetric method. RESULTS The overall prevalence of falciparum malaria was 34.7%, with no difference between the three communities, but Bambili of high altitude had the highest prevalence (70.7%). A majority of the infected persons had mild malaria, with most cases being asymptomatic (temperature < 37.5 ºC). Patients had significant increase of geometric mean malaria parasite density (GMPD) in Bambili (1755 ± 216 parasites/µL) and Bamenda (1060 ± 2515 parasites/µL of blood) than patients in Santa (737 ± 799 parasites/µL). There was a significant risk to have malaria infection in Bambili (OR = 33.367, p = 0.021) than in Santa (OR = 2.309, p = 0.362). Bambili' participants of 6-10 years showed a high prevalence of malaria (85.7%). GMPD was significantly different between males (p = 0.010) as well as females (p = 0.000). Participants from Santa (11.2 ± 3.2 g/dL) and Bambili (12.6 ± 2.4 g/dL) had a high haemoglobin concentration than those from Bamenda (10.6 ± 2.1 g/dL). There was a significant difference in the WBC counts and platelet counts among infected participants in the study areas. MPO level had an increasing trend among infected participants in Santa (2.378 ± 0.250), Bambili (2.582 ± 0.482) and Bamenda (2.635 ± 0.466). CONCLUSION The results of the present study demonstrated that altitudinal variations significant impact the risk of population to have malaria with high parasitaemia and may contribute to the malaria prevalence and severity by affecting the haemoglobin concentration, WBC and platelet level and plasma MPO in population.
Collapse
Affiliation(s)
- Ntonifor Helen Ngum
- grid.449799.e0000 0004 4684 0857Department of Biological Sciences, Faculty of Science, The University of Bamenda, BP 39, Bambili, N. W. Region Cameroon ,grid.442553.10000 0004 0622 6369African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer’s University, Ede, Osun State Nigeria
| | - Ngahbort Belthine Fakeh
- grid.449799.e0000 0004 4684 0857Department of Biological Sciences, Faculty of Science, The University of Bamenda, BP 39, Bambili, N. W. Region Cameroon
| | - Abongwa Edith Lem
- grid.449799.e0000 0004 4684 0857Department of Biological Sciences, Faculty of Science, The University of Bamenda, BP 39, Bambili, N. W. Region Cameroon ,grid.442553.10000 0004 0622 6369African Centre of Excellence for Genomics of Infectious Diseases (ACEGID), Redeemer’s University, Ede, Osun State Nigeria
| | - Oumar Mahamat
- grid.449799.e0000 0004 4684 0857Department of Biological Sciences, Faculty of Science, The University of Bamenda, BP 39, Bambili, N. W. Region Cameroon
| |
Collapse
|
33
|
Harper AK, Kirsch-Mangu TK, Lutfi H, Morris RT, Saed GM. Binding of Intracellular Myeloperoxidase to αV/β1 Integrin Serves as a Mechanism of Survival in Epithelial Ovarian Cancer. Reprod Sci 2023; 30:291-300. [PMID: 35799017 DOI: 10.1007/s43032-022-01025-7] [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: 03/17/2022] [Accepted: 06/23/2022] [Indexed: 01/11/2023]
Abstract
We were the first to report that epithelial ovarian cancer (EOC) cells and tissues express myeloperoxidase (MPO) that is known to play a role in immune surveillance and inflammation by myeloid cells. Additionally, we reported that MPO is colocalized with inducible nitric oxide synthase (iNOS), a key pro-oxidant enzyme, and plays a key role in regulating apoptosis in EOC cells. Whereas myeloid cells express MPO in a dimeric form, intriguingly, here we report the unique expression of only the monomeric form of MPO in EOC cells, tissues, and blood of an ovarian cancer patient. Additionally, we have identified a cell membrane receptor, αV/β1 integrin, that is uniquely expressed by both chemosensitive and chemoresistant EOC cells with significantly higher expression in chemoresistant EOC cells. More importantly, we have demonstrated that monoclonal antibodies against αV/β1 integrin induced cytotoxicity in EOC cells, but not in normal cells, that is also synergistic with conventional chemotherapies. Cytotoxicity of αV/β1 antibodies is due to conformational changes in αV/β1 integrin which prevents monomeric MPO binding to αV/β1 integrin inhibiting the activation of MPO, leading to increased apoptosis. Since normal epithelial cells and macrophages lack monomeric MPO and αV/β1 integrin system, targeting this unique MPO-dependent survival mechanism will selectively eliminate EOC cells and will be the target for developing specific ovarian cancer therapies.
Collapse
Affiliation(s)
- Amy K Harper
- Division of Gynecologic Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, Detroit, MI, 48201, USA
| | - Thea K Kirsch-Mangu
- Department of Obstetrics and Gynecology, The C. S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E Hancock St, Detroit, MI, 48201, USA
| | - Hala Lutfi
- Department of Obstetrics and Gynecology, The C. S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E Hancock St, Detroit, MI, 48201, USA
| | - Robert T Morris
- Division of Gynecologic Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, The C. S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E Hancock St, Detroit, MI, 48201, USA
| | - Ghassan M Saed
- Division of Gynecologic Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, Detroit, MI, 48201, USA.
- Department of Obstetrics and Gynecology, The C. S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, 275 E Hancock St, Detroit, MI, 48201, USA.
| |
Collapse
|
34
|
Owumi SE, Adedara IA, Otunla MT, Owoeye O. Influence of furan and lead co-exposure at environmentally relevant concentrations on neurobehavioral performance, redox-regulatory system and apoptotic responses in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 97:104011. [PMID: 36396074 DOI: 10.1016/j.etap.2022.104011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 05/10/2023]
Abstract
Furan and lead are contaminants of global concern due to the potential public health threat associated with their exposure. Herein, the neurobehavioral performance, biochemical effects and histological alterations associated with co-exposure to furan (8 mg/kg) and lead acetate at low, environmentally realistic concentrations (1, 10 and 100 µg PbAc/L) for 28 uninterrupted days were investigated in rats. The results demonstrated that locomotor, motor and exploratory deficits associated with separate exposure to furan and lead was exacerbated in the co-exposed rats. Furan and lead co-exposure aggravated the marked decrease in acetylcholinesterase activity and antioxidant status, elevation in oxido-inflammatory stress indices and caspases activation in the cerebrum and cerebellum of exposed rats compared with control. Furan and lead co-exposure worsened neuronal degeneration as verified by histomorphometry and histochemical staining. Collectively, furan and lead acts together to exacerbate neurotoxicity via inhibition of cholinergic system, induction of oxido-inflammatory stress and caspases activation in rats.
Collapse
Affiliation(s)
- Solomon E Owumi
- Cancer Research and Molecular Biology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Isaac A Adedara
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Moses T Otunla
- Cancer Research and Molecular Biology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olatunde Owoeye
- Department of Anatomy, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
35
|
Short term deuterium depletion in drinking water reduced tumor induced oxidative stress in mice liver. Pathol Res Pract 2022; 240:154186. [DOI: 10.1016/j.prp.2022.154186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/07/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
|
36
|
Pan G, Zhang P, Yang J, Wu Y. The regulatory effect of specialized pro-resolving mediators on immune cells. Biomed Pharmacother 2022; 156:113980. [DOI: 10.1016/j.biopha.2022.113980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/22/2022] [Accepted: 11/04/2022] [Indexed: 11/08/2022] Open
|
37
|
Salem MOA, Taştan Y, Bilen S, Terzi E, Sönmez AY. Effects of white mustard (Sinapis alba) oil on growth performance, immune response, blood parameters, digestive and antioxidant enzyme activities in rainbow trout (Oncorhynchusmykiss). FISH & SHELLFISH IMMUNOLOGY 2022; 131:283-299. [PMID: 36210002 DOI: 10.1016/j.fsi.2022.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/24/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
A study was conducted to evaluate the effects of dietary supplementation of white mustard (Sinapis alba) oil (WMO) on growth performance, immune responses, digestive and antioxidant enzyme activities in juvenile rainbow trout (Oncorhynchus mykiss). For this purpose, fish (initial weight: 25.77 ± 0.13 g) were divided into four experimental groups in triplicate and fed ad libitum twice a day with diets containing WMO at 0 (control), 0.5, 1, and 1.5% of diet for 9 weeks. Three fish from each tank (n:9 per treatment) were sampled on 21st, 42nd, and 63rd days for further analyses. At the end of the feeding period, fish were challenged with Aeromonas hydrophila and Yersinia ruckeri in two separate experimental setups. Results showed that final weight, weight gain, and specific growth rate were significantly increased in all experimental groups compared to the control. Feed conversion ratio was similar among treatments. Respiratory burst and potential killing activity decreased in all experimental groups compared to the control (P < 0.05). Lysozyme and myeloperoxidase activities were elevated in all experimental groups at the end of the experiment compared to the control (P < 0.05). Cytokine gene expressions in the head kidney and intestine were elevated in all experimental groups compared to that of the control in general (P < 0.05). Hematological responses of the experimental fish groups were similar to that of the control (P > 0.05). Pepsin and trypsin levels decreased in all experimental groups (P < 0.05). In terms of antioxidant enzyme activities, significant improvement in liver superoxide dismutase, catalase, and glutathione s-transferase activities in all treatment groups were determined (P < 0.05). In addition, a significant decline in liver lipid peroxidation levels was recorded in all treated groups at all sampling times compared to the control (P < 0.05). At the end of this feeding trial, no significant differences (P > 0.05) were observed in survival against A. hydrophila among experimental groups compared to the control (P > 0.05). However, increased survival against Y. ruckeri was determined in experimental fish groups (P < 0.05). This study suggests that white mustard oil had a favorable effect on the overall health and growth of rainbow trout.
Collapse
Affiliation(s)
- Mohamed Omar Abdalla Salem
- Kastamonu University Institute of Science, Department of Aquaculture, Kastamonu, Turkey; Bani Waleed University, Faculty of Education, Department of Biology, Bani Walid, Libya
| | - Yiğit Taştan
- Kastamonu University, Faculty of Fisheries, Department of Aquaculture, Kastamonu, Turkey
| | - Soner Bilen
- Kastamonu University, Faculty of Fisheries, Department of Basic Sciences, Kastamonu, Turkey
| | - Ertugrul Terzi
- Kastamonu University, Faculty of Fisheries, Department of Aquaculture, Kastamonu, Turkey
| | - Adem Yavuz Sönmez
- Kastamonu University, Faculty of Fisheries, Department of Basic Sciences, Kastamonu, Turkey.
| |
Collapse
|
38
|
Rizo-Téllez SA, Sekheri M, Filep JG. Myeloperoxidase: Regulation of Neutrophil Function and Target for Therapy. Antioxidants (Basel) 2022; 11:antiox11112302. [PMID: 36421487 PMCID: PMC9687284 DOI: 10.3390/antiox11112302] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/04/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Neutrophils, the most abundant white blood cells in humans, are critical for host defense against invading pathogens. Equipped with an array of antimicrobial molecules, neutrophils can eradicate bacteria and clear debris. Among the microbicide proteins is the heme protein myeloperoxidase (MPO), stored in the azurophilic granules, and catalyzes the formation of the chlorinating oxidant HOCl and other oxidants (HOSCN and HOBr). MPO is generally associated with killing trapped bacteria and inflicting collateral tissue damage to the host. However, the characterization of non-enzymatic functions of MPO suggests additional roles for this protein. Indeed, evolving evidence indicates that MPO can directly modulate the function and fate of neutrophils, thereby shaping immunity. These actions include MPO orchestration of neutrophil trafficking, activation, phagocytosis, lifespan, formation of extracellular traps, and MPO-triggered autoimmunity. This review scrutinizes the multifaceted roles of MPO in immunity, focusing on neutrophil-mediated host defense, tissue damage, repair, and autoimmunity. We also discuss novel therapeutic approaches to target MPO activity, expression, or MPO signaling for the treatment of inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Salma A. Rizo-Téllez
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
- Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada
| | - Meriem Sekheri
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
- Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada
| | - János G. Filep
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
- Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada
- Correspondence: ; Tel.: +1-514-252-3400 (ext. 4662)
| |
Collapse
|
39
|
Salem GA, Mohamed AAR, Ghonimi WAM, Abdallah HM, Rhouma NR, Ali RI. The synbiotic mixture of Bacillus licheniformis and Saccharomyces cerevisiae extract aggravates dextran sulfate sodium induced colitis in rats. BMC Vet Res 2022; 18:405. [PMID: 36384756 PMCID: PMC9667625 DOI: 10.1186/s12917-022-03479-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Uncertain effects of probiotics and/or prebiotics have been reported in experimental and clinical colitis. This study aims to examine the effects of a synbiotic combination comprising Bacillus licheniformis DSM 17236 and Saccharomyces cerevisiae cell wall extract on dextran sulfate sodium (DSS)-induced colitis in Sprague Dawley rats. METHODS Acute colitis was induced in rats by oral administration of DSS 3.5% for 7 days. Fifty rats were divided equally into five groups; one control group and the other groups were induced with colitis and treated with or without the tested synbiotic, mixed with diet, for 28 days and sulfasalazine (100 mg/kg) via intragastric tube once daily for 14 days. RESULTS Symptomatically, the synbiotic administration raised the disease activity index (DAI) to comparable scores of the DSS group, specially from the 2nd to 7th days post DSS intoxication. It also induced a significant (p < 0.05) amplification of WBCs, myeloperoxidase (MPO), malondialdehyde (MDA), nuclear factor kappa B (NF-kB) expression and proinflammatory cytokines tumor necrosis factor alpha (TNFα), interferon gamma (INFγ), and interleukin-1 beta (IL-1β) while depressed the antioxidant enzymes glutathione peroxidase (GPx), catalase (CAT), and superoxide dismutase (SOD) when compared with the DSS and control groups. The DSS intoxicated and Synbiotic+DSS groups showed desquamations of the covering epithelium, noticeable diffuse leukocytic infiltrations, sever catarrhal enteritis, ischemic colitis with diffuse coagulative necrosis of the entire colonic mucosa. Contrarily, sulfasalazine proved to be effective in the reduction of the tested inflammatory markers and the pathological degenerative changes of the DSS ulcerative colitis. CONCLUSION The examined synbiotic did not ameliorate but aggravated the DSS-induced colitis, so it should be subjected to intensive experimental and clinical testing before their use in animals and human.
Collapse
Affiliation(s)
- Gamal A Salem
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, P.O. Box 44519, Zagazig, Egypt.
| | - Amany Abdel-Rahman Mohamed
- Departments of Forensic Medicine and Toxicology and Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Wael A M Ghonimi
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - H M Abdallah
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Nasreddin R Rhouma
- Department of Micobiology, Faculty of Science, Misurata University, Misurata, P.O. Box 2478, Libya
| | - Reem I Ali
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Banha University, Banha, 13518, Egypt
| |
Collapse
|
40
|
Lian N, Chen Y, Chen S, Xiao T, Song C, Ke Y, Wei X, Gong C, Yu H, Gu H, Chen Q, Li M, Chen X. Necroptosis-mediated HMGB1 secretion of keratinocytes as a key step for inflammation development in contact hypersensitivity. Cell Death Dis 2022; 8:451. [PMID: 36344541 PMCID: PMC9640721 DOI: 10.1038/s41420-022-01228-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
Keratinocyte necroptosis (with proinflammatory characteristic) is required for epidermal damage in contact hypersensitivity (CHS). In DNCB-induced CHS mice model, we observed the aggravated keratinocyte death and increased phosphorylation level of MLKL, RIPK3 and RIPK1. However, CHS skin lesion did not present in keratinocyte-specific Mlkl knockout mice. We validated that MLKL-mediated keratinocyte necroptosis is required for epidermal damage in response to immune microenvironment in CHS. Moreover, MLKL-mediated necroptosis deficiency or inhibition resulted in blocking recruitment and activation of inflammatory cells in CHS via reducing HMGB1 release in keratinocytes. This study suggests that MLKL-mediated keratinocyte necroptosis functions as a self-amplified actor in inflammatory responses and could be considered as an effective therapeutic target. It proposes an innovative prospective that inhibiting keratinocyte necroptosis can prevent the development of epidermal damage in CHS. ![]()
Collapse
|
41
|
Lockhart JS, Sumagin R. Non-Canonical Functions of Myeloperoxidase in Immune Regulation, Tissue Inflammation and Cancer. Int J Mol Sci 2022; 23:ijms232012250. [PMID: 36293108 PMCID: PMC9603794 DOI: 10.3390/ijms232012250] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
Myeloperoxidase (MPO) is one of the most abundantly expressed proteins in neutrophils. It serves as a critical component of the antimicrobial defense system, facilitating microbial killing via generation of reactive oxygen species (ROS). Interestingly, emerging evidence indicates that in addition to the well-recognized canonical antimicrobial function of MPO, it can directly or indirectly impact immune cells and tissue responses in homeostatic and disease states. Here, we highlight the emerging non-canonical functions of MPO, including its impact on neutrophil longevity, activation and trafficking in inflammation, its interactions with other immune cells, and how these interactions shape disease outcomes. We further discuss MPO interactions with barrier forming endothelial and epithelial cells, specialized cells of the central nervous system (CNS) and its involvement in cancer progression. Such diverse function and the MPO association with numerous inflammatory disorders make it an attractive target for therapies aimed at resolving inflammation and limiting inflammation-associated tissue damage. However, while considering MPO inhibition as a potential therapy, one must account for the diverse impact of MPO activity on various cellular compartments both in health and disease.
Collapse
|
42
|
Coadministration of Stigmasterol and Dexamethasone (STIG+DEX) Modulates Steroid-Resistant Asthma. Mediators Inflamm 2022; 2022:2222270. [PMID: 36060927 PMCID: PMC9433298 DOI: 10.1155/2022/2222270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/25/2022] [Accepted: 07/24/2022] [Indexed: 11/18/2022] Open
Abstract
Airway inflammation in asthma is managed with anti-inflammatory steroids such as dexamethasone (DEX). However, about 20% of asthmatics do not respond to this therapy and are classified as steroid-resistant. Currently, no effective therapy is available for steroid-resistant asthma. This work therefore evaluated the effect of a plant sterol, stigmasterol (STIG), and stigmasterol-dexamethasone combination (STIG+DEX) in LPS-ovalbumin-induced steroid-resistant asthma in Guinea pigs. To do this, the effect of drugs on inflammatory features such as airway hyperreactivity and histopathology of lung tissue was evaluated. Additionally, the possible pathway of drug action was assessed by measuring events such neutrophil levels, oxidative and nitrative stress, and histone deacetylase 2 (HDAC2) and interleukin 17 (IL-17) levels. STIG alone did not affect inflammatory features, although it caused some changes in the molecular events associated with steroid-resistant asthma. However, STIG+DEX caused significant modulation of inflammatory features by protecting against destruction of lung tissue. The modulation of inflammatory features was associated with significant inhibition of neutrophilia and oxidative and nitrative stress, decrease in HDAC2, and increase in IL-17 levels that are usually associated with steroid-resistant asthma. Our findings show that although STIG and DEX individually do not protect against steroid-resistant asthma, their coadministration results in significant modulation of inflammatory features and the associated molecular events that lead to steroid-resistant asthma.
Collapse
|
43
|
Higashisaka K. Health Effects and Safety Assurance of Nanoparticles in Vulnerable Generations. Biol Pharm Bull 2022; 45:806-812. [PMID: 35786586 DOI: 10.1248/bpb.b22-00277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nanoparticles have a variety of useful functions. They have already been put to practical use in products in many industrial arenas, such as the cosmetics and food fields. Therefore, we cannot avoid the unintentional nanoparticle exposure of vulnerable people such as pregnant women and infants, and the importance of evaluating the safety of such vulnerable generations, who are highly sensitive to chemical substances, has been pointed out worldwide. However, it is still difficult to determine the hazards posed by nanoparticle exposure in everyday life. From this perspective, to analyze the risk from nanoparticles to vulnerable generations, nano-safety science research has been conducted through the collection of toxicity information on nanoparticles based on their physicochemical properties and kinetics via the association analysis of physicochemical properties, kinetics, and toxicity. The results of this nano-safety science research have been used in nano-safety design research to develop safer forms of nanoparticles. The findings of these studies will not only provide insights that will help us to formulate new policies for the risk management of nanoparticles; they will also lead directly to the development of sustainable nanotechnology (nanotechnology that can be safely, usefully, and sustainably used). These developments will contribute not only to the development of the nano-industry and the promotion of its social acceptance, but also to future developments in the field of health science.
Collapse
Affiliation(s)
- Kazuma Higashisaka
- Institute for Advanced Co-Creation Studies, Osaka University.,Graduate School of Pharmaceutical Sciences, Osaka University
| |
Collapse
|
44
|
Gilly A, Klaric L, Park YC, Png G, Barysenka A, Marsh JA, Tsafantakis E, Karaleftheri M, Dedoussis G, Wilson JF, Zeggini E. Gene-based whole genome sequencing meta-analysis of 250 circulating proteins in three isolated European populations. Mol Metab 2022; 61:101509. [PMID: 35504531 PMCID: PMC9118462 DOI: 10.1016/j.molmet.2022.101509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Deep sequencing offers unparalleled access to rare variants in human populations. Understanding their role in disease is a priority, yet prohibitive sequencing costs mean that many cohorts lack the sample size to discover these effects on their own. Meta-analysis of individual variant scores allows the combination of rare variants across cohorts and study of their aggregated effect at the gene level, boosting discovery power. However, the methods involved have largely not been field-tested. In this study, we aim to perform the first meta-analysis of gene-based rare variant aggregation optimal tests, applied to the human cardiometabolic proteome. METHODS Here, we carry out this analysis across MANOLIS, Pomak and ORCADES, three isolated European cohorts with whole-genome sequencing (total N = 4,422). We examine the genetic architecture of 250 proteomic traits of cardiometabolic relevance. We use a containerised pipeline to harmonise variant lists across cohorts and define four sets of qualifying variants. For every gene, we interrogate protein-damaging variants, exonic variants, exonic and regulatory variants, and regulatory only variants, using the CADD and Eigen scores to weigh variants according to their predicted functional consequence. We perform single-cohort rare variant analysis and meta-analyse variant scores using the SMMAT package. RESULTS We describe 5 rare variant pQTLs (RV-pQTL) which pass our stringent significance threshold (7.45 × 10-11) and quality control procedure. These were split between four cis signals for MARCO, TEK, MMP2 and MPO, and one trans association for GDF2 in the SERPINA11 gene. We show that the cis-MPO association, which was not detectable using the single-point data alone, is driven by 5 missense and frameshift variants. These include rs140636390 and rs119468010, which are specific to MANOLIS and ORCADES, respectively. We show how this kind of signal could improve the predictive accuracy of genetic factors in common complex disease such as stroke and cardiovascular disease. CONCLUSIONS Our proof-of-concept study demonstrates the power of gene-based meta-analyses for discovering disease-relevant associations complementing common-variant signals by incorporating population-specific rare variation.
Collapse
Affiliation(s)
- Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Lucija Klaric
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Grace Png
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; TUM School of Medicine, Technical University of Munich and Klinikum Rechts der Isar, Ismaninger Straße 22, 8167 Munich, Germany
| | - Andrei Barysenka
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Joseph A Marsh
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | | | | | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 70, El. Venizelou ave. 17671, Kallithea, Greece
| | - James F Wilson
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK; Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, EH8 9AG, UK
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; TUM School of Medicine, Technical University of Munich and Klinikum Rechts der Isar, Ismaninger Straße 22, 8167 Munich, Germany.
| |
Collapse
|
45
|
Xu X, Li T, Jin K. Bioinspired and Biomimetic Nanomedicines for Targeted Cancer Therapy. Pharmaceutics 2022; 14:1109. [PMID: 35631695 PMCID: PMC9147382 DOI: 10.3390/pharmaceutics14051109] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 02/05/2023] Open
Abstract
Undesirable side effects and multidrug resistance are the major obstacles in conventional chemotherapy towards cancers. Nanomedicines provide alternative strategies for tumor-targeted therapy due to their inherent properties, such as nanoscale size and tunable surface features. However, the applications of nanomedicines are hampered in vivo due to intrinsic disadvantages, such as poor abilities to cross biological barriers and unexpected off-target effects. Fortunately, biomimetic nanomedicines are emerging as promising therapeutics to maximize anti-tumor efficacy with minimal adverse effects due to their good biocompatibility and high accumulation abilities. These bioengineered agents incorporate both the physicochemical properties of diverse functional materials and the advantages of biological materials to achieve desired purposes, such as prolonged circulation time, specific targeting of tumor cells, and immune modulation. Among biological materials, mammalian cells (such as red blood cells, macrophages, monocytes, and neutrophils) and pathogens (such as viruses, bacteria, and fungi) are the functional components most often used to confer synthetic nanoparticles with the complex functionalities necessary for effective nano-biointeractions. In this review, we focus on recent advances in the development of bioinspired and biomimetic nanomedicines (such as mammalian cell-based drug delivery systems and pathogen-based nanoparticles) for targeted cancer therapy. We also discuss the biological influences and limitations of synthetic materials on the therapeutic effects and targeted efficacies of various nanomedicines.
Collapse
Affiliation(s)
- Xiaoqiu Xu
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tong Li
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Ke Jin
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; (X.X.); (T.L.)
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
46
|
Blanter M, Cambier S, De Bondt M, Vanbrabant L, Pörtner N, Abouelasrar Salama S, Metzemaekers M, Marques PE, Struyf S, Proost P, Gouwy M. Method Matters: Effect of Purification Technology on Neutrophil Phenotype and Function. Front Immunol 2022; 13:820058. [PMID: 35222394 PMCID: PMC8866851 DOI: 10.3389/fimmu.2022.820058] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and the first cells responding to infection and injury. Due to their limited ex vivo lifespan and the impossibility to cryopreserve or expand them in vitro, neutrophils need to be purified from fresh blood for immediate use in experiments. Importantly, neutrophil purification methods may artificially modify the phenotype and functional characteristics of the isolated cells. The aim of this study was to expose the effects of ‘classical’ density-gradient purification versus the more expensive but faster immunomagnetic isolation on neutrophil phenotype and functionality. We found that in the absence of inflammatory stimuli, density-gradient-derived neutrophils showed increased polarization responses as well as enhanced release of reactive oxygen species (ROS), neutrophil extracellular traps (NETs) and granular proteins compared to cells derived from immunomagnetic isolation, which yields mostly quiescent neutrophils. Upon exposure to pro-inflammatory mediators, immunomagnetic isolation-derived neutrophils were significantly more responsive in polarization, ROS production, phagocytosis, NETosis and degranulation assays, in comparison to density-gradient-derived cells. We found no difference in chemotactic response in Multiscreen and under-agarose migration assays, but Boyden assays showed reduced chemotaxis of immunomagnetic isolation-derived neutrophils. Finally, we confirmed that density-gradient purification induces artificial activation of neutrophils, evidenced by e.g. higher expression of CD66b, formyl peptide receptor 1 (FPR1) and CD35, and the appearance of a separate neutrophil population expressing surface molecules atypical for neutrophils (e.g. CXCR3, MHC-II and CD14). Based on these results, we recommend using immunomagnetic separation of neutrophils for studying neutrophil polarization, phagocytosis, ROS production, degranulation and NETosis, whereas for Boyden chemotaxis assays, the density-gradient purification is more suitable.
Collapse
Affiliation(s)
- Marfa Blanter
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Seppe Cambier
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Mirre De Bondt
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Lotte Vanbrabant
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Noëmie Pörtner
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Sara Abouelasrar Salama
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Mieke Metzemaekers
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
47
|
Lau D, Lechermann LM, Gallagher FA. Clinical Translation of Neutrophil Imaging and Its Role in Cancer. Mol Imaging Biol 2022; 24:221-234. [PMID: 34637051 PMCID: PMC8983506 DOI: 10.1007/s11307-021-01649-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 01/22/2023]
Abstract
Neutrophils are the first line of defense against pathogens and abnormal cells. They regulate many biological processes such as infections and inflammation. Increasing evidence demonstrated a role for neutrophils in cancer, where different subpopulations have been found to possess both pro- or anti-tumorigenic functions in the tumor microenvironment. In this review, we discuss the phenotypic and functional diversity of neutrophils in cancer, their prognostic significance, and therapeutic relevance in human and preclinical models. Molecular imaging methods are increasingly used to probe neutrophil biology in vivo, as well as the cellular changes that occur during tumor progression and over the course of treatment. This review will discuss the role of neutrophil imaging in oncology and the lessons that can be drawn from imaging in infectious diseases and inflammatory disorders. The major factors to be considered when developing imaging techniques and biomarkers for neutrophils in cancer are reviewed. Finally, the potential clinical applications and the limitations of each method are discussed, as well as the challenges for future clinical translation.
Collapse
Affiliation(s)
- Doreen Lau
- Department of Radiology, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Centre, Cambridge, UK.
- Department of Oncology, University of Oxford, Oxford, UK.
| | | | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Centre, Cambridge, UK.
| |
Collapse
|
48
|
Kumar S, Choubey AK, Srivastava PK. The effects of dietary immunostimulants on the innate immune response of Indian major carp: A review. FISH & SHELLFISH IMMUNOLOGY 2022; 123:36-49. [PMID: 35217196 DOI: 10.1016/j.fsi.2022.02.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 06/14/2023]
Abstract
Immunostimulants, as feed additives, play an important role in maintaining fish health and enhancing their overall growth by providing resistance against diseases in cultured fish. At the initial stages of life of fish, innate immunity is the essential mechanism in their survival. Later, innate immunity has an instructive role in adapting acquired immune response and homeostasis through different receptor proteins. Several studies have been conducted to analyze the effect of dietary immunostimulants like algae, plant extracts, vitamins, herbs, probiotics, and prebiotics-containing diets in Indian major carps. Many bacterial, fungal and viral pathogens are responsible for high death rates in both wild and cultured fish. It's a major limiting factor for world aquaculture industries. Recognition of invading pathogens by different pathogen recognition receptor plays an important role for the activation of different pathways to initiate protective immune responses. Hence, there is a growing need to control the devastating effects of diseases without recourse to toxic chemicals or antibiotics. Keeping with alternative approaches without using toxic chemicals to control fish diseases in mind, many immunostimulants are used, which enhance immune responses along with their gene expression level through different signaling pathway. The objective of this review is to summarize and evaluate the current knowledge of various immunostimulants and their immune responses in three Indian major carps namely Catla catla, Labeo rohita and Cirrhinus mrigala, which are preferred by the people.
Collapse
Affiliation(s)
- Sudhir Kumar
- Institute of Biosciences & Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh- 225003, India
| | - Abhay Kumar Choubey
- Department of Sciences and Humanities, Rajiv Gandhi Institute of Petroleum Technology, Jais, Amethi, Uttar Pradesh-229304, India
| | - Praveen Kumar Srivastava
- Department of Sciences and Humanities, Rajiv Gandhi Institute of Petroleum Technology, Jais, Amethi, Uttar Pradesh-229304, India.
| |
Collapse
|
49
|
Li L, Zhang C, Lin Q, Zhu M, Mei F, Jian S, Zhao D. Role of peroxinectin in the antibacterial immune response of the Chinese mitten crab, Eriocheir sinensis. FISH & SHELLFISH IMMUNOLOGY 2022; 123:496-505. [PMID: 35331883 DOI: 10.1016/j.fsi.2022.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/12/2022] [Accepted: 03/19/2022] [Indexed: 06/14/2023]
Abstract
To elucidate the antibacterial role of peroxinectin (referred to as PXN) and its molecular mechanism in Chinese mitten crab Eriocheir sinensis, we analyzed the bacterial binding and removal of the peroxinectin recombinant protein in vitro and the interaction of peroxinectin with integrin and CuZn-SOD through GST-pulldown and bimolecular fluorescence complementation methods. Concurrently, the effect of peroxinectin interference on the expression of other immune-related genes was studied using RNA interference. The results showed that the recombinant peroxinectin protein could bind to Bacillus subtilis, Staphylococcus aureus, Aeromonas hydrophila, and Vibrio parahaemolyticus with different affinities in vitro and could eliminate Vibrio parahaemolyticus in vivo. The findings also indicated that peroxinectin could establish interactions with integrin and CuZn-SOD in vitro. Furthermore, 48 h after the injection of the peroxinectin gene siRNA in vivo, the expression of peroxinectin mRNA decreased significantly (P < 0.05), integrin mRNA expression decreased by 16.8%, and CuZn-SOD mRNA expression decreased by 62.84% (P < 0.01). The expression levels of Dorsal, GPx, GST, PPAF, and Relish (P < 0.01), as well as that of lectin (P < 0.001) were significantly decreased. When peroxinectin siRNA was injected in vivo for 48 h and Aeromonas hydrophila was injected into mitten crabs, the expression of immune-related genes significantly increased. All data indicate that the recombinant peroxinectin protein in Chinese mitten crabs can recognize and bind different bacteria and promote the elimination of Vibrio parahaemolyticus from the body. Furthermore, peroxinectin may establish interactions with integrin and CuZn-SOD to activate the expression of related immune genes to elicit responses to bacterial infections and achieve immune protection.
Collapse
Affiliation(s)
- Linjie Li
- School of Life Sciences, Nanchang University, Jiangxi, 330013, PR China
| | - Cuizhen Zhang
- School of Life Sciences, Nanchang University, Jiangxi, 330013, PR China
| | - Qichen Lin
- School of Life Sciences, Nanchang University, Jiangxi, 330013, PR China
| | - Minjie Zhu
- School of Life Sciences, Nanchang University, Jiangxi, 330013, PR China
| | - Feng Mei
- School of Life Sciences, Nanchang University, Jiangxi, 330013, PR China
| | - Shaoqing Jian
- School of Life Sciences, Nanchang University, Jiangxi, 330013, PR China; Key Laboratory of Aquatic Animal Resources and Utilization of Jiangxi Province, Jiangxi, 330013, PR China.
| | - Daxian Zhao
- School of Life Sciences, Nanchang University, Jiangxi, 330013, PR China; Key Laboratory of Aquatic Animal Resources and Utilization of Jiangxi Province, Jiangxi, 330013, PR China.
| |
Collapse
|
50
|
Heithoff DM, Pimienta G, Mahan SP, Yang WH, Le DT, House JK, Marth JD, Smith JW, Mahan MJ. Coagulation factor protein abundance in the pre-septic state predicts coagulopathic activities that arise during late-stage murine sepsis. EBioMedicine 2022; 78:103965. [PMID: 35349828 PMCID: PMC8965145 DOI: 10.1016/j.ebiom.2022.103965] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/21/2022] [Accepted: 03/10/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although sepsis accounts for 1 in 5 deaths globally, few molecular therapies exist for this condition. The development of effective biomarkers and treatments for sepsis requires a more complete understanding of host responses and pathogenic mechanisms at early stages of disease to minimize host-driven pathology. METHODS An alternative to the current symptom-based approach used to diagnose sepsis is a precise assessment of blood proteomic changes during the onset and progression of Salmonella Typhimurium (ST) murine sepsis. FINDINGS A distinct pattern of coagulation factor protein abundance was identified in the pre-septic state- prior to overt disease symptoms or bacteremia- that was predictive of the dysregulation of fibrinolytic and anti-coagulant activities and resultant consumptive coagulopathy during ST murine sepsis. Moreover, the changes in protein abundance observed generally have the same directionality (increased or decreased abundance) reported for human sepsis. Significant overlap of ST coagulopathic activities was observed in Gram-negative Escherichia coli- but not in Gram-positive staphylococcal or pneumococcal murine sepsis models. Treatment with matrix metalloprotease inhibitors prevented aberrant inflammatory and coagulopathic activities post-ST infection and increased survival. Antibiotic treatment regimens initiated after specific changes arise in the plasma proteome post-ST infection were predictive of an increase in disease relapse and death after cessation of antibiotic treatment. INTERPRETATION Altered blood proteomics provides a platform to develop rapid and easy-to-perform tests to predict sepsis for early intervention via biomarker incorporation into existing blood tests prompted by patient presentation with general malaise, and to stratify Gram-negative and Gram-positive infections for appropriate treatment. Antibiotics are less effective in microbial clearance when initiated after the onset of altered blood proteomics as evidenced by increased disease relapse and death after termination of antibiotic therapy. Treatment failure is potentially due to altered bacterial / host-responses and associated increased host-driven pathology, providing insight into why delays in antibiotic administration in human sepsis are associated with increased risk for death. Delayed treatment may thus require prolonged therapy for microbial clearance despite the prevailing notion of antibiotic de-escalation and shortened courses of antibiotics to improve drug stewardship. FUNDING National Institutes of Health, U.S. Army.
Collapse
Affiliation(s)
- Douglas M Heithoff
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106, USA
| | - Genaro Pimienta
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Scott P Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106, USA; Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis CA 95616, USA
| | - Won Ho Yang
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Glycosylation Network Research Center and Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Infectious and Inflammatory Diseases Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Dzung T Le
- Department of Pathology, University of California, La Jolla, San Diego, CA 92093, USA
| | - John K House
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Camden, New South Wales 2570, Australia
| | - Jamey D Marth
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Infectious and Inflammatory Diseases Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Jeffrey W Smith
- Cancer Metabolism and Signaling Networks Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Michael J Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara CA 93106, USA; Institute for Collaborative Biotechnologies, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|