1
|
Wu E, Guan J, Yu Y, Lin S, Ding T, Chu Y, Pan F, Liu M, Yang Y, Zhang Z, Zhang J, Zhan C, Qian J. Exemplifying interspecies variation of liposome in vivo fate by the effects of anti-PEG antibodies. Acta Pharm Sin B 2024; 14:4994-5007. [PMID: 39664439 PMCID: PMC11628802 DOI: 10.1016/j.apsb.2024.07.009] [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: 04/24/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 12/13/2024] Open
Abstract
The different fate of liposomes among species has been discovered and mentioned in many studies, but the underlying mechanisms have not been explored. In the present work, we concentrated on the in vivo fate of PEGylated liposomes (sLip) in three commonly used species (mice, rats, and dogs). It was exhibited that the accelerated blood clearance (ABC) phenomenon and hypersensitivity in large animals (beagle dogs) were much more significant than that in rodents. We demonstrated that anti-PEG IgM (partially) and complement (mostly) determined the elimination of sLip and linked the distinct interspecies performances with the diverse complement capacity among species. Based on the data from animals and clinical patients, it was revealed that the fate of sLip in large animals was closer to that in humans, for the sufficient complement capacity could expose the potential adverse reactions caused by anti-PEG antibodies. Our results suggested that the distinctive interspecies performances of sLip were highly related to the physiological variabilities among species, which should not be overlooked in the innovation and translation of nanomedicines.
Collapse
Affiliation(s)
- Ercan Wu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Juan Guan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
| | - Yifei Yu
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Shiqi Lin
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Tianhao Ding
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Yuxiu Chu
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Feng Pan
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
| | - Mengyuan Liu
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
| | - Yang Yang
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Zui Zhang
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Changyou Zhan
- Department of Pharmacy, Shanghai Pudong Hospital, Pudong Medical Center & Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Jun Qian
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education & Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 201203, China
| |
Collapse
|
2
|
Perdrizet UG, Hill JE, Sobchishin L, Singh B, Fernando C, Bollinger TK, Misra V. Tissue and cellular tropism of Eptesicus fuscus gammaherpesvirus in big brown bats, potential role of pulmonary intravascular macrophages. Vet Pathol 2024; 61:550-561. [PMID: 38619093 PMCID: PMC11264566 DOI: 10.1177/03009858241244849] [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] [Indexed: 04/16/2024]
Abstract
Gammaherpesviruses (γHVs) are recognized as important pathogens in humans but their relationship with other animal hosts, especially wildlife species, is less well characterized. Our objectives were to examine natural Eptesicus fuscus gammaherpesvirus (EfHV) infections in their host, the big brown bat (Eptesicus fuscus), and determine whether infection is associated with disease. In tissue samples from 132 individual big brown bats, EfHV DNA was detected by polymerase chain reaction in 41 bats. Tissues from 59 of these cases, including 17 from bats with detectable EfHV genomes, were analyzed. An EfHV isolate was obtained from one of the cases, and electron micrographs and whole genome sequencing were used to confirm that this was a unique isolate of EfHV. Although several bats exhibited various lesions, we did not establish EfHV infection as a cause. Latent infection, defined as RNAScope probe binding to viral latency-associated nuclear antigen in the absence of viral envelope glycoprotein probe binding, was found within cells of the lymphoid tissues. These cells also had colocalization of the B-cell probe targeting CD20 mRNA. Probe binding for both latency-associated nuclear antigen and a viral glycoprotein was observed in individual cells dispersed throughout the alveolar capillaries of the lung, which had characteristics of pulmonary intravascular macrophages. Cells with a similar distribution in bat lungs expressed major histocompatibility class II, a marker for antigen presenting cells, and the existence of pulmonary intravascular macrophages in bats was confirmed with transmission electron microscopy. The importance of this cell type in γHVs infections warrants further investigation.
Collapse
Affiliation(s)
| | | | | | - Baljit Singh
- University of Saskatchewan, Saskatoon, SK, Canada
| | | | | | - Vikram Misra
- University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
3
|
Stucki AO, Sauer UG, Allen DG, Kleinstreuer NC, Perron MM, Yozzo KL, Lowit AB, Clippinger AJ. Differences in the anatomy and physiology of the human and rat respiratory tracts and impact on toxicological assessments. Regul Toxicol Pharmacol 2024; 150:105648. [PMID: 38772524 PMCID: PMC11198871 DOI: 10.1016/j.yrtph.2024.105648] [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: 01/30/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024]
Abstract
Inhalation is a critical route through which substances can exert adverse effects in humans; therefore, it is important to characterize the potential effects that inhaled substances may have on the human respiratory tract by using fit for purpose, reliable, and human relevant testing tools. In regulatory toxicology testing, rats have primarily been used to assess the effects of inhaled substances as they-being mammals-share similarities in structure and function of the respiratory tract with humans. However, questions about inter-species differences impacting the predictability of human effects have surfaced. Disparities in macroscopic anatomy, microscopic anatomy, or physiology, such as breathing mode (e.g., nose-only versus oronasal breathing), airway structure (e.g., complexity of the nasal turbinates), cell types and location within the respiratory tract, and local metabolism may impact inhalation toxicity testing results. This review shows that these key differences describe uncertainty in the use of rat data to predict human effects and supports an opportunity to harness modern toxicology tools and a detailed understanding of the human respiratory tract to develop testing approaches grounded in human biology. Ultimately, as the regulatory purpose is protecting human health, there is a need for testing approaches based on human biology and mechanisms of toxicity.
Collapse
Affiliation(s)
| | - Ursula G Sauer
- Scientific Consultancy - Animal Welfare, Neubiberg, Germany
| | - David G Allen
- International Collaboration on Cosmetics Safety (ICCS), Mount Royal, NJ, USA
| | - Nicole C Kleinstreuer
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods (NICEATM), National Institute of Environmental Health Sciences, NC, USA
| | - Monique M Perron
- US Environmental Protection Agency, Office of Pesticide Programs, Washington, DC, USA
| | - Krystle L Yozzo
- US Environmental Protection Agency, Office of Pesticide Programs, Washington, DC, USA
| | - Anna B Lowit
- US Environmental Protection Agency, Office of Pollution Prevention and Toxics, Washington, DC, USA
| | | |
Collapse
|
4
|
Maina JN. A critical assessment of the cellular defences of the avian respiratory system: are birds in general and poultry in particular relatively more susceptible to pulmonary infections/afflictions? Biol Rev Camb Philos Soc 2023; 98:2152-2187. [PMID: 37489059 DOI: 10.1111/brv.13000] [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: 02/13/2023] [Revised: 07/01/2023] [Accepted: 07/07/2023] [Indexed: 07/26/2023]
Abstract
In commercial poultry farming, respiratory diseases cause high morbidities and mortalities, begetting colossal economic losses. Without empirical evidence, early observations led to the supposition that birds in general, and poultry in particular, have weak innate and adaptive pulmonary defences and are therefore highly susceptible to injury by pathogens. Recent findings have, however, shown that birds possess notably efficient pulmonary defences that include: (i) a structurally complex three-tiered airway arrangement with aerodynamically intricate air-flow dynamics that provide efficient filtration of inhaled air; (ii) a specialised airway mucosal lining that comprises air-filtering (ciliated) cells and various resident phagocytic cells such as surface and tissue macrophages, dendritic cells and lymphocytes; (iii) an exceptionally efficient mucociliary escalator system that efficiently removes trapped foreign agents; (iv) phagocytotic atrial and infundibular epithelial cells; (v) phagocytically competent surface macrophages that destroy pathogens and injurious particulates; (vi) pulmonary intravascular macrophages that protect the lung from the vascular side; and (vii) proficiently phagocytic pulmonary extravasated erythrocytes. Additionally, the avian respiratory system rapidly translocates phagocytic cells onto the respiratory surface, ostensibly from the subepithelial space and the circulatory system: the mobilised cells complement the surface macrophages in destroying foreign agents. Further studies are needed to determine whether the posited weak defence of the avian respiratory system is a global avian feature or is exclusive to poultry. This review argues that any inadequacies of pulmonary defences in poultry may have derived from exacting genetic manipulation(s) for traits such as rapid weight gain from efficient conversion of food into meat and eggs and the harsh environmental conditions and severe husbandry operations in modern poultry farming. To reduce pulmonary diseases and their severity, greater effort must be directed at establishment of optimal poultry housing conditions and use of more humane husbandry practices.
Collapse
Affiliation(s)
- John N Maina
- Department of Zoology, University of Johannesburg, Auckland Park Campus, Kingsway Avenue, Johannesburg, 2006, South Africa
| |
Collapse
|
5
|
Ma W, Loving CL, Driver JP. From Snoot to Tail: A Brief Review of Influenza Virus Infection and Immunity in Pigs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1187-1194. [PMID: 37782856 PMCID: PMC10824604 DOI: 10.4049/jimmunol.2300385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/07/2023] [Indexed: 10/04/2023]
Abstract
Pigs play an important role in influenza A virus (IAV) epidemiology because they support replication of human, avian, and swine origin viruses and act as an IAV reservoir for pigs and other species, including humans. Moreover, novel IAVs with human pandemic potential may be generated in pigs. To minimize the threat of IAVs to human and swine health, it is crucial to understand host defense mechanisms that restrict viral replication and pathology in pigs. In this article, we review IAV strains circulating in the North American swine population, as well as porcine innate and acquired immune responses to IAV, including recent advances achieved through immunological tools developed specifically for swine. Furthermore, we highlight unique aspects of the porcine pulmonary immune system, which warrant consideration when developing vaccines and therapeutics to limit IAV in swine or when using pigs to model human IAV infections.
Collapse
Affiliation(s)
- Wenjun Ma
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO
| | - Crystal L. Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA
| | - John P. Driver
- Division of Animal Sciences, University of Missouri, Columbia, MO
| |
Collapse
|
6
|
Clift SJ, Martí-Garcia B, Lawrence JA, Mitchell EP, Fehrsen J, Martínez J, Williams JH, Steyl JCA. Theileriosis in naturally infected roan antelope ( Hippotragus equinus). Vet Pathol 2022; 59:1031-1046. [PMID: 36052867 DOI: 10.1177/03009858221120011] [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: 11/16/2022]
Abstract
Cases of Theileria-associated mortality are rarely reported in African wild artiodactyls. Descriptions of lesions are limited, particularly in endangered hippotraginids. Here, we analyzed retrospectively the gross and histologic findings in 55 roan antelope (Hippotragus equinus) with fatal natural theileriosis. The most frequently recorded gross findings in 40 cases were widespread petechiae and ecchymoses (72.5%), probable anemia (67.5%), icterus (60%), splenomegaly (60%), hepatomegaly (52.5%), and pulmonary edema (50%). Histologic lesions in 34 cases were characterized by multi-organ infiltrates of parasitized and nonparasitized mononuclear leukocytes (MLs), and fewer multinucleate giant cells (MNGCs). Liver, lung, kidney, adrenal gland, and heart were most consistently infiltrated, followed by spleen and lymph nodes. Leukocytes were phenotyped in lung, liver, kidney, and heart specimens from 16 cases, using immunohistochemistry to detect CD20, CD3, myeloid/histiocyte antigen (MAC387), IBA-1, and CD204 surface receptors. A roan polyclonal anti-Theileria sp. (sable) antibody was applied to the same tissues to identify intraleukocytic parasite antigens. Similar proportions of intravascular and extravascular IBA-1-, CD204-, and MAC387-reactive putative monocyte-macrophages and fewer CD3-positive putative T-lymphocytes were identified in all organs, especially the lungs in infected roan. CD20-positive putative B-lymphocytes were significantly scarcer than in uninfected controls. Intraleukocytic Theileria parasites labeled consistently in affected tissues. Some parasitized and nonparasitized MLs and the MNGCs failed to label with selected leukocyte markers. Fatal theileriosis in roans may largely be the result of multi-organ monocyte-macrophage activation with associated tissue injury and overwhelming systemic inflammation. The identity of the parasitized leukocytes and characteristics of the lymphohistiocytic response require further clarification in roans.
Collapse
Affiliation(s)
| | | | | | | | - Jeanni Fehrsen
- University of Pretoria, Onderstepoort, South Africa.,ARC-Onderstepoort Veterinary Research, Onderstepoort, South Africa
| | | | | | | |
Collapse
|
7
|
Jeong S, Kim B, Byun DJ, Jin S, Seo BS, Shin MH, Leem AY, Choung JJ, Park MS, Hyun YM. Lysophosphatidylcholine Alleviates Acute Lung Injury by Regulating Neutrophil Motility and Neutrophil Extracellular Trap Formation. Front Cell Dev Biol 2022; 10:941914. [PMID: 35859904 PMCID: PMC9289271 DOI: 10.3389/fcell.2022.941914] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/14/2022] [Indexed: 11/28/2022] Open
Abstract
Sepsis is predominantly initiated by bacterial infection and can cause systemic inflammation, which frequently leads to rapid death of the patient. However, this acute systemic inflammatory response requires further investigation from the perspectives of clinical judgment criteria and early treatment strategies for the relief of symptoms. Lysophosphatidylcholine (LPC) 18:0 may relieve septic symptoms, but the relevant mechanism is not clearly understood. Therefore, we aimed to assess the effectiveness of LPC as a therapeutic treatment for acute inflammation in the lung induced by lipopolysaccharide in mice. Systemic inflammation of mice was induced by lipopolysaccharide (LPS) inoculation to investigate the role of LPC in the migration and the immune response of neutrophils during acute lung injury. By employing two-photon intravital imaging of the LPS-stimulated LysM-GFP mice and other in vitro and in vivo assays, we examined whether LPC alleviates the inflammatory effect of sepsis. We also tested the effect of LPC to human neutrophils from healthy control and sepsis patients. Our data showed that LPC treatment reduced the infiltration of innate immune cells into the lung. Specifically, LPC altered neutrophil migratory patterns and enhanced phagocytic efficacy in the damaged lung. Moreover, LPC treatment reduced the release of neutrophil extracellular trap (NET), which can damage tissue in the inflamed organ and exacerbate disease. It also reduced human neutrophil migration under inflammatory environment. Our results suggest that LPC can alleviate sepsis-induced lung inflammation by regulating the function of neutrophils. These findings provide evidence for the beneficial application of LPC treatment as a potential therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- Soi Jeong
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Bora Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Da Jeong Byun
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sunmin Jin
- R&D Center, AriBio Co., Ltd., Sengnam, South Korea
| | - Bo Seung Seo
- R&D Center, AriBio Co., Ltd., Sengnam, South Korea
| | - Mi Hwa Shin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Ah Young Leem
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Moo Suk Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Moo Suk Park, ; Young-Min Hyun,
| | - Young-Min Hyun
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- *Correspondence: Moo Suk Park, ; Young-Min Hyun,
| |
Collapse
|
8
|
Carvallo FR, Stevenson VB. Interstitial pneumonia and diffuse alveolar damage in domestic animals. Vet Pathol 2022; 59:586-601. [DOI: 10.1177/03009858221082228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Classification of pneumonia in animals has been controversial, and the most problematic pattern is interstitial pneumonia. This is true from the gross and histologic perspectives, and also from a mechanistic point of view. Multiple infectious and noninfectious diseases are associated with interstitial pneumonia, all of them converging in the release of inflammatory mediators that generate local damage and attract inflammatory cells that inevitably trigger a second wave of damage. Diffuse alveolar damage is one of the more frequently identified histologic types of interstitial pneumonia and involves injury to alveolar epithelial and/or endothelial cells, with 3 distinct stages. The first is the “exudative” stage, with alveolar edema and hyaline membranes. The second is the “proliferative” stage, with hyperplasia and reactive atypia of type II pneumocytes, infiltration of lymphocytes, plasma cells, and macrophages in the interstitium and early proliferation of fibroblasts. These stages are reversible and often nonfatal. If damage persists, there is a third “fibrosing” stage, characterized by fibrosis of the interstitium due to proliferation of fibroblasts/myofibroblasts, persistence of type II pneumocytes, segments of squamous metaplasia of alveolar epithelium, plus inflammation. Understanding the lesion patterns associated with interstitial pneumonias, their causes, and the underlying mechanisms aid in accurate diagnosis that involves an interdisciplinary collaborative approach involving pathologists, clinicians, and radiologists.
Collapse
Affiliation(s)
- Francisco R. Carvallo
- Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA
- Virginia Department of Agriculture and Consumer Services, Harrisonburg, VA
| | | |
Collapse
|
9
|
Wang Z, Hood ED, Nong J, Ding J, Marcos-Contreras OA, Glassman PM, Rubey KM, Zaleski M, Espy CL, Gullipali D, Miwa T, Muzykantov VR, Song WC, Myerson JW, Brenner JS. Combating Complement's Deleterious Effects on Nanomedicine by Conjugating Complement Regulatory Proteins to Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107070. [PMID: 34910334 PMCID: PMC9062787 DOI: 10.1002/adma.202107070] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/19/2021] [Indexed: 05/07/2023]
Abstract
Complement opsonization is among the biggest challenges facing nanomedicine. Nearly instantly after injection into blood, nanoparticles are opsonized by the complement protein C3, leading to clearance by phagocytes, fouling of targeting moieties, and release of anaphylatoxins. While surface polymers such as poly(ethylene glycol) (PEG) partially decrease complement opsonization, most nanoparticles still suffer from extensive complement opsonization, especially when linked to targeting moieties. To ameliorate the deleterious effects of complement, two of mammals' natural regulators of complement activation (RCAs), Factors H and I, are here conjugated to the surface of nanoparticles. In vitro, Factor H or I conjugation to PEG-coated nanoparticles decrease their C3 opsonization, and markedly reduce nanoparticle uptake by phagocytes. In an in vivo mouse model of sepsis-induced lung injury, Factor I conjugation abrogates nanoparticle uptake by intravascular phagocytes in the lungs, allowing the blood concentration of the nanoparticle to remain elevated much longer. For nanoparticles targeted to the lung's endothelium by conjugation to anti-ICAM antibodies, Factor I conjugation shifts the cell-type distribution away from phagocytes and toward endothelial cells. Finally, Factor I conjugation abrogates the severe anaphylactoid responses common to many nanoparticles, preventing systemic capillary leak and preserving blood flow to visceral organs and the brain. Thus, conjugation of RCAs, like Factor I, to nanoparticles is likely to help in nanomedicine's long battle against complement, improving several key parameters critical for clinical success.
Collapse
Affiliation(s)
- Zhicheng Wang
- Departments of Medicine and Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Elizabeth D. Hood
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Jia Nong
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Jing Ding
- Department of Pediatrics, Peking University People’s Hospital, Beijing, 100044, China
| | | | - Patrick M. Glassman
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Kathryn M. Rubey
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104 USA
| | - Michael Zaleski
- Departments of Medicine and Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Carolann L. Espy
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Damodara Gullipali
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Takashi Miwa
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | | | - Wen-Chao Song
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Jacob W. Myerson
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Jacob S. Brenner
- Departments of Medicine and Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104 USA
| |
Collapse
|
10
|
Morelli S, Diakou A, Di Cesare A, Colombo M, Traversa D. Canine and Feline Parasitology: Analogies, Differences, and Relevance for Human Health. Clin Microbiol Rev 2021; 34:e0026620. [PMID: 34378954 PMCID: PMC8404700 DOI: 10.1128/cmr.00266-20] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cats and dogs are treated as family members by most pet owners. Therefore, a high quality of veterinary care and preventive medicine is imperative for animal health and welfare and for the protection of humans from zoonotic pathogens. There is a general perception of cats being treated as "small dogs," especially in the field of clinical parasitology. As a result, several important differences between the two animal species are not taken into proper consideration and are often overlooked. Dogs and cats are profoundly different under evolutionary, biological, ethological, behavioral, and immunological standpoints. These differences impact clinical features, diagnosis, and control of canine and feline parasites and transmission risk for humans. This review outlines the most common parasitoses and vector-borne diseases of dogs and cats, with a focus on major convergences and divergences, and discusses parasites that have (i) evolved based on different preys for dogs and cats, (ii) adapted due to different immunological or behavioral animal profiles, and (iii) developed more similarities than differences in canine and feline infections and associated diseases. Differences, similarities, and peculiarities of canine and feline parasitology are herein reviewed in three macrosections: (i) carnivorism, vegetarianism, anatomy, genetics, and parasites, (ii) evolutionary adaptation of nematodes, including veterinary reconsideration and zoonotic importance, and (iii) behavior and immune system driving ectoparasites and transmitted diseases. Emphasis is given to provide further steps toward a more accurate evaluation of canine and feline parasitology in a changing world in terms of public health relevance and One Health approach.
Collapse
Affiliation(s)
- Simone Morelli
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Anastasia Diakou
- Faculty of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Angela Di Cesare
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | | | - Donato Traversa
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| |
Collapse
|
11
|
Wann SR, Chi PL, Huang WC, Cheng CC, Chang YT. Combination therapy of iPSC-derived conditioned medium with ceftriaxone alleviates bacteria-induced lung injury by targeting the NLRP3 inflammasome. J Cell Physiol 2021; 237:1299-1314. [PMID: 34612516 DOI: 10.1002/jcp.30596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 01/14/2023]
Abstract
The lung is the first and most frequent organ to fail among sepsis patients. The mortality rate of sepsis-related acute lung injury (ALI) is high. Despite appropriate antimicrobial therapy, no treatment strategies are available for sepsis-induced ALI. Stem cell-mediated paracrine signaling is a potential treatment method for various diseases. This study aimed to examine the effects of induced pluripotent stem cell-derived conditioned medium (iPSC-CM) combined with antibiotics on ALI in a rat model of Escherichia coli-induced sepsis. Rats were administered either iPSC-CM or the vehicle (saline) with antibiotics (ceftriaxone). After 72 h, liquid biopsy, bronchoalveolar lavage fluid (BALF), and tissues were harvested for analysis. Survival rates were observed for up to 3 days. Furthermore, we examined the effects of iPSC-CM on cytokine production, metalloproteinase 9 (MMP-9) expression, and NLRP3-ASC interaction in RAW264.7 cells stimulated with lipopolysaccharide/interferon-γ (LPS/IFN-γ). Combined treatment of iPSC-CM with antibiotics significantly improved survival in E. coli-infected rats (p = 0.0006). iPSC-CM ameliorated E. coli-induced infiltration of macrophages, reducing the number of cells in BALF, and suppressing interleukin (IL)-1β, MIP-2, IL-6, and MMP-9 messenger RNA in lung sections. iPSC-CM treatment attenuated NLRP3 expression and inhibited NLRP3 inflammasome activation by disrupting NLRP3-mediated ASC complex formation in LPS/IFN-γ-primed RAW264.7 cells. This study reveals the mechanisms underlying iPSC-CM-conferred anti-inflammatory activity in ALI through the attenuation of macrophage recruitment to the lung, thus inactivating NLRP3 inflammasomes in macrophages. iPSC-CM therapy may be a useful adjuvant treatment to reduce sepsis-related mortality by ameliorating ALI.
Collapse
Affiliation(s)
- Shue-Ren Wann
- Department of Nursing, Shu-Zen Junior College of Medicine and Management, Kaohsiung City, Taiwan.,Department of Medicine, Kaohsiung Veterans General Hospital, Pingtung Branch, Pintung, Taiwan
| | - Pei-Ling Chi
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
| | - Wei-Chun Huang
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei City, Taiwan.,Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan.,Department of Physical Therapy, Fooyin University, Kaohsiung, Taiwan.,Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Chin-Chang Cheng
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan
| | - Yun-Te Chang
- Department of Nursing, Shu-Zen Junior College of Medicine and Management, Kaohsiung City, Taiwan.,School of Medicine, National Yang-Ming Chiao Tung University, Taipei City, Taiwan.,Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung City, Taiwan.,Departement of Nursing, Yuh-Ing Junior College of Health Care and Management, Kaohsiung City, Taiwan
| |
Collapse
|
12
|
Skydsgaard M, Dincer Z, Haschek WM, Helke K, Jacob B, Jacobsen B, Jeppesen G, Kato A, Kawaguchi H, McKeag S, Nelson K, Rittinghausen S, Schaudien D, Vemireddi V, Wojcinski ZW. International Harmonization of Nomenclature and Diagnostic Criteria (INHAND): Nonproliferative and Proliferative Lesions of the Minipig. Toxicol Pathol 2021; 49:110-228. [PMID: 33393872 DOI: 10.1177/0192623320975373] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The INHAND (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions) Project (www.toxpath.org/inhand.asp) is a joint initiative of the Societies of Toxicologic Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP), and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative lesions in laboratory animals. The purpose of this publication is to provide a standardized nomenclature for classifying microscopic lesions observed in most tissues and organs from the minipig used in nonclinical safety studies. Some of the lesions are illustrated by color photomicrographs. The standardized nomenclature presented in this document is also available electronically on the internet (http://www.goreni.org/). Sources of material included histopathology databases from government, academia, and industrial laboratories throughout the world. Content includes spontaneous lesions as well as lesions induced by exposure to test materials. Relevant infectious and parasitic lesions are included as well. A widely accepted and utilized international harmonization of nomenclature for lesions in laboratory animals will provide a common language among regulatory and scientific research organizations in different countries and increase and enrich international exchanges of information among toxicologists and pathologists.
Collapse
Affiliation(s)
| | - Zuhal Dincer
- Pathology Department, Covance Laboratories Limited, Harrogate, United Kingdom
| | - Wanda M Haschek
- Department of Pathobiology, University of Illinois, Urbana, IL, USA
| | - Kris Helke
- Medical University of South Carolina, Charleston, SC, USA
| | | | - Bjoern Jacobsen
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - Gitte Jeppesen
- Charles River Laboratories Copenhagen, Lille Skensved, Denmark
| | - Atsuhiko Kato
- Chugai Pharmaceutical Co, Ltd Research Division, Shizuoka, Japan
| | | | - Sean McKeag
- Pathology Department, Covance Laboratories Limited, Harrogate, United Kingdom
| | | | - Susanne Rittinghausen
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | | | | |
Collapse
|
13
|
Tiba MH, McCracken BM, Leander DC, Colmenero CI, Nemzek JA, Sjoding MW, Konopka KE, Flott TL, VanEpps JS, Daniels RC, Ward KR, Stringer KA, Dickson RP. A novel swine model of the acute respiratory distress syndrome using clinically relevant injury exposures. Physiol Rep 2021; 9:e14871. [PMID: 33991456 PMCID: PMC8123544 DOI: 10.14814/phy2.14871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/18/2022] Open
Abstract
To date, existing animal models of the acute respiratory distress syndrome (ARDS) have failed to translate preclinical discoveries into effective pharmacotherapy or diagnostic biomarkers. To address this translational gap, we developed a high-fidelity swine model of ARDS utilizing clinically relevant lung injury exposures. Fourteen male swine were anesthetized, mechanically ventilated, and surgically instrumented for hemodynamic monitoring, blood, and tissue sampling. Animals were allocated to one of three groups: (1) Indirect lung injury only: animals were inoculated by direct injection of Escherichia coli into the kidney parenchyma, provoking systemic inflammation and distributive shock physiology; (2) Direct lung injury only: animals received volutrauma, hyperoxia, and bronchoscope-delivered gastric particles; (3) Combined indirect and direct lung injury: animals were administered both above-described indirect and direct lung injury exposures. Animals were monitored for up to 12 h, with serial collection of physiologic data, blood samples, and radiographic imaging. Lung tissue was acquired postmortem for pathological examination. In contrast to indirect lung injury only and direct lung injury only groups, animals in the combined indirect and direct lung injury group exhibited all of the physiological, radiographic, and histopathologic hallmarks of human ARDS: impaired gas exchange (mean PaO2 /FiO2 ratio 124.8 ± 63.8), diffuse bilateral opacities on chest radiographs, and extensive pathologic evidence of diffuse alveolar damage. Our novel porcine model of ARDS, built on clinically relevant lung injury exposures, faithfully recapitulates the physiologic, radiographic, and histopathologic features of human ARDS and fills a crucial gap in the translational study of human lung injury.
Collapse
Affiliation(s)
- Mohamad H. Tiba
- Department of Emergency MedicineUniversity of MichiganAnn ArborMIUSA
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
| | - Brendan M. McCracken
- Department of Emergency MedicineUniversity of MichiganAnn ArborMIUSA
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
| | - Danielle C. Leander
- Department of Emergency MedicineUniversity of MichiganAnn ArborMIUSA
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
| | - Carmen I. Colmenero
- Department of Emergency MedicineUniversity of MichiganAnn ArborMIUSA
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
| | - Jean A. Nemzek
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
- Unit of Laboratory Animal MedicineUniversity of MichiganAnn ArborMIUSA
| | - Michael W. Sjoding
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of MichiganAnn ArborMIUSA
- Institute for Healthcare Policy and InnovationUniversity of MichiganAnn ArborMIUSA
- Department of Computational Medicine and BioinformaticsUniversity of MichiganAnn ArborMIUSA
| | - Kristine E. Konopka
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
- Department of PathologyUniversity of MichiganAnn ArborMIUSA
| | - Thomas L. Flott
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
- Department of Clinical PharmacyCollege of PharmacyUniversity of MichiganAnn ArborMIUSA
| | - J. Scott VanEpps
- Department of Emergency MedicineUniversity of MichiganAnn ArborMIUSA
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMIUSA
- Biointerfaces InstituteUniversity of MichiganAnn ArborMIUSA
| | - Rodney C. Daniels
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMIUSA
- Department of PediatricsPediatric Critical Care MedicineUniversity of MichiganAnn ArborMIUSA
| | - Kevin R. Ward
- Department of Emergency MedicineUniversity of MichiganAnn ArborMIUSA
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMIUSA
| | - Kathleen A. Stringer
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of MichiganAnn ArborMIUSA
- Department of Clinical PharmacyCollege of PharmacyUniversity of MichiganAnn ArborMIUSA
| | - Robert P. Dickson
- Michigan Center for Integrative Research in Critical CareUniversity of MichiganAnn ArborMIUSA
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineUniversity of MichiganAnn ArborMIUSA
- Department of Microbiology & ImmunologyUniversity of MichiganAnn ArborMIUSA
| |
Collapse
|
14
|
Bertho N, Meurens F. The pig as a medical model for acquired respiratory diseases and dysfunctions: An immunological perspective. Mol Immunol 2021; 135:254-267. [PMID: 33933817 DOI: 10.1016/j.molimm.2021.03.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/04/2021] [Accepted: 03/13/2021] [Indexed: 12/21/2022]
Abstract
By definition no model is perfect, and this also holds for biology and health sciences. In medicine, murine models are, and will be indispensable for long, thanks to their reasonable cost and huge choice of transgenic strains and molecular tools. On the other side, non-human primates remain the best animal models although their use is limited because of financial and obvious ethical reasons. In the field of respiratory diseases, specific clinical models such as sheep and cotton rat for bronchiolitis, or ferret and Syrian hamster for influenza and Covid-19, have been successfully developed, however, in these species, the toolbox for biological analysis remains scarce. In this view the porcine medical model is appearing as the third, intermediate, choice, between murine and primate. Herein we would like to present the pros and cons of pig as a model for acquired respiratory conditions, through an immunological point of view. Indeed, important progresses have been made in pig immunology during the last decade that allowed the precise description of immune molecules and cell phenotypes and functions. These progresses might allow the use of pig as clinical model of human respiratory diseases but also as a species of interest to perform basic research explorations.
Collapse
Affiliation(s)
| | - François Meurens
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon S7N5E3, Canada
| |
Collapse
|
15
|
Pentraxin 3 expression in lungs and neutrophils of calves. Vet Immunol Immunopathol 2021; 236:110251. [PMID: 33901710 DOI: 10.1016/j.vetimm.2021.110251] [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: 12/30/2020] [Revised: 04/15/2021] [Accepted: 04/18/2021] [Indexed: 11/23/2022]
Abstract
Bacterial lung disease caused by Mannheimia haemolytica inflict significant mortality and morbidity resulting in enormous economic losses to cattle industry. The use of antibiotics is becoming more challenging because of development of anti-microbial resistance. The innate immune system plays a critical role in the initiation of immune response in the lung. Pentraxin 3 (PTX3), a pattern-recognition receptor is produced at sites of inflammation by many cell types, recognizes and binds to many pathogens, activates the complement cascade, and has a role in the clearance of apoptotic and necrotic cells. Because there are very few data on the expression of PTX3 in the lungs, we examined PTX3 expression in lungs of normal and M. haemolytica-infected calves and normal and E. coli lipopolysaccharide-treated cattle neutrophils using light and electron microscopic immunochemistry and Western blots. Immunohistology showed the presence of PTX3 in airway epithelial cells, alveolar septa and macrophages in normal and inflamed lungs of calves and the blots showed a significant increase in the expression of PTX3 in lungs from infected calves. Immuno-gold electron microscopy showed PTX3 in the nuclei, cytoplasm, and vesicular organelles of alveolar macrophages, endothelial cells and pulmonary intravascular macrophages (PIMs). Immunohistochemical staining for PTX3 in peripheral blood neutrophils shows an altered staining pattern in neutrophils stimulated with lipopolysachharide (LPS). However, western blots no significant change in PTX3 amount in LPS-treated neutrophils compared to the controls. These are the first data on the expression of PTX3 in the lungs and the neutrophils of cattle which may add to our understanding of innate immunity in cattle lungs.
Collapse
|
16
|
Abouelfetouh MM, Salah E, Ding M, Ding Y. Application of α 2 -adrenergic agonists combined with anesthetics and their implication in pulmonary intravascular macrophages-insulted pulmonary edema and hypoxemia in ruminants. J Vet Pharmacol Ther 2021; 44:478-502. [PMID: 33709435 DOI: 10.1111/jvp.12960] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/08/2021] [Indexed: 11/29/2022]
Abstract
Alpha2 -adrenergic agonists have been implicated in the development of pulmonary edema (PE) and sustained hypoxemia that lead to life-threatening pulmonary distress in ruminants, especially with sensitive and compromised animals. Recently, there is limited understanding of exact mechanism underlying pulmonary alterations associated with α2 -adrenergic agonist administration. Ruminants have a rich population of pulmonary intravascular macrophages (PIMs) in the pulmonary circulation, which may be involved in the development of pulmonary alveolo-capillary barrier damage. Hence, the central thesis of this review is overviewing the literatures regarding the systemic use of α2 -adrenergic agonists in domestic ruminants, focusing on their pulmonary side effects, especially on the influence of PIMs on the lung. At this moment, further studies are needed to provide a clear emphasis and better understanding of the potential role of PIMs in the lung pathophysiology associated with α2 -adrenergic agonists. These preliminary studies would be potentially to develop future medications and intervention targets that may be helpful to alleviate or prevent the critical striking pulmonary effects, and thereby improving the safety of α2 -agonist application in ruminants.
Collapse
Affiliation(s)
- Mahmoud M Abouelfetouh
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China.,Department of Surgery, Radiology and Anaesthesiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Egypt
| | - Eman Salah
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, China.,Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Egypt
| | - Mingxing Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yi Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
17
|
van Valenberg FJP, Brummelhuis ISG, Lindner LH, Kuhnle F, Wedmann B, Schweizer P, Hossann M, Witjes JA, Oosterwijk E. DPPG 2-Based Thermosensitive Liposomes with Encapsulated Doxorubicin Combined with Hyperthermia Lead to Higher Doxorubicin Concentrations in the Bladder Compared to Conventional Application in Pigs: A Rationale for the Treatment of Muscle-Invasive Bladder Cancer. Int J Nanomedicine 2021; 16:75-88. [PMID: 33447028 PMCID: PMC7802347 DOI: 10.2147/ijn.s280034] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/10/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose Current treatment options for muscle-invasive bladder cancer (MIBC) are associated with substantial morbidity. Local release of doxorubicin (DOX) from phosphatidyldiglycerol-based thermosensitive liposomes (DPPG2-TSL-DOX) potentiated by hyperthermia (HT) in the bladder wall may result in bladder sparing without toxicity of systemic chemotherapy. We investigated whether this approach, compared to conventional DOX application, increases DOX concentrations in the bladder wall while limiting DOX in essential organs. Materials and Methods Twenty-one pigs were anaesthetized, and a urinary catheter equipped with a radiofrequency-emitting antenna for HT (60 minutes) was placed. Experimental groups consisted of iv low or full dose (20 or 60 mg/m2) DPPG2-TSL-DOX with/without HT, iv low dose (20 mg/m2) free DOX with HT, and full dose (50 mg/50 mL) intravesical DOX with/without HT. After the procedure, animals were immediately sacrificed. HPLC was used to measure DOX levels in the bladder, essential organs and serum, and fluorescence microscopy to evaluate DOX distribution in the bladder wall. Results Iv DPPG2-TSL-DOX with HT resulted in a significantly higher bladder wall DOX concentration which was more homogeneous distributed, than iv and intravesical free DOX administration with HT. Specifically in the detrusor, DPPG2-TSL-DOX with HT led to a >7- and 44-fold higher DOX concentration, compared to iv free DOX with HT and intravesical DOX, respectively. Organ DOX concentrations were significantly lower in heart and kidneys, and similar in liver, spleen and lungs, following iv DPPG2-TSL-DOX with HT, compared to iv free DOX. Intravesical DOX led to the lowest organ DOX concentrations. Conclusion Iv DPPG2-TSL-DOX combined with HT achieved higher DOX concentrations in the bladder wall including the detrusor, compared to conventional iv and intravesical DOX application. In combination with lower DOX accumulation in heart and kidneys, compared to iv free chemotherapy, DPPG2-TSL-DOX with HT has great potential to attain a role as a bladder-sparing treatment for MIBC.
Collapse
Affiliation(s)
| | - Iris S G Brummelhuis
- Department of Urology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lars H Lindner
- Department of Medicine III, University Hospital LMU Munich, Munich, Germany
| | - Felix Kuhnle
- Department of Medicine III, University Hospital LMU Munich, Munich, Germany
| | - Barbara Wedmann
- Department of Medicine III, University Hospital LMU Munich, Munich, Germany
| | | | | | - J Alfred Witjes
- Department of Urology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Egbert Oosterwijk
- Department of Urology, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
18
|
Evren E, Ringqvist E, Tripathi KP, Sleiers N, Rives IC, Alisjahbana A, Gao Y, Sarhan D, Halle T, Sorini C, Lepzien R, Marquardt N, Michaëlsson J, Smed-Sörensen A, Botling J, Karlsson MCI, Villablanca EJ, Willinger T. Distinct developmental pathways from blood monocytes generate human lung macrophage diversity. Immunity 2020; 54:259-275.e7. [PMID: 33382972 DOI: 10.1016/j.immuni.2020.12.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/15/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
The study of human macrophages and their ontogeny is an important unresolved issue. Here, we use a humanized mouse model expressing human cytokines to dissect the development of lung macrophages from human hematopoiesis in vivo. Human CD34+ hematopoietic stem and progenitor cells (HSPCs) generated three macrophage populations, occupying separate anatomical niches in the lung. Intravascular cell labeling, cell transplantation, and fate-mapping studies established that classical CD14+ blood monocytes derived from HSPCs migrated into lung tissue and gave rise to human interstitial and alveolar macrophages. In contrast, non-classical CD16+ blood monocytes preferentially generated macrophages resident in the lung vasculature (pulmonary intravascular macrophages). Finally, single-cell RNA sequencing defined intermediate differentiation stages in human lung macrophage development from blood monocytes. This study identifies distinct developmental pathways from circulating monocytes to lung macrophages and reveals how cellular origin contributes to human macrophage identity, diversity, and localization in vivo.
Collapse
Affiliation(s)
- Elza Evren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Emma Ringqvist
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Kumar Parijat Tripathi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Natalie Sleiers
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Inés Có Rives
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Arlisa Alisjahbana
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Yu Gao
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Tor Halle
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Chiara Sorini
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Rico Lepzien
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Nicole Marquardt
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Jakob Michaëlsson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Tim Willinger
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden.
| |
Collapse
|
19
|
Anderson SL, Duke-Novakovski T, Robinson AR, Townsend HGG, Singh B. Depletion of pulmonary intravascular macrophages rescues inflammation-induced delayed neutrophil apoptosis in horses. Am J Physiol Lung Cell Mol Physiol 2020; 320:L126-L136. [PMID: 33146566 DOI: 10.1152/ajplung.00392.2019] [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] Open
Abstract
The objective of this study was to determine the effect of pulmonary intravascular macrophage depletion on systemic inflammation and ex vivo neutrophil apoptosis using an experimental model of intestinal ischemia and reperfusion injury in horses. Neutrophils were isolated before and after surgery from horses that were randomized to three treatment groups, namely, sham celiotomy (CEL, n = 4), intestinal ischemia and reperfusion (IR, n = 6), and intestinal ischemia and reperfusion with gadolinium chloride treatment to deplete pulmonary intravascular macrophages (PIMs, IRGC, n = 6). Neutrophil apoptosis was assessed with Annexin V and propidium iodide staining quantified with flow cytometry and caspase-3, caspase-8, and caspase-9 activities in neutrophil lysates. All horses experienced a systemic inflammatory response following surgery. Following surgery, ex vivo neutrophil apoptosis was significantly delayed after 12 or 24 h in culture, except in IRGC horses (12 h: CEL: P = 0.03, IR: P = 0.05, IRGC: P = 0.2; 24 h: CEL: P = 0.001, IR: P = 0.004, IRGC: P = 0.3). Caspase-3, caspase-8, and caspase-9 activities were significantly reduced in neutrophils isolated after surgery and cultured for 12 h in IR horses, but not in IRGC horses (IR caspase-3: P = 0.002, IR caspase-8: P = 0.002, IR caspase-9: P = 0.04). Serum TNF-α concentration was increased in IRGC horses for 6-18 h following jejunal ischemia. Following surgery, ex vivo equine neutrophil apoptosis was delayed via downregulation of caspase activity, which was ameliorated by PIM depletion potentially via upregulation of TNF-α.
Collapse
Affiliation(s)
- Stacy L Anderson
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Tanya Duke-Novakovski
- Department of Small Animal Clinical Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Alexandra R Robinson
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Hugh G G Townsend
- Department of Large Animal Clinical Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Baljit Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
20
|
Wang A, Ali A, Keshavjee S, Liu M, Cypel M. Ex vivo lung perfusion for donor lung assessment and repair: a review of translational interspecies models. Am J Physiol Lung Cell Mol Physiol 2020; 319:L932-L940. [PMID: 32996780 DOI: 10.1152/ajplung.00295.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
For patients with end-stage lung disease, lung transplantation is a lifesaving therapy. Currently however, the number of patients who require a transplant exceeds the number of donor lungs available. One of the contributing factors to this is the conservative mindset of physicians who are concerned about transplanting marginal lungs due to the potential risk of primary graft dysfunction. Ex vivo lung perfusion (EVLP) technology has allowed for the expansion of donor pool of organs by enabling assessment and reconditioning of these marginal grafts before transplant. Ongoing efforts to optimize the therapeutic potential of EVLP are underway. Researchers have adopted the use of different large and small animal models to generate translational preclinical data. This includes the use of rejected human lungs, pig lungs, and rat lungs. In this review, we summarize some of the key current literature studies relevant to each of the major EVLP model platforms and identify the advantages and disadvantages of each platform. The review aims to guide investigators in choosing an appropriate species model to suit their specific goals of study, and ultimately aid in translation of therapy to meet the growing needs of the patient population.
Collapse
Affiliation(s)
- Aizhou Wang
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Aadil Ali
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Karagianni AE, Lisowski ZM, Hume DA, Scott Pirie R. The equine mononuclear phagocyte system: The relevance of the horse as a model for understanding human innate immunity. Equine Vet J 2020; 53:231-249. [PMID: 32881079 DOI: 10.1111/evj.13341] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/07/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022]
Abstract
The mononuclear phagocyte system (MPS) is a family of cells of related function that includes bone marrow progenitors, blood monocytes and resident tissue macrophages. Macrophages are effector cells in both innate and acquired immunity. They are a major resident cell population in every organ and their numbers increase in response to proinflammatory stimuli. Their function is highly regulated by a wide range of agonists, including lymphokines, cytokines and products of microorganisms. Macrophage biology has been studied most extensively in mice, yet direct comparisons of rodent and human macrophages have revealed many functional differences. In this review, we provide an overview of the equine MPS, describing the variation in the function and phenotype of macrophages depending on their location and the similarities and differences between the rodent, human and equine immune response. We discuss the use of the horse as a large animal model in which to study macrophage biology and pathological processes shared with humans. Finally, following the recent update to the horse genome, facilitating further comparative analysis of regulated gene expression between the species, we highlight the importance of future transcriptomic macrophage studies in the horse, the findings of which may also be applicable to human as well as veterinary research.
Collapse
Affiliation(s)
- Anna E Karagianni
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Zofia M Lisowski
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - David A Hume
- Mater Research Institute-UQ, Translational Research Institute, Woolloongabba, QLD, Australia
| | - R Scott Pirie
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| |
Collapse
|
22
|
Human Clinical Relevance of the Porcine Model of Pseudoallergic Infusion Reactions. Biomedicines 2020; 8:biomedicines8040082. [PMID: 32276476 PMCID: PMC7235862 DOI: 10.3390/biomedicines8040082] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/28/2022] Open
Abstract
Pigs provide a highly sensitive animal model for pseudoallergic infusion reactions, which are mild-to-severe hypersensitivity reactions (HSRs) that arise following intravenous administration of certain nanoparticulate drugs (nanomedicines) and other macromolecular structures. This model has been used in research for three decades and was also proposed by regulatory bodies for preclinical assessment of the risk of HSRs in the clinical stages of nano-drug development. However, there are views challenging the human relevance of the model and its utility in preclinical safety evaluation of nanomedicines. The argument challenging the model refers to the “global response” of pulmonary intravascular macrophages (PIM cells) in the lung of pigs, preventing the distinction of reactogenic from non-reactogenic particles, therefore overestimating the risk of HSRs relative to its occurrence in the normal human population. The goal of this review is to present the large body of experimental and clinical evidence negating the “global response” claim, while also showing the concordance of symptoms caused by different reactogenic nanoparticles in pigs and hypersensitive man. Contrary to the model’s demotion, we propose that the above features, together with the high reproducibility of quantifiable physiological endpoints, validate the porcine “complement activation-related pseudoallergy” (CARPA) model for safety evaluations. However, it needs to be kept in mind that the model is a disease model in the context of hypersensitivity to certain nanomedicines. Rather than toxicity screening, its main purpose is specific identification of HSR hazard, also enabling studies on the mechanism and mitigation of potentially serious HSRs.
Collapse
|
23
|
Spengler D, Rintz N, Krause MF. An Unsettled Promise: The Newborn Piglet Model of Neonatal Acute Respiratory Distress Syndrome (NARDS). Physiologic Data and Systematic Review. Front Physiol 2019; 10:1345. [PMID: 31736777 PMCID: PMC6831728 DOI: 10.3389/fphys.2019.01345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
Abstract
Despite great advances in mechanical ventilation and surfactant administration for the newborn infant with life-threatening respiratory failure no specific therapies are currently established to tackle major pro-inflammatory pathways. The susceptibility of the newborn infant with neonatal acute respiratory distress syndrome (NARDS) to exogenous surfactant is linked with a suppression of most of the immunologic responses by the innate immune system, however, additional corticosteroids applied in any severe pediatric lung disease with inflammatory background do not reduce morbidity or mortality and may even cause harm. Thus, the neonatal piglet model of acute lung injury serves as an excellent model to study respiratory failure and is the preferred animal model for reasons of availability, body size, similarities of porcine and human lung, robustness, and costs. In addition, similarities to the human toll-like receptor 4, the existence of intraalveolar macrophages, the sensitivity to lipopolysaccharide, and the production of nitric oxide make the piglet indispensable in anti-inflammatory research. Here we present the physiologic and immunologic data of newborn piglets from three trials involving acute lung injury secondary to repeated airway lavage (and others), mechanical ventilation, and a specific anti-inflammatory intervention via the intratracheal route using surfactant as a carrier substance. The physiologic data from many organ systems of the newborn piglet—but with preference on the lung—are presented here differentiating between baseline data from the uninjured piglet, the impact of acute lung injury on various parameters (24 h), and the follow up data after 72 h of mechanical ventilation. Data from the control group and the intervention groups are listed separately or combined. A systematic review of the newborn piglet meconium aspiration model and the repeated airway lavage model is finally presented. While many studies assessed lung injury scores, leukocyte infiltration, and protein/cytokine concentrations in bronchoalveolar fluid, a systematic approach to tackle major upstream pro-inflammatory pathways of the innate immune system is still in the fledgling stages. For the sake of newborn infants with life-threatening NARDS the newborn piglet model still is an unsettled promise offering many options to conquer neonatal physiology/immunology and to establish potent treatment modalities.
Collapse
Affiliation(s)
- Dietmar Spengler
- Department of Pediatrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Nele Rintz
- Department of Pediatrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Martin F Krause
- Department of Pediatrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
24
|
Liu H, Liu J, Huang J, Bai X, Wang Q. Heterogeneity and plasticity of porcine alveolar macrophage and pulmonary interstitial macrophage isolated from healthy pigs in vitro. Biol Open 2019; 8:bio.046342. [PMID: 31615770 PMCID: PMC6826289 DOI: 10.1242/bio.046342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
This study investigated the heterogeneity and plasticity of porcine alveolar macrophages (PAM) and pulmonary interstitial macrophages (IM) isolated from healthy pigs, including phenotype, function and gene expression. Dynamic changes of nitric oxide (NO) levels secreted by PAM and IM with stimulation of different doses of lipopolysaccharide (LPS) were investigated by Griess method, and the viability of the PAM and IM cells was investigated by MTT assay. Flow cytometry, fluorescence quantitative PCR and ELISA techniques were used to measure cell phenotype, gene expression and cytokine secretion, respectively. The PAM and IM cells in normal healthy pigs showed heterogeneity with 95.42±1.51% and 31.99±5.84% of CD163+ macrophage, respectively. The NO level in IM was significantly higher versus PAM after LPS treatment. Consistently, the ratio of Arg I/iNOS in IM was much lower than that in PAM, suggesting that the PAM belong to M2 macrophages and the IM belong to M1 macrophages. The PAM and IM cells in normal healthy pigs also showed plasticity. The Arg I/iNOS ratio and TIMP1/MMP12 ratio were significantly decreased in LPS- or LPS+IFNγ-treated PAM and IM, suggesting that cells were polarized towards M1 macrophages under LPS or LPS+IFNγ stimulation. On the contrary, IL-4 and IL-13 stimulation on PAM and IM lead to M2 polarization. A similar result was found in IL-1β gene expression and TNFα secretion. In conclusion, porcine macrophages have shown heterogeneity and plasticity on polarization under the stimulation of LPS, IFNγ, IL-4 and IL-13.
Collapse
Affiliation(s)
- Huan Liu
- College of Life Science and Technology, Dalian University, Dalian 116622, China
| | - Jia Liu
- Dalian Modern Agricultural Production Development Service Center, Dalian 116037, China
| | - Jing Huang
- College of Life Science and Technology, Dalian University, Dalian 116622, China
| | - Xianchang Bai
- College of Life Science and Technology, Dalian University, Dalian 116622, China
| | - Qinfu Wang
- College of Life Science and Technology, Dalian University, Dalian 116622, China
| |
Collapse
|
25
|
Weber B, Lackner I, Haffner-Luntzer M, Palmer A, Pressmar J, Scharffetter-Kochanek K, Knöll B, Schrezenemeier H, Relja B, Kalbitz M. Modeling trauma in rats: similarities to humans and potential pitfalls to consider. J Transl Med 2019; 17:305. [PMID: 31488164 PMCID: PMC6728963 DOI: 10.1186/s12967-019-2052-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/29/2019] [Indexed: 12/27/2022] Open
Abstract
Trauma is the leading cause of mortality in humans below the age of 40. Patients injured by accidents frequently suffer severe multiple trauma, which is life-threatening and leads to death in many cases. In multiply injured patients, thoracic trauma constitutes the third most common cause of mortality after abdominal injury and head trauma. Furthermore, 40-50% of all trauma-related deaths within the first 48 h after hospital admission result from uncontrolled hemorrhage. Physical trauma and hemorrhage are frequently associated with complex pathophysiological and immunological responses. To develop a greater understanding of the mechanisms of single and/or multiple trauma, reliable and reproducible animal models, fulfilling the ethical 3 R's criteria (Replacement, Reduction and Refinement), established by Russell and Burch in 'The Principles of Human Experimental Technique' (published 1959), are required. These should reflect both the complex pathophysiological and the immunological alterations induced by trauma, with the objective to translate the findings to the human situation, providing new clinical treatment approaches for patients affected by severe trauma. Small animal models are the most frequently used in trauma research. Rattus norvegicus was the first mammalian species domesticated for scientific research, dating back to 1830. To date, there exist numerous well-established procedures to mimic different forms of injury patterns in rats, animals that are uncomplicated in handling and housing. Nevertheless, there are some physiological and genetic differences between humans and rats, which should be carefully considered when rats are chosen as a model organism. The aim of this review is to illustrate the advantages as well as the disadvantages of rat models, which should be considered in trauma research when selecting an appropriate in vivo model. Being the most common and important models in trauma research, this review focuses on hemorrhagic shock, blunt chest trauma, bone fracture, skin and soft-tissue trauma, burns, traumatic brain injury and polytrauma.
Collapse
Affiliation(s)
- Birte Weber
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Ina Lackner
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma-Immunology, University of Ulm, Ulm, Germany
| | - Jochen Pressmar
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| | | | - Bernd Knöll
- Institute of Physiological Chemistry, University of Ulm, Ulm, Germany
| | - Hubert Schrezenemeier
- Institute of Transfusion Medicine, University of Ulm and Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg – Hessen and University Hospital Ulm, Ulm, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, Frankfurt, Germany
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm Medical School, Albert-Einstein-Allee 23, 89081 Ulm, Germany
| |
Collapse
|
26
|
Moghimi SM, Simberg D, Skotland T, Yaghmur A, Hunter AC. The Interplay Between Blood Proteins, Complement, and Macrophages on Nanomedicine Performance and Responses. J Pharmacol Exp Ther 2019; 370:581-592. [PMID: 30940695 PMCID: PMC11047092 DOI: 10.1124/jpet.119.258012] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 03/28/2019] [Indexed: 12/17/2022] Open
Abstract
In the blood, depending on their physicochemical characteristics, nanoparticles attract a wide range of plasma biomolecules. The majority of blood biomolecules bind nonspecifically to nanoparticles. On the other hand, biomolecules such as pattern-recognition complement-sensing proteins may recognize some structural determinants of the pristine surface, causing complement activation. Adsorption of nonspecific blood proteins could also recruit natural antibodies and initiate complement activation, and this seems to be a global process with many preclinical and clinical nanomedicines. We discuss these issues, since complement activation has ramifications in nanomedicine stability and pharmacokinetics, as well as in inflammation and disease progression. Some studies have also predicted a role for complement systems in infusion-related reactions, whereas others show a direct role for macrophages and other immune cells independent of complement activation. We comment on these discrepancies and suggest directions for exploring the underlying mechanisms.
Collapse
Affiliation(s)
- S Moein Moghimi
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| | - Dmitri Simberg
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| | - Tore Skotland
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| | - Anan Yaghmur
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| | - A Christy Hunter
- School of Pharmacy and Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (S.M.M.); Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus (S.M.M., D.S.), and Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences (D.S.), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway (T.S.); Department of Pharmacy, University of Copenhagen, Copenhagen Ø, Denmark (A.Y.); and Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, United Kingdom (A.C.H.)
| |
Collapse
|
27
|
Valic MS, Zheng G. Research tools for extrapolating the disposition and pharmacokinetics of nanomaterials from preclinical animals to humans. Theranostics 2019; 9:3365-3387. [PMID: 31244958 PMCID: PMC6567967 DOI: 10.7150/thno.34509] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/26/2019] [Indexed: 11/30/2022] Open
Abstract
A critical step in the translational science of nanomaterials from preclinical animal studies to humans is the comprehensive investigation of their disposition (or ADME) and pharmacokinetic behaviours. Disposition and pharmacokinetic data are ideally collected in different animal species (rodent and nonrodent), at different dose levels, and following multiple administrations. These data are used to assess the systemic exposure and effect to nanomaterials, primary determinants of their potential toxicity and therapeutic efficacy. At toxic doses in animal models, pharmacokinetic (termed toxicokinetic) data are related to toxicologic findings that inform the design of nonclinical toxicity studies and contribute to the determination of the maximum recommended starting dose in clinical phase 1 trials. Nanomaterials present a unique challenge for disposition and pharmacokinetic investigations owing to their prolonged circulation times, nonlinear pharmacokinetic profiles, and their extensive distribution into tissues. Predictive relationships between nanomaterial physicochemical properties and behaviours in vivo are lacking and are confounded by anatomical, physiological, and immunological differences amongst preclinical animal models and humans. These challenges are poorly understood and frequently overlooked by investigators, leading to inaccurate assumptions of disposition, pharmacokinetic, and toxicokinetics profiles across species that can have profoundly detrimental impacts for nonclinical toxicity studies and clinical phase 1 trials. Herein are highlighted two research tools for analysing and interpreting disposition and pharmacokinetic data from multiple species and for extrapolating this data accurately in humans. Empirical methodologies and mechanistic mathematical modelling approaches are discussed with emphasis placed on important considerations and caveats for representing nanomaterials, such as the importance of integrating physiological variables associated with the mononuclear phagocyte system (MPS) into extrapolation methods for nanomaterials. The application of these tools will be examined in recent examples of investigational and clinically approved nanomaterials. Finally, strategies for applying these extrapolation tools in a complementary manner to perform dose predictions and in silico toxicity assessments in humans will be explained. A greater familiarity with the available tools and prior experiences of extrapolating nanomaterial disposition and pharmacokinetics from preclinical animal models to humans will hopefully result in a more straightforward roadmap for the clinical translation of promising nanomaterials.
Collapse
Affiliation(s)
- Michael S. Valic
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, CANADA, M5G 1L7
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, CANADA, M5G 1L7
- Department of Medical Biophysics, Institute of Biomaterials and Biomedical Engineering, and Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, CANADA, M5G 1L7
| |
Collapse
|
28
|
Starbæk SMR, Brogaard L, Dawson HD, Smith AD, Heegaard PMH, Larsen LE, Jungersen G, Skovgaard K. Animal Models for Influenza A Virus Infection Incorporating the Involvement of Innate Host Defenses: Enhanced Translational Value of the Porcine Model. ILAR J 2018; 59:323-337. [PMID: 30476076 DOI: 10.1093/ilar/ily009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 06/19/2018] [Indexed: 01/05/2025] Open
Abstract
Influenza is a viral respiratory disease having a major impact on public health. Influenza A virus (IAV) usually causes mild transitory disease in humans. However, in specific groups of individuals such as severely obese, the elderly, and individuals with underlying inflammatory conditions, IAV can cause severe illness or death. In this review, relevant small and large animal models for human IAV infection, including the pig, ferret, and mouse, are discussed. The focus is on the pig as a large animal model for human IAV infection as well as on the associated innate immune response. Pigs are natural hosts for the same IAV subtypes as humans, they develop clinical disease mirroring human symptoms, they have similar lung anatomy, and their respiratory physiology and immune responses to IAV infection are remarkably similar to what is observed in humans. The pig model shows high face and target validity for human IAV infection, making it suitable for modeling many aspects of influenza, including increased risk of severe disease and impaired vaccine response due to underlying pathologies such as low-grade inflammation. Comparative analysis of proteins involved in viral pattern recognition, interferon responses, and regulation of interferon-stimulated genes reveals a significantly higher degree of similarity between pig, ferret, and human compared with mice. It is concluded that the pig is a promising animal model displaying substantial human translational value with the ability to provide essential insights into IAV infection, pathogenesis, and immunity.
Collapse
Affiliation(s)
- Sofie M R Starbæk
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Louise Brogaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Harry D Dawson
- Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland
| | - Allen D Smith
- Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, Maryland
| | - Peter M H Heegaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lars E Larsen
- National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gregers Jungersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Kerstin Skovgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
29
|
Szebeni J, Simberg D, González-Fernández Á, Barenholz Y, Dobrovolskaia MA. Roadmap and strategy for overcoming infusion reactions to nanomedicines. NATURE NANOTECHNOLOGY 2018; 13:1100-1108. [PMID: 30348955 PMCID: PMC6320688 DOI: 10.1038/s41565-018-0273-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/03/2018] [Indexed: 05/20/2023]
Abstract
Infusion reactions (IRs) are complex, immune-mediated side effects that mainly occur within minutes to hours of receiving a therapeutic dose of intravenously administered pharmaceutical products. These products are diverse and include both traditional pharmaceuticals (for example biological agents and small molecules) and new ones (for example nanotechnology-based products). Although IRs are not unique to nanomedicines, they represent a hurdle for the translation of nanotechnology-based drug products. This Perspective offers a big picture of the pharmaceutical field and examines current understanding of mechanisms responsible for IRs to nanomedicines. We outline outstanding questions, review currently available experimental evidence to provide some answers and highlight the gaps. We review advantages and limitations of the in vitro tests and animal models used for studying IRs to nanomedicines. Finally, we propose a roadmap to improve current understanding, and we recommend a strategy for overcoming the problem.
Collapse
Affiliation(s)
- Janos Szebeni
- Nanomedicine Research and Education Center, Institute of Pathophysiology, Semmelweis University, Budapest, Hungary
- SeroScience Ltd, Budapest, Hungary
- Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO, USA
| | - África González-Fernández
- Immunology, Centro de Investigaciones Biomédicas (CINBIO), Centro de Investigación Singular de Galicia, Instituto de Investigación Sanitaria Galicia Sur (IIS-GS), University of Vigo, Vigo, Spain
| | - Yechezkel Barenholz
- Department of Biochemistry, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
30
|
Granton E, Kim JH, Podstawka J, Yipp BG. The Lung Microvasculature Is a Functional Immune Niche. Trends Immunol 2018; 39:890-899. [PMID: 30253910 DOI: 10.1016/j.it.2018.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/28/2018] [Accepted: 09/04/2018] [Indexed: 01/15/2023]
Abstract
Lung capillaries, best known for vital gas exchange, also contribute to neutrophil margination, a phenomenon resulting in large numbers of pulmonary vascular neutrophils. Importantly, the functional relevance of neutrophil margination is unknown. Recent advances in microscopy have altered our understanding of why neutrophils marginate. Specifically, data show that lung capillaries provide a unique anatomical site for neutrophils to capture bloodstream pathogens, which contrasts the conventional monophagocytic-dominated vascular host defense of the spleen and liver. Moreover, lung capillaries provide an efficient site for direct cell-cell communication required for the induction of apoptosis in aged neutrophils. These new ideas transform our views of the pulmonary circulation as a site for immediate neutrophil-mediated host defense and regulation of their life cycle.
Collapse
Affiliation(s)
- Elise Granton
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Co-first authors, equal contribution
| | - Jung Hwan Kim
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Current address: Systems Genomics and Bioinformatics Unit, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Co-first authors, equal contribution
| | - John Podstawka
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Co-first authors, equal contribution
| | - Bryan G Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
31
|
Núñez A, Sánchez-Cordón PJ, Pedrera M, Gómez-Villamandos JC, Carrasco L. Pulmonary intravascular macrophages regulate the pathogenetic mechanisms of pulmonary lesions during acute courses of classical swine fever. Transbound Emerg Dis 2018; 65:1885-1897. [DOI: 10.1111/tbed.12970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/15/2018] [Accepted: 07/03/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Alejandro Núñez
- Department of Comparative Pathology; Veterinary Faculty; University of Córdoba; Córdoba Spain
| | - Pedro J. Sánchez-Cordón
- Department of Comparative Pathology; Veterinary Faculty; University of Córdoba; Córdoba Spain
| | - Miriam Pedrera
- Department of Comparative Pathology; Veterinary Faculty; University of Córdoba; Córdoba Spain
| | | | - Librado Carrasco
- Department of Comparative Pathology; Veterinary Faculty; University of Córdoba; Córdoba Spain
| |
Collapse
|
32
|
Brenner JS, Pan DC, Myerson JW, Marcos-Contreras OA, Villa CH, Patel P, Hekierski H, Chatterjee S, Tao JQ, Parhiz H, Bhamidipati K, Uhler TG, Hood ED, Kiseleva RY, Shuvaev VS, Shuvaeva T, Khoshnejad M, Johnston I, Gregory JV, Lahann J, Wang T, Cantu E, Armstead WM, Mitragotri S, Muzykantov V. Red blood cell-hitchhiking boosts delivery of nanocarriers to chosen organs by orders of magnitude. Nat Commun 2018; 9:2684. [PMID: 29992966 PMCID: PMC6041332 DOI: 10.1038/s41467-018-05079-7] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Drug delivery by nanocarriers (NCs) has long been stymied by dominant liver uptake and limited target organ deposition, even when NCs are targeted using affinity moieties. Here we report a universal solution: red blood cell (RBC)-hitchhiking (RH), in which NCs adsorbed onto the RBCs transfer from RBCs to the first organ downstream of the intravascular injection. RH improves delivery for a wide range of NCs and even viral vectors. For example, RH injected intravenously increases liposome uptake in the first downstream organ, lungs, by ~40-fold compared with free NCs. Intra-carotid artery injection of RH NCs delivers >10% of the injected NC dose to the brain, ~10× higher than that achieved with affinity moieties. Further, RH works in mice, pigs, and ex vivo human lungs without causing RBC or end-organ toxicities. Thus, RH is a clinically translatable platform technology poised to augment drug delivery in acute lung disease, stroke, and several other diseases.
Collapse
Affiliation(s)
- Jacob S Brenner
- Pulmonary, Allergy, & Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Daniel C Pan
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jacob W Myerson
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Oscar A Marcos-Contreras
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Carlos H Villa
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Transfusion Medicine and Therapeutic Pathology, Department of Pathology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Priyal Patel
- Pulmonary, Allergy, & Critical Care Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hugh Hekierski
- Department of Anesthesiology & Critical Care, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Shampa Chatterjee
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jian-Qin Tao
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hamideh Parhiz
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kartik Bhamidipati
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Thomas G Uhler
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Elizabeth D Hood
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Raisa Yu Kiseleva
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vladimir S Shuvaev
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tea Shuvaeva
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Makan Khoshnejad
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ian Johnston
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason V Gregory
- Department of Chemical Engineering and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joerg Lahann
- Department of Chemical Engineering and Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tao Wang
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Edward Cantu
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William M Armstead
- Department of Anesthesiology & Critical Care, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Samir Mitragotri
- School of Engineering & Applied Sciences, Harvard University, Wyss Institute, Cambridge, MA, 02138, USA
| | - Vladimir Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics and Center for Translational Targeted Therapeutics and Nanomedicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
33
|
Moghimi SM, Simberg D. Translational gaps in animal models of human infusion reactions to nanomedicines. Nanomedicine (Lond) 2018; 13:973-975. [DOI: 10.2217/nnm-2018-0064] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Adverse infusion reactions to regulatory approved nanomedicines in human subjects are idiosyncratic, but outwardly reproducible in pigs. A large body of evidence suggests that the porcine reactions are related to robust nanoparticle clearance by pulmonary intravascular macrophages (PIMs), and rapid release of arachidonate metabolites from these cells. Similar to pigs, other animals that have resident PIMs in their lungs also respond to intravenously injected particles, where rapid particle clearance by PIMs correlate with peak periods of cardiopulmonary distress. Normal human lungs, however, do not have PIMs, but ‘induced’ PIMs have been identified in pulmonary circulation under certain pathological conditions. We question suitability, and limitation of these preclinical models for global assessment of nanomedicine safety, and discuss alternative models and approaches.
Collapse
Affiliation(s)
- S Moein Moghimi
- School of Pharmacy, The Faculty of Medical Sciences, King George VI Building, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
- Division of Stratified Medicine, Biomarkers & Therapeutics, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
- Translational Bio-Nanosciences Laboratory & Colorado Center for Nanomedicine & Nanosafety, The Skaggs School of Pharmacy & Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, 1250 East Mountview Blvd, Aurora, CO 80045, USA
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory & Colorado Center for Nanomedicine & Nanosafety, The Skaggs School of Pharmacy & Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, 1250 East Mountview Blvd, Aurora, CO 80045, USA
| |
Collapse
|
34
|
Parhiz H, Khoshnejad M, Myerson JW, Hood E, Patel PN, Brenner JS, Muzykantov VR. Unintended effects of drug carriers: Big issues of small particles. Adv Drug Deliv Rev 2018; 130:90-112. [PMID: 30149885 PMCID: PMC6588191 DOI: 10.1016/j.addr.2018.06.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/11/2018] [Accepted: 06/26/2018] [Indexed: 02/06/2023]
Abstract
Humoral and cellular host defense mechanisms including diverse phagocytes, leukocytes, and immune cells have evolved over millions of years to protect the body from microbes and other external and internal threats. These policing forces recognize engineered sub-micron drug delivery systems (DDS) as such a threat, and react accordingly. This leads to impediment of the therapeutic action, extensively studied and discussed in the literature. Here, we focus on side effects of DDS interactions with host defenses. We argue that for nanomedicine to reach its clinical potential, the field must redouble its efforts in understanding the interaction between drug delivery systems and the host defenses, so that we can engineer safer interventions with the greatest potential for clinical success.
Collapse
Affiliation(s)
- Hamideh Parhiz
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Makan Khoshnejad
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob W Myerson
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Hood
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Priyal N Patel
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob S Brenner
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Vladimir R Muzykantov
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Targeted Therapeutics and Translational Nanomedicine (CT3N), University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Descotes J, Allais L, Ancian P, Pedersen HD, Friry-Santini C, Iglesias A, Rubic-Schneider T, Skaggs H, Vestbjerg P. Nonclinical evaluation of immunological safety in Göttingen Minipigs: The CONFIRM initiative. Regul Toxicol Pharmacol 2018; 94:271-275. [PMID: 29481836 DOI: 10.1016/j.yrtph.2018.02.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/05/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
There is a growing need to consider non-rodent species for the immunological safety evaluation of drug candidates. The EU Framework-6 RETHINK Project demonstrated that the Göttingen Minipig is a relevant animal model for regulatory toxicology studies. Extensive knowledge on the immune system of domestic pigs is available and fewer differences from humans have been identified as compared to other species, such as mice or non-human primates. Minipig data are too scarce to allow for claiming full immunological comparability with domestic pigs. Another gap limiting minipig use for immunological safety evaluation is the lack of a qualified and validated database. However, available data lend support to the use of minipigs. The need for a COllaborative Network For Immunological safety Research in Minipigs (the CONFIRM Initiative) was obvious. It is intended to trigger immunological safety research in Göttingen Minipigs, to assist and synergize fundamental, translational and regulatory investigative efforts relevant to the immunological safety evaluation of pharmaceuticals and biologics, and to spread current knowledge and new findings to the scientific and regulatory toxicology community.
Collapse
Affiliation(s)
- Jacques Descotes
- ImmunoSafe Consulting & University of Lyon, 38480 Saint Jean d'Avelanne, France.
| | - Linda Allais
- Charles River Laboratories, 69210 Saint Germain-Nuelles, France
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Schneberger D, Sethi RS, Singh B. Comparative View of Lung Vascular Endothelium of Cattle, Horses, and Water Buffalo. MOLECULAR AND FUNCTIONAL INSIGHTS INTO THE PULMONARY VASCULATURE 2018; 228:21-39. [DOI: 10.1007/978-3-319-68483-3_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
Moghimi SM. Nanomedicine safety in preclinical and clinical development: focus on idiosyncratic injection/infusion reactions. Drug Discov Today 2017; 23:1034-1042. [PMID: 29146517 DOI: 10.1016/j.drudis.2017.11.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/20/2017] [Accepted: 11/09/2017] [Indexed: 11/18/2022]
Abstract
Injection/infusion reactions to nanopharmaceuticals (and particulate drug carriers) are idiosyncratic and well documented. The molecular basis of nanoparticle-mediated injection reactions is debatable, with two hypotheses as front-runners. The first is complement-activation-related 'pseudoallergy', where a causal role for nanoparticle-mediated complement activation in injection/infusion reactions is considered. However, the second hypothesis (the rapid phagocytic response hypothesis) states a transitional link from robust clearance of nanoparticles (NPs) from the blood by strategically placed responsive macrophages to adverse hemodynamic and cardiopulmonary reactions, regardless of complement activation. Here, I critically examine and discuss these hypotheses. Current experimentally derived evidence appears to be more in support of the rapid phagocytic response hypothesis than of the 'pseudoallergy' hypothesis.
Collapse
Affiliation(s)
- Seyed Moein Moghimi
- School of Pharmacy, The Faculty of Medical Sciences, King George VI Building, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; Division of Stratified Medicine, Biomarkers & Therapeutics, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
38
|
Molecular Ultrasound Imaging of αvβ3-Integrin Expression in Carotid Arteries of Pigs After Vessel Injury. Invest Radiol 2017; 51:767-775. [PMID: 27119438 DOI: 10.1097/rli.0000000000000282] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Interventions such as balloon angioplasty can cause vascular injury leading to platelet activation, thrombus formation, and inflammatory response. This induces vascular smooth muscle cell activation and subsequent re-endothelialization with expression of αvβ3-integrin by endothelial cells and vascular smooth muscle cell. Thus, poly-N-butylcyanoacrylate microbubbles (MBs) targeted to αvβ3-integrin were evaluated for monitoring vascular healing after vessel injury in pigs using molecular ultrasound imaging. MATERIALS AND METHODS Approval for animal experiments was obtained. The binding specificity of αvβ3-integrin-targeted MB to human umbilical vein endothelial cells was tested with fluorescence microscopy. In vivo imaging was performed using a clinical ultrasound system and an 8-MHz probe. Six mini pigs were examined after vessel injury in the left carotid artery. The right carotid served as control. Uncoated MB, cDRG-coated MB, and αvβ3-integrin-specific cRGD-coated MB were injected sequentially. Bound MBs were assessed 8 minutes after injection using ultrasound replenishment analysis. Measurements were performed 2 hours, 1 and 5 weeks, and 3 and 6 months after injury. In vivo data were validated by immunohistochemistry. RESULTS Significantly stronger binding of cRGD-MB than MB and cDRG-MB to human umbilical vein endothelial cells was found (P < 0.01). As vessel injury leads to upregulation of αvβ3-integrin, cRGD-MBs bound significantly stronger (P < 0.05) in injured carotid arteries than at the counter side 1 week after vessel injury and significant differences could also be observed after 5 weeks. After 3 months, αvβ3-integrin expression decreased to baseline and binding of cRGD-MB was comparable in both vessels. Values remained at baseline also after 6 months. CONCLUSIONS Ultrasound imaging with RGD-MB is promising for monitoring vascular healing after vessel injury. This may open new perspectives to assess vascular damage after radiological interventions.
Collapse
|
39
|
Yamada S, Kawaguchi H, Yamada T, Guo X, Matsuo K, Hamada T, Miura N, Tasaki T, Tanimoto A. Cholic Acid Enhances Visceral Adiposity, Atherosclerosis and Nonalcoholic Fatty Liver Disease in Microminipigs. J Atheroscler Thromb 2017; 24:1150-1166. [PMID: 28496045 PMCID: PMC5684480 DOI: 10.5551/jat.39909] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/21/2017] [Indexed: 01/02/2023] Open
Abstract
AIM We have recently established a novel swine model for studies of atherosclerosis using MicrominipigsTM (µMPs) fed a high-fat/high-cholesterol diet (HcD). Using this swine model, we re-evaluated the effects of dietary cholic acid (CA) on serum lipid profile, atherosclerosis and hepatic injuries. METHODS The µMPs were fed HcD supplemented with 0.7% CA (HcD+CA) for eight weeks, and the effect of CA on serum lipoprotein levels, expression of oxidative stress markers, adiposity and lesion formation in the aorta, liver, and other organs was investigated. RESULTS The HcD+CA-fed group exhibited more visceral adiposity, progression of atherosclerosis and higher serum levels of oxidative stress markers than the HcD-fed group, even though they showed similar serum lipid levels. The liver demonstrated increased lipid accumulation, higher expression of oxidative stress markers, accelerated activation of foamy Kupffer cells and stellate cells, and increased hepatocyte apoptosis, indicating non-alcoholic fatty liver disease (NAFLD). Intriguingly, foamy macrophage mobilization was observed in various organs, including the reticuloendothelial system, pulmonary capillary vessels and skin very often in HcD+CA-fed µMPs. CONCLUSION To our knowledge, this is the first large animal model, in which visceral obesity, NAFLD and atherosclerosis are concomitantly induced by dietary manipulation. These data suggest the detrimental effects of CA, potentially through local and systemic activation of oxidative stress-induced signaling to macrophage mobilization, on the acceleration of visceral adiposity, atherosclerosis and NAFLD.
Collapse
Affiliation(s)
- Sohsuke Yamada
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hiroaki Kawaguchi
- Department of Hygiene and Health Promotion Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | | | - Xin Guo
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kei Matsuo
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Taiji Hamada
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Naoki Miura
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| | - Takashi Tasaki
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akihide Tanimoto
- Department of Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
40
|
Abstract
Tuberculosis remains one of the greatest threats to human health. The causative bacterium, Mycobacterium tuberculosis, is acquired by the respiratory route. It is exquisitely adapted to humans and is a prototypic intracellular pathogen of macrophages, with alveolar macrophages being the primary conduit of infection and disease. However, M. tuberculosis bacilli interact with and are affected by several soluble and cellular components of the innate immune system which dictate the outcome of primary infection, most commonly a latently infected healthy human host, in whom the bacteria are held in check by the host immune response within the confines of tissue granuloma, the host histopathologic hallmark. Such individuals can develop active TB later in life with impairment in the immune system. In contrast, in a minority of infected individuals, the early host immune response fails to control bacterial growth, and progressive granulomatous disease develops, facilitating spread of the bacilli via infectious aerosols. The molecular details of the M. tuberculosis-host innate immune system interaction continue to be elucidated, particularly those occurring within the lung. However, it is clear that a number of complex processes are involved at the different stages of infection that may benefit either the bacterium or the host. In this article, we describe a contemporary view of the molecular events underlying the interaction between M. tuberculosis and a variety of cellular and soluble components and processes of the innate immune system.
Collapse
|
41
|
Wibroe PP, Anselmo AC, Nilsson PH, Sarode A, Gupta V, Urbanics R, Szebeni J, Hunter AC, Mitragotri S, Mollnes TE, Moghimi SM. Bypassing adverse injection reactions to nanoparticles through shape modification and attachment to erythrocytes. NATURE NANOTECHNOLOGY 2017; 12:589-594. [PMID: 28396605 DOI: 10.1038/nnano.2017.47] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 02/27/2017] [Indexed: 05/18/2023]
Abstract
Intravenously injected nanopharmaceuticals, including PEGylated nanoparticles, induce adverse cardiopulmonary reactions in sensitive human subjects, and these reactions are highly reproducible in pigs. Although the underlying mechanisms are poorly understood, roles for both the complement system and reactive macrophages have been implicated. Here, we show the dominance and importance of robust pulmonary intravascular macrophage clearance of nanoparticles in mediating adverse cardiopulmonary distress in pigs irrespective of complement activation. Specifically, we show that delaying particle recognition by macrophages within the first few minutes of injection overcomes adverse reactions in pigs using two independent approaches. First, we changed the particle geometry from a spherical shape (which triggers cardiopulmonary distress) to either rod- or disk-shape morphology. Second, we physically adhered spheres to the surface of erythrocytes. These strategies, which are distinct from commonly leveraged stealth engineering approaches such as nanoparticle surface functionalization with poly(ethylene glycol) and/or immunological modulators, prevent robust macrophage recognition, resulting in the reduction or mitigation of adverse cardiopulmonary distress associated with nanopharmaceutical administration.
Collapse
Affiliation(s)
- Peter Popp Wibroe
- Nanomedicine Laboratory, Centre for Pharmaceutical Nanotechnology and Nanotoxicology, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Aaron C Anselmo
- Department of Chemical Engineering and Center for Bioengineering, University of California at Santa Barbara, Santa Barbara, California 93106, USA
| | - Per H Nilsson
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- K.G. Jebsen IRC, University of Oslo, 0372 Oslo, Norway
- Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, 391 82 Kalmar, Sweden
| | - Apoorva Sarode
- Department of Chemical Engineering and Center for Bioengineering, University of California at Santa Barbara, Santa Barbara, California 93106, USA
| | - Vivek Gupta
- College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York 11439, USA
| | - Rudolf Urbanics
- Nanomedicine Research and Education Center, Semmelweis University, Budapest &SeroScience Ltd, Budapest, Hungary
| | - Janos Szebeni
- Nanomedicine Research and Education Center, Semmelweis University, Budapest &SeroScience Ltd, Budapest, Hungary
| | - Alan Christy Hunter
- Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Samir Mitragotri
- Department of Chemical Engineering and Center for Bioengineering, University of California at Santa Barbara, Santa Barbara, California 93106, USA
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway
- K.G. Jebsen IRC, University of Oslo, 0372 Oslo, Norway
- Reserach Laboratory, Nordland Hospital, 8092 Bodø, Norway
- K.G. Jebsen TREC, University of Tromsø, 9037 Tromsø, Norway
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Seyed Moein Moghimi
- Nanomedicine Laboratory, Centre for Pharmaceutical Nanotechnology and Nanotoxicology, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
- Nano-Science Center, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen Ø, Denmark
- School of Medicine, Pharmacy and Health, Durham University, Queen's Campus, Stockton-on-Tees TS17 6BH, UK
| |
Collapse
|
42
|
Thanabalasuriar A, Surewaard BG, Willson ME, Neupane AS, Stover CK, Warrener P, Wilson G, Keller AE, Sellman BR, DiGiandomenico A, Kubes P. Bispecific antibody targets multiple Pseudomonas aeruginosa evasion mechanisms in the lung vasculature. J Clin Invest 2017; 127:2249-2261. [PMID: 28463232 DOI: 10.1172/jci89652] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 03/02/2017] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa is a major cause of severe infections that lead to bacteremia and high patient mortality. P. aeruginosa has evolved numerous evasion and subversion mechanisms that work in concert to overcome immune recognition and effector functions in hospitalized and immunosuppressed individuals. Here, we have used multilaser spinning-disk intravital microscopy to monitor the blood-borne stage in a murine bacteremic model of P. aeruginosa infection. P. aeruginosa adhered avidly to lung vasculature, where patrolling neutrophils and other immune cells were virtually blind to the pathogen's presence. This cloaking phenomenon was attributed to expression of Psl exopolysaccharide. Although an anti-Psl mAb activated complement and enhanced neutrophil recognition of P. aeruginosa, neutrophil-mediated clearance of the pathogen was suboptimal owing to a second subversion mechanism, namely the type 3 secretion (T3S) injectisome. Indeed, T3S prevented phagosome acidification and resisted killing inside these compartments. Antibody-mediated inhibition of the T3S protein PcrV did not enhance bacterial phagocytosis but did enhance killing of the few bacteria ingested by neutrophils. A bispecific mAb targeting both Psl and PcrV enhanced neutrophil uptake of P. aeruginosa and also greatly increased inhibition of T3S function, allowing for phagosome acidification and bacterial killing. These data highlight the need to block multiple evasion and subversion mechanisms in tandem to kill P. aeruginosa.
Collapse
Affiliation(s)
- Ajitha Thanabalasuriar
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Bas Gj Surewaard
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Michelle E Willson
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Arpan S Neupane
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | - Paul Kubes
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
43
|
Yipp BG, Kim JH, Lima R, Zbytnuik LD, Petri B, Swanlund N, Ho M, Szeto VG, Tak T, Koenderman L, Pickkers P, Tool ATJ, Kuijpers TW, van den Berg TK, Looney MR, Krummel MF, Kubes P. The Lung is a Host Defense Niche for Immediate Neutrophil-Mediated Vascular Protection. Sci Immunol 2017. [PMID: 28626833 DOI: 10.1126/sciimmunol.aam8929] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bloodstream infection is a hallmark of sepsis, a medically emergent condition requiring rapid treatment. However, upregulation of host defense proteins through toll-like receptors and NFκB requires hours after endotoxin detection. Using confocal pulmonary intravital microscopy, we identified that the lung provides a TLR4-Myd88-and abl tyrosine kinase-dependent niche for immediate CD11b-dependent neutrophil responses to endotoxin and Gram-negative bloodstream pathogens. In an in vivo model of bacteremia, neutrophils crawled to and rapidly phagocytosed Escherichia coli sequestered to the lung endothelium. Therefore, the lung capillaries provide a vascular defensive niche whereby endothelium and neutrophils cooperate for immediate detection and capture of disseminating pathogens.
Collapse
Affiliation(s)
- Bryan G Yipp
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jung Hwan Kim
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ronald Lima
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lori D Zbytnuik
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Björn Petri
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nick Swanlund
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - May Ho
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Vivian G Szeto
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Tamar Tak
- Department of Respiratory Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands Emma Children's Hospital, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Leo Koenderman
- Department of Respiratory Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands Emma Children's Hospital, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Anton T J Tool
- Department of Blood Cell Research, Sanquin Research, and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Blood Cell Research, Sanquin Research, and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Timo K van den Berg
- Department of Blood Cell Research, Sanquin Research, and Landsteiner Laboratory, Amsterdam, The Netherlands.,Department of Molecular Cell Biology and Immunology, VU Medical Center, Amsterdam, The Netherlands
| | - Mark R Looney
- Departments of Medicine and Laboratory Medicine, University of California, San Francisco, 513 Parnassus Avenue, HSW512, California 94143-0511, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, 513 Parnassus Ave, HSW512, San Francisco, California 94143-0511, USA
| | - Paul Kubes
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
44
|
Harrison JME, Quanstrom LM, Robinson AR, Wobeser B, Anderson SL, Singh B. Expression of von Willebrand factor, pulmonary intravascular macrophages, and Toll-like receptors in lungs of septic foals. J Vet Sci 2017; 18:17-23. [PMID: 27297419 PMCID: PMC5366298 DOI: 10.4142/jvs.2017.18.1.17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/02/2016] [Accepted: 05/12/2016] [Indexed: 01/19/2023] Open
Abstract
Sepsis causes significant mortality in neonatal foals; however, there is little data describing the cellular and molecular pathways of lung inflammation in septic foals. This study was conducted to characterize lung inflammation in septic foals. Lung tissue sections from control (n = 6) and septic (n = 17) foals were compared using histology and immunohistology. Blinded pathologic scoring of hematoxylin and eosin stained samples revealed increased features of lung inflammation such as thickened alveolar septa and sequestered inflammatory cells in septic foals. Septic foal lungs showed increased expression of von Willebrand factor in blood vessels, demonstrating vascular inflammation. Use of MAC387 antibody to detect calprotectin as a reflection of mononuclear cell infiltration revealed a significant increase in their numbers in alveolar septa of lungs from septic foals compared to those from control foals. The mononuclear cells appeared to be mature macrophages and were located in the septal capillaries, suggesting they were pulmonary intravascular macrophages (PIMs). Finally, lungs from septic foals showed increased expression of Toll-like receptor 4 and 9 in mononuclear cells relative to the control. Taken together, this study is the first to show the expression of inflammatory molecules and an increase in PIMs in lungs from foals that died from sepsis.
Collapse
Affiliation(s)
- Jacqueline M E Harrison
- Department of Veterinary Biomedical Sciences Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Leah M Quanstrom
- Department of Veterinary Biomedical Sciences Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Alex R Robinson
- Department of Veterinary Biomedical Sciences Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Bruce Wobeser
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Stacy L Anderson
- Department of Veterinary Biomedical Sciences Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Baljit Singh
- Department of Veterinary Biomedical Sciences Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| |
Collapse
|
45
|
Abstract
Respiratory immunity is accomplished using multiple mechanisms including structure/anatomy of the respiratory tract, mucosal defense in the form of the mucociliary apparatus, innate immunity using cells and molecules and acquired immunity. There are species differences of the respiratory immune system that influence the response to environmental challenges and pharmaceutical, industrial and agricultural compounds assessed in nonclinical safety testing and hazard identification. These differences influence the interpretation of respiratory system changes after exposure to these challenges and compounds in nonclinical safety assessment and hazard identification and their relevance to humans.
Collapse
|
46
|
Pulmonary innate inflammatory responses to agricultural occupational contaminants. Cell Tissue Res 2017; 367:627-642. [PMID: 28168324 DOI: 10.1007/s00441-017-2573-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/09/2017] [Indexed: 10/20/2022]
Abstract
Agricultural workers are exposed to many contaminants and suffer from respiratory and other symptoms. Dusts, gases, microbial products and pesticide residues from farms have been linked to effects on the health of agricultural workers. Growing sets of data from in vitro and in vivo models demonstrate the role of the innate immune system, especially Toll-like receptor 4 (TLR4) and TLR9, in lung inflammation induced following exposure to contaminants in agricultural environments. Interestingly, inflammation and lung function changes appear to be discordant indicating the complexity of inflammatory responses to exposures. Whereas the recent development of rodent models and exposure systems have yielded valuable data, we need new systems to examine the combined effects of multiple contaminants in order to increase our understanding of farm-exposure-induced negative health effects.
Collapse
|
47
|
Nedredal GI, Elvevold K, Chedid MF, Ytrebø LM, Rose CF, Sen S, Smedsrød B, Jalan R, Revhaug A. Pulmonary vascular clearance of harmful endogenous macromolecules in a porcine model of acute liver failure. Ann Hepatol 2017; 15:427-35. [PMID: 27049497 DOI: 10.5604/16652681.1198821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Pulmonary complications are common in acute liver failure (ALF). The role of the lungs in the uptake of harmful soluble endogenous macromolecules was evaluated in a porcine model of ALF induced by hepatic devascularization (n = 8) vs. controls (n = 8). In additional experiments, pulmonary uptake was investigated in healthy pigs. Fluorochrome-labeled modified albumin (MA) was applied to investigate the cellular uptake. RESULTS As compared to controls, the ALF group displayed a 4-fold net increased lung uptake of hyaluronan, and 5-fold net increased uptake of both tissue plasminogen activator and lysosomal enzymes. Anatomical distribution experiments in healthy animals revealed that radiolabeled MA uptake (taken up by the same receptor as hyaluronan) was 53% by the liver, and 24% by the lungs. The lung uptake of LPS was 14% whereas 60% remained in the blood. Both fluorescence and electron microscopy revealed initial uptake of MA by pulmonary endothelial cells (PECs) with later translocation to pulmonary intravascular macrophages (PIMs). Moreover, the presence of PIMs was evident 10 min after injection. Systemic inflammatory markers such as leukopenia and increased serum TNF-α levels were evident after 20 min in the MA and LPS groups. CONCLUSION Significant lung uptake of harmful soluble macromolecules compensated for the defect liver scavenger function in the ALF-group. Infusion of MA induced increased TNF-α serum levels and leukopenia, similar to the effect of the known inflammatory mediator LPS. These observations suggest a potential mechanism that may contribute to lung damage secondary to liver disease.
Collapse
Affiliation(s)
- Geir I Nedredal
- Department of Digestive Surgery, University Hospital Northern Norway
| | - Kjetil Elvevold
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø
| | - Marcio F Chedid
- Liver and Pancreas Transplant and Hepatobiliary Surgery Unit, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Lars M Ytrebø
- Department of Anesthesia and Intensive Care, University Hospital Northern Norway
| | | | - Sambit Sen
- Department of Gastroenterology, Luton & Dunstable University Hospital, Luton, UK
| | - Bård Smedsrød
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø
| | - Rajiv Jalan
- Institute of Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Arthur Revhaug
- Department of Digestive Surgery, University Hospital Northern Norway
| |
Collapse
|
48
|
Vrolyk V, Wobeser BK, Al-Dissi AN, Carr A, Singh B. Lung Inflammation Associated With Clinical Acute Necrotizing Pancreatitis in Dogs. Vet Pathol 2016; 54:129-140. [DOI: 10.1177/0300985816646432] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Although dogs with acute necrotizing pancreatitis (ANP) can develop respiratory complications, there are no data describing lung injury in clinical cases of ANP in dogs. Therefore, we conducted a study to characterize lung injury and determine if pulmonary intravascular macrophages (PIMs) are induced in dogs with ANP ( n = 21) compared with control dogs ( n = 6). Two pathologists independently graded histologic sections of pancreas from clinical cases to characterize the severity of ANP (total scores of 3–10) compared with controls showing histologically normal pancreas (total scores of 0). Based on histological grading, lungs from dogs with ANP showed inflammation (median score, 1.5; range, 0–3), but the scores did not differ statistically from the control lungs (median score, 0.5; range, 0–2). A grid intersects-counting method showed an increase in the numbers of MAC387-positive alveolar septal mononuclear phagocyte profiles in lungs of dogs with ANP (ratio median, 0.0243; range, 0.0093–0.0734, with 2 outliers at 0.1523 and 0.1978) compared with controls (ratio median, 0.0019; range, 0.0017–0.0031; P < .0001). Only dogs with ANP showed labeling for von Willebrand factor in alveolar septal capillary endothelial cells, septal inflammatory cells, and alveolar macrophages. Toll-like receptor 4 and interleukin 6 were variably expressed in alveolar macrophages and septal inflammatory cells in lungs from both ANP and control dogs. Inducible nitric oxide synthase was detected in alveolar macrophages of dogs with ANP only. These data show that dogs with ANP have lung inflammation, including the recruitment of PIMs and expression of inflammatory mediators.
Collapse
Affiliation(s)
- V. Vrolyk
- Departments of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - B. K. Wobeser
- Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - A. N. Al-Dissi
- Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - A. Carr
- Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - B. Singh
- Departments of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
49
|
Porta C, Riboldi E, Ippolito A, Sica A. Molecular and epigenetic basis of macrophage polarized activation. Semin Immunol 2016; 27:237-48. [PMID: 26561250 DOI: 10.1016/j.smim.2015.10.003] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/16/2015] [Accepted: 10/19/2015] [Indexed: 12/15/2022]
Abstract
Macrophages are unique cells for origin, heterogeneity and plasticity. At steady state most of macrophages are derived from fetal sources and maintained in adulthood through self-renewing. Despite sharing common progenitors, a remarkable heterogeneity characterized tissue-resident macrophages indicating that local signals educate them to express organ-specific functions. Macrophages are extremely plastic: chromatin landscape and transcriptional programs can be dynamically re-shaped in response to microenvironmental changes. Owing to their ductility, macrophages are crucial orchestrators of both initiation and resolution of immune responses and key supporters of tissue development and functions in homeostatic and pathological conditions. Herein, we describe current understanding of heterogeneity and plasticity of macrophages using the M1-M2 dichotomy as operationally useful simplification of polarized activation. We focused on the complex network of signaling cascades, metabolic pathways, transcription factors, and epigenetic changes that control macrophage activation. In particular, this network was addressed in sepsis, as a paradigm of a pathological condition determining dynamic macrophage reprogramming.
Collapse
Affiliation(s)
- Chiara Porta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy.
| | - Elena Riboldi
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy.
| | - Alessandro Ippolito
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy.
| | - Antonio Sica
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan 20089, Italy.
| |
Collapse
|
50
|
Moghimi SM. Complement Propriety and Conspiracy in Nanomedicine: Perspective and a Hypothesis. Nucleic Acid Ther 2016; 26:67-72. [DOI: 10.1089/nat.2015.0587] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Seyed Moein Moghimi
- Centre for Pharmaceutical Nanotechnology and Nanotoxicology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|