1
|
Odunitan TT, Apanisile BT, Afolabi JA, Adeniwura PO, Akinboade MW, Ibrahim NO, Alare KP, Saibu OA, Adeosun OA, Opeyemi HS, Ayiti KS. Beyond Conventional Drug Design: Exploring the Broad-Spectrum Efficacy of Antimicrobial Peptides. Chem Biodivers 2025; 22:e202401349. [PMID: 39480053 DOI: 10.1002/cbdv.202401349] [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: 05/31/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/02/2024]
Abstract
In the fight against pathogenic infections, antimicrobial peptides (AMPs) constitute a novel and promising class of compounds that defies accepted drug development conventions like Lipinski's rule. AMPs are remarkably effective against a variety of pathogens, including viruses, bacteria, parasites, and fungi. Their effectiveness, despite differing from traditional drug-like properties defies accepted standards. This review investigates the complex world of AMPs with an emphasis on their structural and physicochemical properties, which include size, sequence, structure, charge, and half-life. These distinguishing characteristics set AMPs apart from conventional therapeutics that adhere to Lipinski's rules and greatly contribute to their selective targeting, reduction of resistance, multifunctionality, and broad-spectrum efficacy. In contrast to traditional drugs that follow Lipinski's guidelines, AMPs have special qualities that play a big role in their ability to target specific targets, lower resistance, and work across a wide range of conditions. Our work is unique because of this nuanced investigation, which offers a new viewpoint on the potential of AMPs in tackling the worldwide problem of antibiotic resistance. In the face of the escalating global challenge of antibiotic resistance, antimicrobial peptides (AMPs) are innovative antimicrobial agents with unique mechanisms of action that challenge traditional Lipinski's Rule. They can withstand various microbial threats through membrane disruption, intracellular targeting, and immunomodulation. AMP versatility sets them apart from other antibiotics and their potential to address microbial infections and antibiotic resistance is growing. To fully unlock their potential, traditional drug development approaches need to be reconsidered. AMPs have revolutionary potential, paving the way for innovative solutions to health issues and transforming the antimicrobial therapy landscape.
Collapse
Affiliation(s)
- Tope T Odunitan
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
- Genomics Unit, Helix Biogen Institute, Ogbomosho, Nigeria
| | - Boluwatife T Apanisile
- Department of Nutrition and Dietetics, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Justinah A Afolabi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Praise O Adeniwura
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Modinat W Akinboade
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
- Genomics Unit, Helix Biogen Institute, Ogbomosho, Nigeria
| | - Najahtulahi O Ibrahim
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Kehinde P Alare
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Oluwatosin A Saibu
- Department of Chemistry and Biochemistry, New Mexico State University, USA, Ibadan
| | - Oyindamola A Adeosun
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Hammed S Opeyemi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Kolawole S Ayiti
- Department of Biochemistry, College of Medicine, University of Ibadan, Nigeria
| |
Collapse
|
2
|
O ET. Amphibian-Derived Antimicrobial Peptides: Essential Components of Innate Immunity and Potential Leads for New Antibiotic Development. Protein Pept Lett 2025; 32:97-110. [PMID: 39791145 DOI: 10.2174/0109298665356946241218103145] [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: 09/26/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 01/12/2025]
Abstract
Like other vertebrates, amphibians possess innate and adaptive immune systems. At the center of the adaptive immune system is the Major Histocompatibility Complex. The important molecules of innate immunity are antimicrobial peptides (AMPs). These peptides are secreted by granular glands in the skin and protect the animal against microorganisms entering its body through the skin. AMPs offer an effective and rapid defense against pathogenic microorganisms and have cationic and amphiphilic structures. These peptides are small gene-encoded molecules of 8-50 amino acid residues synthesized by ribosomes. These small molecules typically exhibit activity against bacteria, viruses, fungi, and even cancer cells. It is known that today's amphibian AMPs originated from a common precursor gene 150 million years ago and that the origin of these peptides is preprodermaseptins. Today, antibiotic resistance has occurred due to the incorrect use of antibiotics. Traditional antibiotics are becoming increasingly inadequate. AMPs are considered promising candidates for the development of new-generation antibiotics. Therefore, new antibiotic discoveries are needed. AMPs are suitable molecules for new-generation antibiotics that are both fast and have different killing mechanisms. One of the biggest problems in the clinical applications of AMPs is their poor stability. AMPs generally have limited tropical applications because they are sensitive to protease degradation. Coating these peptides with nanomaterials to make them more stable can solve this problem.
Collapse
Affiliation(s)
- Ebru Tanriverdi O
- Department of Biology, Faculty of Science, Ege University, Izmir, Turkey
| |
Collapse
|
3
|
Javadi A, Nikhbakht MR, Ghasemian Yadegari J, Rustamzadeh A, Mohammadi M, Shirazinejad A, Azadbakht S, Abdi Z. In-vivo and in vitro assessments of the radioprotective potential natural and chemical compounds: a review. Int J Radiat Biol 2023; 99:155-165. [PMID: 35549605 DOI: 10.1080/09553002.2022.2078007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE The study of the radioactive role of natural and chemical substances on human and animal studies has been the subject of research by some researchers. Therefore, the review of some of the past and current studies conducted in this field, can provide helpful information to elucidate of the importance of radioprotective components in reducing radiation exposure side effects. METHODS The authors search for keywords including In vitro, In vivo, Radioprotective, Ionizing radiation, and Vitamin in ScienceDirect, Scopus, Pubmed, and Google Scholar databases to access previously published articles and search for more reference articles on the role of radioprotective materials from natural and chemical compounds. RESULTS Radiation exposure can produce reactive oxygen species (ROS) in the body, however most of which are eliminated by the body's natural mechanisms, but when the body's antioxidant systems do not have enough ability to neutralize free radicals, oxidative stress occurs, which causes damage to DNA and body tissues. Therefore, it is necessary use of alternative substances that reduce and inhibit free radicals. CONCLUSION In general, recommended that antioxidant component(s) can be protect tissue damages in humans or animals, due to the their ability to scavenge free radicals generated by ionizing radiation.
Collapse
Affiliation(s)
- Anis Javadi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammad Reza Nikhbakht
- Department of Physiology and Pharmacology, School of Medicine Medicinal Plants Research Center Yasuj, University of Medical Sciences, Yasuj, Iran
| | - Javad Ghasemian Yadegari
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Auob Rustamzadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Mohammadi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran.,Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.,Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Alireza Shirazinejad
- Department of Food Science and Technology, Sarvestan Branch, Islamic Azad University, Sarvestan, Iran
| | - Saleh Azadbakht
- Department of Internal Medicine, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Zahra Abdi
- Department of Medical Biotechnology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
4
|
Dijksteel GS, Ulrich MMW, Middelkoop E, Boekema BKHL. Review: Lessons Learned From Clinical Trials Using Antimicrobial Peptides (AMPs). Front Microbiol 2021; 12:616979. [PMID: 33692766 PMCID: PMC7937881 DOI: 10.3389/fmicb.2021.616979] [Citation(s) in RCA: 227] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial peptides (AMPs) or host defense peptides protect the host against various pathogens such as yeast, fungi, viruses and bacteria. AMPs also display immunomodulatory properties ranging from the modulation of inflammatory responses to the promotion of wound healing. More interestingly, AMPs cause cell disruption through non-specific interactions with the membrane surface of pathogens. This is most likely responsible for the low or limited emergence of bacterial resistance against many AMPs. Despite the increasing number of antibiotic-resistant bacteria and the potency of novel AMPs to combat such pathogens, only a few AMPs are in clinical use. Therefore, the current review describes (i) the potential of AMPs as alternatives to antibiotics, (ii) the challenges toward clinical implementation of AMPs and (iii) strategies to improve the success rate of AMPs in clinical trials, emphasizing the lessons we could learn from these trials.
Collapse
Affiliation(s)
- Gabrielle S Dijksteel
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Magda M W Ulrich
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther Middelkoop
- Association of Dutch Burn Centres, Beverwijk, Netherlands.,Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | |
Collapse
|
5
|
Mao A, Sun C, Katsube T, Wang B. A Minireview on Gastrointestinal Microbiota and Radiosusceptibility. Dose Response 2020; 18:1559325820963859. [PMID: 33239996 PMCID: PMC7672743 DOI: 10.1177/1559325820963859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
Gastrointestinal (GI) microbiota maintains a symbiotic relationship with the host and plays a key role in modulating many important biological processes and functions of the host, such as metabolism, inflammation, immune and stress response. It is becoming increasingly apparent that GI microbiota is susceptible to a wide range of environmental factors and insults, for examples, geographic location of birth, diet, use of antibiotics, and exposure to radiation. Alterations in GI microbiota link to various diseases, including radiation-induced disorders. In addition, GI microbiota composition could be used as a biomarker to estimate radiosusceptibility and radiation health risk in the host. In this minireview, we summarized the documented studies on radiation-induced alterations in GI microbiota and the relationship between GI microbiota and radiosusceptibility of the host, and mainly discussed the possible mechanisms underlying GI microbiota influencing the outcome of radiation response in humans and animal models. Furthermore, we proposed that GI microbiota manipulation may be used to reduce radiation injury and improve the health of the host.
Collapse
Affiliation(s)
- Aihong Mao
- Gansu Provincial Academic Institute for Medical Research, Lanzhou, People's Republic of China
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People's Republic of China
| | - Takanori Katsube
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
6
|
Extracellular vesicles derived from mesenchymal stromal cells mitigate intestinal toxicity in a mouse model of acute radiation syndrome. Stem Cell Res Ther 2020; 11:371. [PMID: 32854778 PMCID: PMC7457304 DOI: 10.1186/s13287-020-01887-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/04/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Human exposure to high doses of radiation resulting in acute radiation syndrome and death can rapidly escalate to a mass casualty catastrophe in the event of nuclear accidents or terrorism. The primary reason is that there is presently no effective treatment option, especially for radiation-induced gastrointestinal syndrome. This syndrome results from disruption of mucosal barrier integrity leading to severe dehydration, blood loss, and sepsis. In this study, we tested whether extracellular vesicles derived from mesenchymal stromal cells (MSC) could reduce radiation-related mucosal barrier damage and reduce radiation-induced animal mortality. Methods Human MSC-derived extracellular vesicles were intravenously administered to NUDE mice, 3, 24, and 48 h after lethal whole-body irradiation (10 Gy). Integrity of the small intestine epithelial barrier was assessed by morphologic analysis, immunostaining for tight junction protein (claudin-3), and in vivo permeability to 4 kDa FITC-labeled dextran. Renewal of the small intestinal epithelium was determined by quantifying epithelial cell apoptosis (TUNEL staining) and proliferation (Ki67 immunostaining). Statistical analyses were performed using one-way ANOVA followed by a Tukey test. Statistical analyses of mouse survival were performed using Kaplan-Meier and Cox methods. Results We demonstrated that MSC-derived extracellular vesicle treatment reduced by 85% the instantaneous mortality risk in mice subjected to 10 Gy whole-body irradiation and so increased their survival time. This effect could be attributed to the efficacy of MSC-derived extracellular vesicles in reducing mucosal barrier disruption. We showed that the MSC-derived extracellular vesicles improved the renewal of the small intestinal epithelium by stimulating proliferation and inhibiting apoptosis of the epithelial crypt cells. The MSC-derived extracellular vesicles also reduced radiation-induced mucosal permeability as evidenced by the preservation of claudin-3 immunostaining at the tight junctions of the epithelium. Conclusions MSC-derived extracellular vesicles promote epithelial repair and regeneration and preserve structural integrity of the intestinal epithelium in mice exposed to radiation-induced gastrointestinal toxicity. Our results suggest that the administration of MSC-derived extracellular vesicles could be an effective therapy for limiting acute radiation syndrome.
Collapse
|
7
|
Jones CB, Davis CM, Sfanos KS. The Potential Effects of Radiation on the Gut-Brain Axis. Radiat Res 2020; 193:209-222. [DOI: 10.1667/rr15493.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
| | - Catherine M. Davis
- Division of Behavioral Biology, Department of Psychiatry and Behavioral Sciences
| | | |
Collapse
|
8
|
Torisawa YS. Microfluidic Organs-on-Chips to Reconstitute Cellular Microenvironments. Bioanalysis 2019. [DOI: 10.1007/978-981-13-6229-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
9
|
Janec KJ, Yuan H, Norton JE, Kelner RH, Hirt CK, Betensky RA, Guinan EC. rBPI 21 (Opebacan) Promotes Rapid Trilineage Hematopoietic Recovery in a Murine Model of High-Dose Total Body Irradiation. Am J Hematol 2018; 93:10.1002/ajh.25136. [PMID: 29752735 PMCID: PMC6230507 DOI: 10.1002/ajh.25136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 11/11/2022]
Abstract
The complexity of providing adequate care after radiation exposure has drawn increasing attention. While most therapeutic development has focused on improving survival at lethal radiation doses, acute hematopoietic syndrome (AHS) occurs at substantially lower exposures. Thus, it is likely that a large proportion of such a radiation-exposed population will manifest AHS of variable degree and that the medical and socioeconomic costs of AHS will accrue. Here, we examined the potential of rBPI21 (opebacan), used without supportive care, to accelerate hematopoietic recovery after radiation where expected survival was substantial (42-75%) at 30 days). rBPI21 administration was associated with accelerated recovery of hematopoietic precursors and normal marrow cellularity, with increases in megakaryocyte numbers particularly marked. This translated into attaining normal trilineage peripheral blood counts 2-3 weeks earlier than controls. Elevations of hematopoietic growth factors observed in plasma and the marrow microenvironment suggest the mechanism is likely multifactorial and not confined to known endotoxin-neutralizing and cytokine down-modulating activities of rBPI21 . These observations deserve further exploration in radiation models and other settings where inadequate hematopoiesis is a prominent feature. These experiments also model the potential of therapeutics to limit the allocation of scarce resources after catastrophic exposures as an endpoint independent of lethality mitigation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kenneth J. Janec
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Huaiping Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA
| | - James E. Norton
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Rowan H. Kelner
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Christian K. Hirt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA
| | - Rebecca A. Betensky
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston MA
| | - Eva C. Guinan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston MA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston MA
- Department of Radiation Oncology, Harvard Medical School, Boston MA
| |
Collapse
|
10
|
Rossmann MP, Orkin SH, Chute JP. Hematopoietic Stem Cell Biology. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
11
|
Duan Y, Yao X, Zhu J, Li Y, Zhang J, Zhou X, Qiao Y, Yang M, Li X. Effects of yak-activated protein on hematopoiesis and related cytokines in radiation-induced injury in mice. Exp Ther Med 2017; 14:5297-5304. [PMID: 29285056 PMCID: PMC5740812 DOI: 10.3892/etm.2017.5256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 03/24/2017] [Indexed: 11/08/2022] Open
Abstract
The aim of the present study was to investigate the protective effects of yak-activated protein on hematopoiesis and cytokine function in radiation-induced injury in mice. A total of 180 Kunming mice were randomly divided into three groups (A, B and C). Of these, 60 were randomly divided into a normal control group, a radiation model group, a positive control group and 3 yak-activated protein groups (high, medium and low dose groups; 10, 5 and 2.5 mg/kg, respectively). The other 120 mice were used for the subsequent experiments on days 7 and 14 following radiation. Yak-activated protein was administered orally to mice in the treatment groups and an equal volume of saline was administered orally to mice in the normal control and radiation model groups for 14 days. The positive control group received amifostine (150 mg/kg) via intraperitoneal injection. With the exception of the control group, the groups of mice received a 5 Gy quantity of X-radiation evenly over their whole body once. Changes in the peripheral hemogram, thymus and spleen indices, DNA content in the bone marrow, interleukin (IL)-2 and IL-6 levels, and the expression levels of B cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax) following irradiation were assessed. The low dose of yak-activated protein significantly increased Spleen indices in mice 14 days after irradiation and the high and middle dose of yak-activated protein significantly increased Thymus indices in mice 14 days after irradiation (P<0.05) compared with the control group. In addition, hemogram results increased gradually in the low-yak-activated protein dose group and were significantly higher 7 days after irradiation compared with the radiation model group (P<0.05). The DNA content in the bone marrow was markedly increased in the yak-activated protein groups, and increased significantly in the low dose group at 7 days post-irradiation compared with the radiation model group (P<0.05). The IL-2 content was significantly increased in the yak-activated protein groups (P<0.05). Furthermore, Bcl-2 expression was increased and Bax expression was decreased (P<0.05). These results suggest that yak-activated protein exerts protective effects against radiation-induced injury in mice. The optimal effects of yak-activated protein were observed in the medium dose group 14 days after irradiation.
Collapse
Affiliation(s)
- Yabin Duan
- Department of Clinical Pharmacy, Qinghai University Affiliated Hospital, Xining, Qinghai 810001, P.R. China
| | - Xingchen Yao
- Department of Radiotherapy Oncology, Qinghai People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Junbo Zhu
- Department of Pharmacy, Medical College, Quinghai University, Xining, Qinghai 810001, P.R. China
| | - Yongping Li
- Department of Traditional Chinese Medicine, Medical College, Quinghai University, Xining, Qinghai 810001, P.R. China
| | - Juanling Zhang
- Department of Biology Resources, College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Xuejiao Zhou
- Department of Biology Resources, College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Yijie Qiao
- Department of Pharmacy, Medical College, Quinghai University, Xining, Qinghai 810001, P.R. China
| | - Meng Yang
- Department of Biology Resources, College of Eco-Environmental Engineering, Qinghai University, Xining, Qinghai 810016, P.R. China
| | - Xiangyang Li
- Medical College, Qinghai University, Xining, Qinghai 810016, P.R. China.,State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai 810016, P.R. China
| |
Collapse
|
12
|
Satyamitra M, Kumar VP, Biswas S, Cary L, Dickson L, Venkataraman S, Ghosh SP. Impact of Abbreviated Filgrastim Schedule on Survival and Hematopoietic Recovery after Irradiation in Four Mouse Strains with Different Radiosensitivity. Radiat Res 2017; 187:659-671. [PMID: 28362168 DOI: 10.1667/rr14555.1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Filgrastim (Neupogen®, granulocyte-colony stimulating factor) is among the few countermeasures recommended for management of patients in the event of lethal total-body irradiation. Despite the plethora of studies using filgrastim as a radiation countermeasure, relatively little is known about the optimal dose schedule of filgrastim to mitigate radiation lethality. We evaluated the efficacy of filgrastim in improving 30-day survival of CD2F1 mice irradiated with a lethal dose (LD70/30) in the AFRRI cobalt-60 facility. We tested different schedules of 1, 3, 5, 10 or 16 once-daily injections of filgrastim initiated one day after irradiation. Time optimization studies with filgrastim treatment were also performed, beginning 6-48 h postirradiation. Maximum survival was observed with 3 daily doses of 0.17 mg/kg filgrastim. Survival efficacy of the 3-day treatment was compared against the conventional 16-day filgrastim treatment after irradiation in four mouse strains with varying radiation sensitivities: C3H/HeN, C57BL/6, B6C3F1 and CD2F1. Blood indices, bone marrow histopathology and colony forming unit assays were also evaluated. Filgrastim significantly increased 30-day survival (P < 0.001) with a 3-day treatment compared to 16-day treatment. Filgrastim did not prevent cytopenia nadirs, but facilitated faster recovery of white blood cells, neutrophils, red blood cells, platelets, lymphocytes and hematocrits in all four strains. Accelerated hematopoietic recovery was also reflected in faster bone marrow reconstitution and significant increase in hematopoietic progenitors (P < 0.001) in all four mouse strains. These data indicate that prompt and abbreviated filgrastim treatment has potential benefit for triage in the event of a radiological incident for treating acute hematopoietic syndrome.
Collapse
Affiliation(s)
- Merriline Satyamitra
- a Radiation and Nuclear Countermeasure Program, DAIT, NIAID, NIH, Bethesda, Maryland 20889
| | - Vidya P Kumar
- b Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Shukla Biswas
- b Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Lynnette Cary
- b Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Leonora Dickson
- b Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Srinivasan Venkataraman
- b Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Sanchita P Ghosh
- b Armed Forces Radiobiology Research Institute (AFRRI), Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| |
Collapse
|
13
|
Battersby AJ, Khara J, Wright VJ, Levy O, Kampmann B. Antimicrobial Proteins and Peptides in Early Life: Ontogeny and Translational Opportunities. Front Immunol 2016; 7:309. [PMID: 27588020 PMCID: PMC4989132 DOI: 10.3389/fimmu.2016.00309] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/29/2016] [Indexed: 12/18/2022] Open
Abstract
While developing adaptive immune responses, young infants are especially vulnerable to serious infections, including sepsis, meningitis, and pneumonia. Antimicrobial proteins and peptides (APPs) are key effectors that function as broad-spectrum anti-infectives. This review seeks to summarize the clinically relevant functional qualities of APPs and the increasing clinical trial evidence for their use to combat serious infections in infancy. Levels of APPs are relatively low in early life, especially in infants born preterm or with low birth weight (LBW). There are several rationales for the potential clinical utility of APPs in the prevention and treatment of infections in infants: (a) APPs may be most helpful in those with reduced levels; (b) during sepsis microbial products signal via pattern recognition receptors causing potentially harmful inflammation that APPs may counteract; and (c) in the era of antibiotic resistance, development of new anti-infective strategies is essential. Evidence supports the potential clinical utility of exogenous APPs to reduce infection-related morbidity in infancy. Further studies should characterize the ontogeny of antimicrobial activity in mucosal and systemic compartments, and examine the efficacy of exogenous-APP formulations to inform translational development of APPs for infant groups.
Collapse
Affiliation(s)
- Anna J Battersby
- Academic Paediatrics, Imperial College London, London, UK; Medical Research Council (MRC) Unit, Vaccines and Immunity Theme, Fajara, Gambia
| | - Jasmeet Khara
- Academic Paediatrics, Imperial College London, London, UK; Department of Pharmacy, National University of Singapore, Singapore
| | | | - Ofer Levy
- Precision Vaccines Program, Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Beate Kampmann
- Academic Paediatrics, Imperial College London, London, UK; Medical Research Council (MRC) Unit, Vaccines and Immunity Theme, Fajara, Gambia
| |
Collapse
|
14
|
Torisawa YS, Mammoto T, Jiang E, Jiang A, Mammoto A, Watters AL, Bahinski A, Ingber DE. Modeling Hematopoiesis and Responses to Radiation Countermeasures in a Bone Marrow-on-a-Chip. Tissue Eng Part C Methods 2016; 22:509-15. [PMID: 26993746 DOI: 10.1089/ten.tec.2015.0507] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Studies on hematopoiesis currently rely on animal models because in vitro culture methods do not accurately recapitulate complex bone marrow physiology. We recently described a bone marrow-on-a-chip microfluidic device that enables the culture of living hematopoietic bone marrow and mimics radiation toxicity in vitro. In the present study, we used this microdevice to demonstrate continuous blood cell production in vitro and model bone marrow responses to potential radiation countermeasure drugs. The device maintained mouse hematopoietic stem and progenitor cells in normal proportions for at least 2 weeks in culture. Increases in the number of leukocytes and red blood cells into the microfluidic circulation also could be detected over time, and addition of erythropoietin induced a significant increase in erythrocyte production. Exposure of the bone marrow chip to gamma radiation resulted in reduction of leukocyte production, and treatment of the chips with two potential therapeutics, granulocyte-colony stimulating factor or bactericidal/permeability-increasing protein (BPI), induced significant increases in the number of hematopoietic stem cells and myeloid cells in the fluidic outflow. In contrast, BPI was not found to have any effect when analyzed using static marrow cultures, even though it has been previously shown to accelerate recovery from radiation-induced toxicity in vivo. These findings demonstrate the potential value of the bone marrow-on-a-chip for modeling blood cell production, monitoring responses to hematopoiesis-modulating drugs, and testing radiation countermeasures in vitro.
Collapse
Affiliation(s)
- Yu-Suke Torisawa
- 1 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts.,2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Tadanori Mammoto
- 2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Elisabeth Jiang
- 2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Amanda Jiang
- 2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Akiko Mammoto
- 2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts
| | - Alexander L Watters
- 1 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts
| | - Anthony Bahinski
- 1 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts
| | - Donald E Ingber
- 1 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts.,2 Vascular Biology Program, Departments of Pathology and Surgery, Children's Hospital Boston and Harvard Medical School , Boston, Massachusetts.,3 School of Engineering and Applied Science, Harvard University , Cambridge, Massachusetts
| |
Collapse
|
15
|
Guinan E, Avigan DE, Soiffer RJ, Bunin NJ, Brennan LL, Bergelson I, Brightman S, Ozonoff A, Scannon PJ, Levy O. Pilot experience with opebacan/rBPI 21 in myeloablative hematopoietic cell transplantation. F1000Res 2016; 4:1480. [PMID: 26835003 PMCID: PMC4722698 DOI: 10.12688/f1000research.7558.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/15/2015] [Indexed: 12/26/2022] Open
Abstract
Bacterial infection and inflammation contribute significantly to the morbidity and mortality of myeloablative allogeneic hematopoietic cell transplantation (HCT). Endotoxin, a component of the outer membrane of Gram-negative bacteria, is a potent inflammatory stimulus in humans. Bactericidal/permeability increasing protein (BPI), a constituent of human neutrophil granules, binds endotoxin thereby precluding endotoxin-induced inflammation and also has direct anti-infective properties against bacteria. As a consequence of myeloablative therapy used in preparation for hematopoietic cell infusion, patients experience gastrointestinal leak of bacteria and bacterial toxins into the systemic circulation and a period of inflammatory cytokine elevation associated with subsequent regimen-related toxicities. Patients frequently become endotoxemic and febrile as well as BPI-deficient due to sustained neutropenia. To examine whether enhancing endotoxin-neutralizing and anti-infective activity by exogenous administration of a recombinant N-terminal fragment of BPI (rBPI
21, generic name opebacan) might ameliorate regimen-related toxicities including infection, we recruited patients scheduled to undergo myeloablative HCT to participate in a proof-of-concept prospective phase I/II trial. After the HCT preparative regimen was completed, opebacan was initiated 18-36 hours prior to administration of allogeneic hematopoietic stem cells (defined as Day 0) and continued for 72 hours. The trial was to have included escalation of rBPI
21 dose and duration but was stopped prematurely due to lack of further drug availability. Therefore, to better understand the clinical course of opebacan-treated patients (n=6), we compared their outcomes with a comparable cohort meeting the same eligibility criteria and enrolled in a non-interventional myeloablative HCT observational study (n = 35). Opebacan-treated participants had earlier platelet engraftment (p=0.005), mirroring beneficial effects of rBPI
21 previously observed in irradiated mice, fewer documented infections (p=0.03) and appeared less likely to experience significant regimen-related toxicities (p=0.05). This small pilot experience supports the potential utility of rBPI
21 in ameliorating HCT-related morbidity and merits further exploration.
Collapse
Affiliation(s)
- Eva Guinan
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA; Boston Children's Hospital, Boston, USA
| | - David E Avigan
- Harvard Medical School, Boston, USA; Beth Israel Deaconess Medical Center, Boston, USA
| | - Robert J Soiffer
- Dana-Farber Cancer Institute, Boston, USA; Harvard Medical School, Boston, USA; Brigham and Women's Hospital, Boston, USA
| | - Nancy J Bunin
- Children's Hospital of Philadelphia, Philadelphia, USA
| | | | | | | | - Al Ozonoff
- Harvard Medical School, Boston, USA; Boston Children's Hospital, Boston, USA
| | | | - Ofer Levy
- Harvard Medical School, Boston, USA; Boston Children's Hospital, Boston, USA
| |
Collapse
|
16
|
Palmer CD, Romero-Tejeda M, Sirignano M, Sharma S, Allen TM, Altfeld M, Jost S. Naturally Occurring Subclinical Endotoxemia in Humans Alters Adaptive and Innate Immune Functions through Reduced MAPK and Increased STAT1 Phosphorylation. THE JOURNAL OF IMMUNOLOGY 2015; 196:668-77. [PMID: 26643479 DOI: 10.4049/jimmunol.1501888] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/02/2015] [Indexed: 11/19/2022]
Abstract
Multiple studies have shown correlates of immune activation with microbial translocation and plasma LPS during HIV infection. It is unclear whether this activation is due to LPS, residual viral replication, or both. Few studies have addressed the effects of persistent in vivo levels of LPS on specific immune functions in humans in the absence of chronic viral infection or pathological settings such as sepsis. We previously reported on a cohort of HIV-negative men with subclinical endotoxemia linked to alterations in CD4/CD8 T cell ratio and plasma cytokine levels. This HIV-negative cohort allowed us to assess cellular immune functions in the context of different subclinical plasma LPS levels ex vivo without confounding viral effects. By comparing two samples of differing plasma LPS levels from each individual, we now show that subclinical levels of plasma LPS in vivo significantly alter T cell proliferative capacity, monocyte cytokine release, and HLA-DR expression, and induce TLR cross-tolerance by decreased phosphorylation of MAPK pathway components. Using this human in vivo model of subclinical endotoxemia, we furthermore show that plasma LPS leads to constitutive activation of STAT1 through autocrine cytokine signaling, suggesting that subclinical endotoxemia in healthy individuals might lead to significant changes in immune function that have thus far not been appreciated.
Collapse
Affiliation(s)
| | | | | | | | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139; and
| | - Marcus Altfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139; and Department of Viral Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Stephanie Jost
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139; and
| |
Collapse
|
17
|
Garbati MR, Hays LE, Rathbun RK, Jillette N, Chin K, Al-Dhalimy M, Agarwal A, Newell AEH, Olson SB, Bagby GC. Cytokine overproduction and crosslinker hypersensitivity are unlinked in Fanconi anemia macrophages. J Leukoc Biol 2015; 99:455-65. [PMID: 26432900 DOI: 10.1189/jlb.3a0515-201r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 09/15/2015] [Indexed: 01/13/2023] Open
Abstract
The Fanconi anemia proteins participate in a canonical pathway that repairs cross-linking agent-induced DNA damage. Cells with inactivated Fanconi anemia genes are universally hypersensitive to such agents. Fanconi anemia-deficient hematopoietic stem cells are also hypersensitive to inflammatory cytokines, and, as importantly, Fanconi anemia macrophages overproduce such cytokines in response to TLR4 and TLR7/8 agonists. We questioned whether TLR-induced DNA damage is the primary cause of aberrantly regulated cytokine production in Fanconi anemia macrophages by quantifying TLR agonist-induced TNF-α production, DNA strand breaks, crosslinker-induced chromosomal breakage, and Fanconi anemia core complex function in Fanconi anemia complementation group C-deficient human and murine macrophages. Although both M1 and M2 polarized Fanconi anemia cells were predictably hypersensitive to mitomycin C, only M1 macrophages overproduced TNF-α in response to TLR-activating signals. DNA damaging agents alone did not induce TNF-α production in the absence of TLR agonists in wild-type or Fanconi anemia macrophages, and mitomycin C did not enhance TLR responses in either normal or Fanconi anemia cells. TLR4 and TLR7/8 activation induced cytokine overproduction in Fanconi anemia macrophages. Also, although TLR4 activation was associated with induced double strand breaks, TLR7/8 activation was not. That DNA strand breaks and chromosome breaks are neither necessary nor sufficient to account for the overproduction of inflammatory cytokines by Fanconi anemia cells suggests that noncanonical anti-inflammatory functions of Fanconi anemia complementation group C contribute to the aberrant macrophage phenotype and suggests that suppression of macrophage/TLR hyperreactivity might prevent cytokine-induced stem cell attrition in Fanconi anemia.
Collapse
Affiliation(s)
- Michael R Garbati
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Laura E Hays
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - R Keaney Rathbun
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Nathaniel Jillette
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Kathy Chin
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Muhsen Al-Dhalimy
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Anupriya Agarwal
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Amy E Hanlon Newell
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Susan B Olson
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Grover C Bagby
- *Northwest Veterans Affairs Cancer Research Center, Portland, Oregon, USA; Oregon Health & Science University, Portland, Oregon, USA; and Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
18
|
Boittin FX, Martigne P, Mayol JF, Denis J, Raffin F, Coulon D, Grenier N, Drouet M, Hérodin F. Experimental Quantification of Delayed Radiation-Induced Organ Damage in Highly Irradiated Rats With Bone Marrow Protection: Effect of Radiation Dose and Organ Sensitivity. HEALTH PHYSICS 2015; 109:134-144. [PMID: 26107434 DOI: 10.1097/hp.0000000000000300] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The evolution of organ damage following extensive high-dose irradiation remains largely unexplored and needs further investigation. Wistar rats [with or without partial bone marrow protection (∼20%)] were irradiated at lethal gamma-ray doses (12, 14, and 16 Gy) and received antibiotic support. While total-body-irradiated rats did not survive, bone marrow protection (achieved by protecting hind limbs behind a lead wall) combined with antibiotic support allowed survival of 12-Gy and 14-Gy irradiated rats for more than 3 mo, with a late phase of body weight loss and altered clinical status. Histological analysis of radiation-induced damages in visceral organs (liver, kidney, and ileum), performed 64 and 104 d after high-dose body irradiation, indicates that the extent and the evolution of damage depend on both the irradiation dose and organ. A dose-related aggravation of lesions was observed in the liver and kidney but not in the ileum. In contrast to the liver, alterations in the kidney and ileum aggravate with time, emphasizing the need to develop new efficient countermeasures to protect both the gastrointestinal tract and kidney from late-occurring radiation effects. Specifically, the complex evolution of organ damage presented in this paper offers the possibility to explore and then validate specific therapeutic windows using candidate drugs targeted to each injured visceral organ.
Collapse
Affiliation(s)
- François-Xavier Boittin
- *Institut de Recherche Biomédicale des Armées (IRBA), Department of Radiobiology, Brétigny-sur-Orge, France
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Singh VK, Romaine PL, Seed TM. Medical Countermeasures for Radiation Exposure and Related Injuries: Characterization of Medicines, FDA-Approval Status and Inclusion into the Strategic National Stockpile. HEALTH PHYSICS 2015; 108:607-630. [PMID: 25905522 PMCID: PMC4418776 DOI: 10.1097/hp.0000000000000279] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/05/2015] [Indexed: 05/28/2023]
Abstract
World events over the past decade have highlighted the threat of nuclear terrorism as well as an urgent need to develop radiation countermeasures for acute radiation exposures and subsequent bodily injuries. An increased probability of radiological or nuclear incidents due to detonation of nuclear weapons by terrorists, sabotage of nuclear facilities, dispersal and exposure to radioactive materials, and accidents provides the basis for such enhanced radiation exposure risks for civilian populations. Although the search for suitable radiation countermeasures for radiation-associated injuries was initiated more than half a century ago, no safe and effective radiation countermeasure for the most severe of these injuries, namely acute radiation syndrome (ARS), has been approved by the United States Food and Drug Administration (FDA). The dearth of FDA-approved radiation countermeasures has prompted intensified research for a new generation of radiation countermeasures. In this communication, the authors have listed and reviewed the status of radiation countermeasures that are currently available for use, or those that might be used for exceptional nuclear/radiological contingencies, plus a limited few medicines that show early promise but still remain experimental in nature and unauthorized for human use by the FDA.
Collapse
Affiliation(s)
- Vijay K. Singh
- *Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Bethesda, MD; †Department of Radiation Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD; ‡Tech Micro Services, Bethesda, MD
| | - Patricia L.P. Romaine
- *Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Bethesda, MD; †Department of Radiation Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD; ‡Tech Micro Services, Bethesda, MD
| | - Thomas M. Seed
- *Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Bethesda, MD; †Department of Radiation Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD; ‡Tech Micro Services, Bethesda, MD
| |
Collapse
|
20
|
Xiao X, Luo H, Vanek KN, LaRue AC, Schulte BA, Wang GY. Catalase inhibits ionizing radiation-induced apoptosis in hematopoietic stem and progenitor cells. Stem Cells Dev 2015; 24:1342-1351. [PMID: 25603016 PMCID: PMC4440990 DOI: 10.1089/scd.2014.0402] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 01/19/2015] [Indexed: 12/19/2022] Open
Abstract
Hematologic toxicity is a major cause of mortality in radiation emergency scenarios and a primary side effect concern in patients undergoing chemo-radiotherapy. Therefore, there is a critical need for the development of novel and more effective approaches to manage this side effect. Catalase is a potent antioxidant enzyme that coverts hydrogen peroxide into hydrogen and water. In this study, we evaluated the efficacy of catalase as a protectant against ionizing radiation (IR)-induced toxicity in hematopoietic stem and progenitor cells (HSPCs). The results revealed that catalase treatment markedly inhibits IR-induced apoptosis in murine hematopoietic stem cells and hematopoietic progenitor cells. Subsequent colony-forming cell and cobble-stone area-forming cell assays showed that catalase-treated HSPCs can not only survive irradiation-induced apoptosis but also have higher clonogenic capacity, compared with vehicle-treated cells. Moreover, transplantation of catalase-treated irradiated HSPCs results in high levels of multi-lineage and long-term engraftments, whereas vehicle-treated irradiated HSPCs exhibit very limited hematopoiesis reconstituting capacity. Mechanistically, catalase treatment attenuates IR-induced DNA double-strand breaks and inhibits reactive oxygen species. Unexpectedly, we found that the radioprotective effect of catalase is associated with activation of the signal transducer and activator of transcription 3 (STAT3) signaling pathway and pharmacological inhibition of STAT3 abolishes the protective activity of catalase, suggesting that catalase may protect HSPCs against IR-induced toxicity via promoting STAT3 activation. Collectively, these results demonstrate a previously unrecognized mechanism by which catalase inhibits IR-induced DNA damage and apoptosis in HSPCs.
Collapse
Affiliation(s)
- Xia Xiao
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
- Department of Hematology, Tianjin First Center Hospital, Tianjin, People's Republic of China
| | - Hongmei Luo
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
- Department of Histology and Embryology, University of South China, Hengyang City, Hunan Province, People's Republic of China
| | - Kenneth N. Vanek
- Department of Radiation Oncology, Medical University of South Carolina, Charleston, South Carolina
| | - Amanda C. LaRue
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
- Research Services, Ralph H. Johnson VAMC, Charleston, South Carolina
- Cancer Genes and Molecular Regulation Program of the Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Bradley A. Schulte
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Gavin Y. Wang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
- Cancer Genes and Molecular Regulation Program of the Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
21
|
Taniguchi CM, Miao YR, Diep AN, Wu C, Rankin EB, Atwood TF, Xing L, Giaccia AJ. PHD inhibition mitigates and protects against radiation-induced gastrointestinal toxicity via HIF2. Sci Transl Med 2014; 6:236ra64. [PMID: 24828078 DOI: 10.1126/scitranslmed.3008523] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Radiation-induced gastrointestinal (GI) toxicity can be a major source of morbidity and mortality after radiation exposure. There is an unmet need for effective preventative or mitigative treatments against the potentially fatal diarrhea and water loss induced by radiation damage to the GI tract. We report that prolyl hydroxylase inhibition by genetic knockout or pharmacologic inhibition of all PHD (prolyl hydroxylase domain) isoforms by the small-molecule dimethyloxallyl glycine (DMOG) increases hypoxia-inducible factor (HIF) expression, improves epithelial integrity, reduces apoptosis, and increases intestinal angiogenesis, all of which are essential for radioprotection. HIF2, but not HIF1, is both necessary and sufficient to prevent radiation-induced GI toxicity and death. Increased vascular endothelial growth factor (VEGF) expression contributes to the protective effects of HIF2, because inhibition of VEGF function reversed the radioprotection and radiomitigation afforded by DMOG. Additionally, mortality from abdominal or total body irradiation was reduced even when DMOG was given 24 hours after exposure. Thus, prolyl hydroxylase inhibition represents a treatment strategy to protect against and mitigate GI toxicity from both therapeutic radiation and potentially lethal radiation exposures.
Collapse
Affiliation(s)
- Cullen M Taniguchi
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Yu Rebecca Miao
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Anh N Diep
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Colleen Wu
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Todd F Atwood
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Lei Xing
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Pettengill MA, van Haren SD, Levy O. Soluble mediators regulating immunity in early life. Front Immunol 2014; 5:457. [PMID: 25309541 PMCID: PMC4173950 DOI: 10.3389/fimmu.2014.00457] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 09/08/2014] [Indexed: 12/15/2022] Open
Abstract
Soluble factors in blood plasma have a substantial impact on both the innate and adaptive immune responses. The complement system, antibodies, and anti-microbial proteins and peptides can directly interact with potential pathogens, protecting against systemic infection. Levels of these innate effector proteins are generally lower in neonatal circulation at term delivery than in adults, and lower still at preterm delivery. The extracellular environment also has a critical influence on immune cell maturation, activation, and effector functions, and many of the factors in plasma, including hormones, vitamins, and purines, have been shown to influence these processes for leukocytes of both the innate and adaptive immune systems. The ontogeny of plasma factors can be viewed in the context of a lower effectiveness of immune responses to infection and immunization in early life, which may be influenced by the striking neonatal deficiency of complement system proteins or enhanced neonatal production of the anti-inflammatory cytokine IL-10, among other ontogenic differences. Accordingly, we survey here a number of soluble mediators in plasma for which age-dependent differences in abundance may influence the ontogeny of immune function, particularly direct innate interaction and skewing of adaptive lymphocyte activity in response to infectious microorganisms and adjuvanted vaccines.
Collapse
Affiliation(s)
- Matthew Aaron Pettengill
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital , Boston, MA , USA ; Harvard Medical School , Boston, MA , USA
| | - Simon Daniël van Haren
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital , Boston, MA , USA ; Harvard Medical School , Boston, MA , USA
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital , Boston, MA , USA ; Harvard Medical School , Boston, MA , USA
| |
Collapse
|
23
|
Himburg HA, Yan X, Doan PL, Quarmyne M, Micewicz E, McBride W, Chao NJ, Slamon DJ, Chute JP. Pleiotrophin mediates hematopoietic regeneration via activation of RAS. J Clin Invest 2014; 124:4753-8. [PMID: 25250571 DOI: 10.1172/jci76838] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 08/21/2014] [Indexed: 02/06/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are highly susceptible to ionizing radiation-mediated death via induction of ROS, DNA double-strand breaks, and apoptotic pathways. The development of therapeutics capable of mitigating ionizing radiation-induced hematopoietic toxicity could benefit both victims of acute radiation sickness and patients undergoing hematopoietic cell transplantation. Unfortunately, therapies capable of accelerating hematopoietic reconstitution following lethal radiation exposure have remained elusive. Here, we found that systemic administration of pleiotrophin (PTN), a protein that is secreted by BM-derived endothelial cells, substantially increased the survival of mice following radiation exposure and after myeloablative BM transplantation. In both models, PTN increased survival by accelerating the recovery of BM hematopoietic stem and progenitor cells in vivo. PTN treatment promoted HSC regeneration via activation of the RAS pathway in mice that expressed protein tyrosine phosphatase receptor-zeta (PTPRZ), whereas PTN treatment did not induce RAS signaling in PTPRZ-deficient mice, suggesting that PTN-mediated activation of RAS was dependent upon signaling through PTPRZ. PTN strongly inhibited HSC cycling following irradiation, whereas RAS inhibition abrogated PTN-mediated induction of HSC quiescence, blocked PTN-mediated recovery of hematopoietic stem and progenitor cells, and abolished PTN-mediated survival of irradiated mice. These studies demonstrate the therapeutic potential of PTN to improve survival after myeloablation and suggest that PTN-mediated hematopoietic regeneration occurs in a RAS-dependent manner.
Collapse
|
24
|
Strunk T, Inder T, Wang X, Burgner D, Mallard C, Levy O. Infection-induced inflammation and cerebral injury in preterm infants. THE LANCET. INFECTIOUS DISEASES 2014; 14:751-762. [PMID: 24877996 DOI: 10.1016/s1473-3099(14)70710-8] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Preterm birth and infectious diseases are the most common causes of neonatal and early childhood deaths worldwide. The rates of preterm birth have increased over recent decades and account for 11% of all births worldwide. Preterm infants are at significant risk of severe infection in early life and throughout childhood. Bacteraemia, inflammation, or both during the neonatal period in preterm infants is associated with adverse outcomes, including death, chronic lung disease, and neurodevelopmental impairment. Recent studies suggest that bacteraemia could trigger cerebral injury even without penetration of viable bacteria into the CNS. Here we review available evidence that supports the concept of a strong association between bacteraemia, inflammation, and cerebral injury in preterm infants, with an emphasis on the underlying biological mechanisms, clinical correlates, and translational opportunities.
Collapse
Affiliation(s)
- Tobias Strunk
- Centre for Neonatal Research and Education, School of Paediatrics and Child Health, The University of Western Australia, Perth, WA, Australia; Neonatal Clinical Care Unit, King Edward Memorial Hospital, Perth, WA, Australia.
| | - Terrie Inder
- Department of Pediatrics, Neurology and Radiology, Washington University, St Louis, USA
| | - Xiaoyang Wang
- Perinatal Center, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Shangjie, Henan, China
| | - David Burgner
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Carina Mallard
- Perinatal Center, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ofer Levy
- Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Hong GU, Kim NG, Ro JY. Expression of airway remodeling proteins in mast cell activated by TGF-β released in OVA-induced allergic responses and their inhibition by low-dose irradiation or 8-oxo-dG. Radiat Res 2014; 181:425-38. [PMID: 24720751 DOI: 10.1667/rr13547.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Allergic asthma is characterized by chronic airway remodeling, which is associated with the expression of extracellular matrix proteins (ECM) by TGF-β. However, to date there are no reports demonstrating that structural proteins are directly expressed in mast cells. This study aimed to investigate whether ECM proteins are expressed in mast cells activated with antigen/antibody reaction, and whether the resolution effects of irradiation or 8-oxo-dG may contribute to allergic asthma prevention. Bone marrow-derived mast cells (BMMCs) were activated with DNP-HSA/anti-DNP IgE antibody (act-BMMCs). C57BL/6 mice were sensitized and challenged with ovalbumin (OVA) to induce allergic asthma. Mice were treated orally with 8-oxo-dG or exposed to whole body irradiation (using (137)Cs gamma ray at a dose of 0.5 Gy) for three consecutive days 24 h after OVA challenge. Expression of extracellular matrix (ECM) proteins, TGF-β signaling molecules and NF-κB/AP-1 was determined in the BMMCs, bronchoalveolar lavage (BAL) cells or lung tissues using Western blot, polymerase chain reaction (PCR) and electrophoretic mobility shift assay (EMSA), respectively. Act-BMMCs increased expression of ECM proteins, TGF-β/TGF-β receptor I, TGF-β signaling molecules and cytokines; and increased both NF-κB and AP-1 activity. In addition, the population of mast cells; expression of mast cell markers, TGF-β signaling molecules, ECM proteins/amounts; OVA-specific serum IgE level; numbers of goblet cells; airway hyperresponsiveness; cytokines/chemokines were increased in BAL cells and lung tissues of OVA-challenged mice. All of the above end points were reduced by irradiation or 8-oxo-dG in vitro and in vivo, respectively. The data suggest that mast cells induce expression of ECM proteins through TGF-β produced in inflammatory cells of OVA mice and that post treatment of irradiation or 8-oxo-dG after OVA-challenge may reduce airway remodeling through down-regulating mast cell re-activation by TGF-β/Smad signals.
Collapse
Affiliation(s)
- Gwan Ui Hong
- Department of Pharmacology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, South Korea
| | | | | |
Collapse
|
26
|
Abstract
Fractionated, high-dose total body irradiation (TBI) is used therapeutically to myeloablate and immune suppress patients undergoing hematopoietic stem cell (HSC) transplantation. Acute exposure to ionizing radiation can have fatal effects on the hematopoietic and immune systems. Currently, therapies aimed at ameliorating ionizing radiation-associated toxicities are limited. In the February 2014 issue of the JCI, Kim and colleagues demonstrated that induction of nuclear factor erythroid 2-related factor 2 (NRF2) enhances HSC regeneration and increases survival following ionizing radiation exposure in mice. The results of this study suggest that NRF2 is a novel potential target for the development of therapeutics aimed at mitigating the toxicities of ionizing radiation exposure.
Collapse
|
27
|
Kim JH, Thimmulappa RK, Kumar V, Cui W, Kumar S, Kombairaju P, Zhang H, Margolick J, Matsui W, Macvittie T, Malhotra SV, Biswal S. NRF2-mediated Notch pathway activation enhances hematopoietic reconstitution following myelosuppressive radiation. J Clin Invest 2014; 124:730-41. [PMID: 24463449 PMCID: PMC3904618 DOI: 10.1172/jci70812] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 10/31/2013] [Indexed: 12/13/2022] Open
Abstract
A nuclear disaster may result in exposure to potentially lethal doses of ionizing radiation (IR). Hematopoietic acute radiation syndrome (H-ARS) is characterized by severe myelosuppression, which increases the risk of infection, bleeding, and mortality. Here, we determined that activation of nuclear factor erythroid-2-related factor 2 (NRF2) signaling enhances hematopoietic stem progenitor cell (HSPC) function and mitigates IR-induced myelosuppression and mortality. Augmenting NRF2 signaling in mice, either by genetic deletion of the NRF2 inhibitor Keap1 or by pharmacological NRF2 activation with 2-trifluoromethyl-2'-methoxychalone (TMC), enhanced hematopoietic reconstitution following bone marrow transplantation (BMT). Strikingly, even 24 hours after lethal IR exposure, oral administration of TMC mitigated myelosuppression and mortality in mice. Furthermore, TMC administration to irradiated transgenic Notch reporter mice revealed activation of Notch signaling in HSPCs and enhanced HSPC expansion by increasing Jagged1 expression in BM stromal cells. Administration of a Notch inhibitor ablated the effects of TMC on hematopoietic reconstitution. Taken together, we identified a mechanism by which NRF2-mediated Notch signaling improves HSPC function and myelosuppression following IR exposure. Our data indicate that targeting this pathway may provide a countermeasure against the damaging effects of IR exposure.
Collapse
Affiliation(s)
- Jung-Hyun Kim
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Rajesh K. Thimmulappa
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Vineet Kumar
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Wanchang Cui
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Sarvesh Kumar
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Ponvijay Kombairaju
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Hao Zhang
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Joseph Margolick
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - William Matsui
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Thomas Macvittie
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Sanjay V. Malhotra
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| | - Shyam Biswal
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.
Laboratory of Synthetic Chemistry, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.
Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland, USA.
Department of Microbiology and Molecular Immunology, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland, USA.
Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Guinan EC, Palmer CD, Mancuso CJ, Brennan L, Stoler-Barak L, Kalish LA, Suter EE, Gallington LC, Huhtelin DP, Mansilla M, Schumann RR, Murray JC, Weiss J, Levy O. Identification of single nucleotide polymorphisms in hematopoietic cell transplant patients affecting early recognition of, and response to, endotoxin. Innate Immun 2013; 20:697-711. [PMID: 24107515 DOI: 10.1177/1753425913505122] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Hematopoietic cell transplant (HCT) is a life-saving therapy for many malignant and non-malignant bone marrow diseases. Associated morbidities are often due to transplant-related toxicities and infections, exacerbated by regimen-induced immune suppression and systemic incursion of bacterial products. Patients undergoing myeloablative conditioning for HCT become endotoxemic and display blood/plasma changes consistent with lipopolysaccharide (LPS)-induced systemic innate immune activation. Herein, we addressed whether patients scheduled for HCT display differences in recognition/response to LPS ex vivo traceable to specific single nucleotide polymorphisms (SNPs). Two SNPs of LPS binding protein (LBP) were associated with changes in plasma LBP levels, with one LBP SNP also associating with differences in efficiency of extraction and transfer of endotoxin to myeloid differentiation factor-2 (MD-2), a step needed for activation of TLR4. None of the examined SNPs of CD14, bactericidal/permeability-increasing protein (BPI), TLR4 or MD-2 were associated with corresponding protein plasma levels or endotoxin delivery to MD-2, but CD14 and BPI SNPs significantly associated with differences in LPS-induced TNF-α release ex vivo and infection frequency, respectively. These findings suggest that specific LBP, CD14 and BPI SNPs might be contributory assessments in studies where clinical outcome may be affected by host response to endotoxin and bacterial infection.
Collapse
Affiliation(s)
- Eva C Guinan
- Boston Children's Hospital, Boston, MA, USA Harvard Medical School, Boston, MA, USA Dana-Farber Cancer Institute, Boston, MA, USA
| | - Christine D Palmer
- Boston Children's Hospital, Boston, MA, USA Harvard Medical School, Boston, MA, USA Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Boston, MA, USA
| | | | | | | | - Leslie A Kalish
- Boston Children's Hospital, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| | | | | | - David P Huhtelin
- University of Iowa and Veterans' Administration Medical Center, Coralville, Iowa City, IA, USA
| | - Maria Mansilla
- Department of Pediatrics, University of Iowa, IA, Iowa City, USA
| | - Ralf R Schumann
- Institute for Microbiology, Charité-University Medical Center, Berlin, Germany
| | - Jeffrey C Murray
- Department of Pediatrics, University of Iowa, IA, Iowa City, USA
| | - Jerrold Weiss
- University of Iowa and Veterans' Administration Medical Center, Coralville, Iowa City, IA, USA
| | - Ofer Levy
- Boston Children's Hospital, Boston, MA, USA Harvard Medical School, Boston, MA, USA
| |
Collapse
|
29
|
Neonatal host defense against Staphylococcal infections. Clin Dev Immunol 2013; 2013:826303. [PMID: 23935651 PMCID: PMC3722842 DOI: 10.1155/2013/826303] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/14/2013] [Accepted: 05/14/2013] [Indexed: 12/17/2022]
Abstract
Preterm infants are especially susceptible to late-onset sepsis that is often due to Gram-positive bacterial infections resulting in substantial morbidity and mortality. Herein, we will describe neonatal innate immunity to Staphylococcus spp. comparing differences between preterm and full-term newborns with adults. Newborn innate immunity is distinct demonstrating diminished skin integrity, impaired Th1-polarizing responses, low complement levels, and diminished expression of plasma antimicrobial proteins and peptides, especially in preterm newborns. Characterization of distinct aspects of the neonatal immune response is defining novel approaches to enhance host defense to prevent and/or treat staphylococcal infection in this vulnerable population.
Collapse
|
30
|
Li K, Liu Y, Xia X, Wang L, Lu M, Hu Y, Xu C. Bactericidal/permeability-increasing protein in the reproductive system of male mice may be involved in the sperm-oocyte fusion. Reproduction 2013; 146:135-44. [PMID: 23740083 DOI: 10.1530/rep-13-0127] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Bactericidal/permeability-increasing protein (BPI) is a 455-residue (∼55 kDa) protein found mainly in the primary (azurophilic) granules of human neutrophils. BPI is an endogenous antibiotic protein that belongs to the family of mammalian lipopolysaccharide (LPS)-binding and lipid transport proteins. Its major function is to kill Gram-negative bacteria, thereby protecting the host from infection. In addition, BPI can inhibit angiogenesis, suppress LPS-mediated platelet activation, increase DNA synthesis, and activate ERK/Akt signaling. In this study, we found that Bpi was expressed in the testis and epididymis but not in the seminal vesicles, prostate, and solidification glands. BPI expression in the epididymis increased upon upregulation of testosterone, caused by injection of GNRH. In orchidectomized mice, BPI expression was significantly reduced, but its expression was restored to 30% of control levels in orchidectomized mice that received supplementary testosterone. The number of sperm fused per egg significantly decreased after incubation with anti-BPI antiserum. These results suggest that BPI may take part in the process of sperm-oocyte fusion and play a unique and significant role in reproduction.
Collapse
Affiliation(s)
- Kun Li
- Department of Histology and Embryology, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | | | | | | | | | | | | |
Collapse
|
31
|
Gaberman E, Pinzur L, Levdansky L, Tsirlin M, Netzer N, Aberman Z, Gorodetsky R. Mitigation of Lethal Radiation Syndrome in Mice by Intramuscular Injection of 3D Cultured Adherent Human Placental Stromal Cells. PLoS One 2013; 8:e66549. [PMID: 23823334 PMCID: PMC3688917 DOI: 10.1371/journal.pone.0066549] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Accepted: 05/12/2013] [Indexed: 12/22/2022] Open
Abstract
Exposure to high lethal dose of ionizing radiation results in acute radiation syndrome with deleterious systemic effects to different organs. A primary target is the highly sensitive bone marrow and the hematopoietic system. In the current study C3H/HeN mice were total body irradiated by 7.7 Gy. Twenty four hrs and 5 days after irradiation 2×106 cells from different preparations of human derived 3D expanded adherent placental stromal cells (PLX) were injected intramuscularly. Treatment with batches consisting of pure maternal cell preparations (PLX-Mat) increased the survival of the irradiated mice from ∼27% to 68% (P<0.001), while cell preparations with a mixture of maternal and fetal derived cells (PLX-RAD) increased the survival to ∼98% (P<0.0001). The dose modifying factor of this treatment for both 50% and 37% survival (DMF50 and DMF37) was∼1.23. Initiation of the more effective treatment with PLX-RAD injection could be delayed for up to 48 hrs after irradiation with similar effect. A delayed treatment by 72 hrs had lower, but still significantly effect (p<0.05). A faster recovery of the BM and improved reconstitution of all blood cell lineages in the PLX-RAD treated mice during the follow-up explains the increased survival of the cells treated irradiated mice. The number of CD45+/SCA1+ hematopoietic progenitor cells within the fast recovering population of nucleated BM cells in the irradiated mice was also elevated in the PLX-RAD treated mice. Our study suggests that IM treatment with PLX-RAD cells may serve as a highly effective “off the shelf” therapy to treat BM failure following total body exposure to high doses of radiation. The results suggest that similar treatments may be beneficial also for clinical conditions associated with severe BM aplasia and pancytopenia.
Collapse
Affiliation(s)
- Elena Gaberman
- Sharett Institute of Oncology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | | | - Lilia Levdansky
- Sharett Institute of Oncology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Maria Tsirlin
- Sharett Institute of Oncology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nir Netzer
- Pluristem Therapeutics Inc., Haifa, Israel
| | | | - Raphael Gorodetsky
- Sharett Institute of Oncology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
32
|
Abstract
Newborns are at increased risk of infection due to genetic, epigenetic, and environmental factors. Herein we examine the roles of the neonatal innate immune system in host defense against bacterial and viral infections. Full-term newborns express a distinct innate immune system biased toward T(H)2-/T(H)17-polarizing and anti-inflammatory cytokine production with relative impairment in T(H)1-polarizing cytokine production that leaves them particularly vulnerable to infection with intracellular pathogens. In addition to these distinct features, preterm newborns also have fragile skin, impaired T(H)17-polarizing cytokine production, and deficient expression of complement and of antimicrobial proteins and peptides (APPs) that likely contribute to susceptibility to pyogenic bacteria. Ongoing research is identifying APPs, including bacterial/permeability-increasing protein and lactoferrin, as well as pattern recognition receptor agonists that may serve to enhance protective newborn and infant immune responses as stand-alone immune response modifiers or vaccine adjuvants.
Collapse
Affiliation(s)
| | - James L Wynn
- Division of Neonatology, Department of Pediatrics, Vanderbilt University
| | | | - Ofer Levy
- Division of Infectious Diseases, Boston Children’s Hospital; Boston MA,Harvard Medical School, Boston, MA
| |
Collapse
|
33
|
Nathe KE, Mancuso CJ, Parad R, Van Marter LJ, Martin CR, Stoler-Barak L, Philbin VJ, Phillips MF, Palmer CD, Levy O. Innate immune activation in neonatal tracheal aspirates suggests endotoxin-driven inflammation. Pediatr Res 2012; 72:203-11. [PMID: 22580716 PMCID: PMC3406551 DOI: 10.1038/pr.2012.61] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Tracheal aspirates (TAs) from critically ill neonates accumulate bacterial endotoxin and demonstrate mobilization of endotoxin-binding proteins, but the potential bioactivity of endotoxin in TAs is unknown. We characterized innate immune activation in TAs of mechanically ventilated neonates. METHODS Innate immune activation in TAs of mechanically ventilated neonates was characterized using a targeted 84-gene quantitative real-time (qRT) PCR array. Protein expression of cytokines was confirmed by multiplex assay. Expression and localization of the endotoxin-inducible antimicrobial protein Calgranulin C (S100A12) was assessed by flow cytometry. Endotoxin levels were measured in TA supernatants using the Limulus amoebocyte lysate assay. RESULTS Analyses by qRT-PCR demonstrated expression of pattern recognition receptors, Toll-like receptor-nuclear factor κB and inflammasome pathways, cytokines/chemokines and their receptors, and anti-infective proteins in TA cells. Endotoxin positivity increased with postnatal age. As compared with endotoxin-negative TAs, endotoxin-positive TAs demonstrated significantly greater tumor necrosis factor (TNF), interleukin (IL)-6, IL-10, and serpin peptidase inhibitor, clade E, member 1 (SERPINE1) mRNA, and IL-10, TNF, and IL-1β protein. Expression of S100A12 protein was localized to TA neutrophils. CONCLUSION Correlation of endotoxin with TA inflammatory responses suggests endotoxin bioactivity and the possibility that endotoxin antagonists could mitigate pulmonary inflammation and its sequelae in this vulnerable population.
Collapse
Affiliation(s)
- Katheryn E. Nathe
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts,Department of Newborn Medicine, Brigham and Women's Hospital, Boston, Massachusetts,Department of Pediatrics, Massachusetts General Hospital for Children at North Shore Medical Center, Salem, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - Christy J. Mancuso
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts
| | - Richard Parad
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts,Department of Newborn Medicine, Brigham and Women's Hospital, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - Linda J. Van Marter
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts,Department of Newborn Medicine, Brigham and Women's Hospital, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - Camilia R. Martin
- Harvard Medical School, Boston, Massachusetts,Department of Neonatology Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Liat Stoler-Barak
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts
| | - Victoria J. Philbin
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - Michele F. Phillips
- Department of Newborn Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Christine D. Palmer
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts
| | - Ofer Levy
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts,Harvard Medical School, Boston, Massachusetts,()
| |
Collapse
|
34
|
Rodgers KE, Espinoza T, Roda N, Meeks CJ, Hill C, Louie SG, Dizerega GS. Accelerated hematopoietic recovery with angiotensin-(1-7) after total body radiation. Int J Radiat Biol 2012; 88:466-76. [PMID: 22433112 DOI: 10.3109/09553002.2012.676228] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Angiotensin (1-7) [A(1-7)] is a component of the renin angiotensin system (RAS) that stimulates hematopoietic recovery after myelosuppression. In a Phase I/IIa clinical trial, thrombocytopenia after chemotherapy was reduced by A(1-7). In this study, the ability of A(1-7) to improve recovery after total body irradiation (TBI) is shown with specific attention to radiation-induced hematopoietic injury. MATERIALS AND METHODS Mice were exposed to TBI (doses of 2-7 Gray [Gy]) of cesium 137 gamma rays, followed by treatment with A(1-7), typical doses were 100-1000 μg/kg given once or once daily for a specified number of days depending on the study. Animals are injected subcutaneously via the nape of the neck with 0.1 ml drug in saline. The recovery of blood and bone marrow cells was determined. Effects of TBI and A(1-7) on survival and bleeding time was also evaluated. RESULTS Daily administration of A(1-7) after radiation exposure improved survival (from 60% to 92-97%) and reduced bleeding time at day 30 after TBI. Further, A(1-7) increased early mixed progenitors (3- to 5-fold), megakaryocyte (2- to 3-fold), myeloid (3- to 6-fold) and erythroid (2- to 5-fold) progenitors in the bone marrow and reduced radiation-induced thrombocytopenia (RIT) (up to 2-fold). Reduction in the number of treatments to 3 per week also improved bone marrow recovery and reduced RIT. As emergency responder and healthcare systems in case of nuclear accident or/and terrorist attack may be overwhelmed, the consequence of delayed initiation of treatment was ascertained. Treatment with A(1-7) can be delayed up to 5 days and still be effective in the reduction of RIT or acceleration of bone marrow recovery. CONCLUSIONS The data presented in this paper indicate that A(1-7) reduces the consequences of critical radiation exposure and can be initiated well after initial exposure with maximal effects on early responding hematopoietic progenitors when treatment is initiated 2 days after exposure and 5 days after exposure for the later responding progenitors and reduced thrombocytopenia. There was some effect of A(1-7) even when given days after radiation exposure.
Collapse
Affiliation(s)
- Kathleen E Rodgers
- School of Pharmacy, University of Southern California, Los Angeles, California, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Tolar J, Tolar M. Reinventing mesenchymal stromal cells. Cytotherapy 2012; 14:388-90. [PMID: 22420833 DOI: 10.3109/14653249.2012.665631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Jakub Tolar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA.
| | | |
Collapse
|
36
|
Double radiation shield. Nature 2011. [DOI: 10.1038/480008b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
37
|
Dynamic duo helps to heal irradiated mice. Nature 2011. [DOI: 10.1038/nature.2011.9463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|