1
|
Wang K, Yuan B, Zhang F, Li Z, Jia X, Hu Y, Chen Z, Hong J, Du L, Jin Y. A bioadhesive antioxidase-overexpressed probiotic prevents radiation enteritis by scavenging the excess reactive oxygen species. Free Radic Biol Med 2025; 227:485-498. [PMID: 39643134 DOI: 10.1016/j.freeradbiomed.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
The scavenging of the excess reactive oxygen species (ROS) induced by radiation is fundamental for radiation protection. However, directly applying antioxidants results in low bioavailability and side effects. Superoxide dismutase (SOD) and catalase (CAT) have high ROS clearance efficiency, whereas their application is limited by the enzyme inactivation, making it difficult to exhibit significant therapeutic effects. Here, we engineered a probiotic Escherichia coli Nissle 1917 (EcN), i.e., AAEcN, serving as a SOD/CAT vehicle to scavenge ROS for the prevention and treatment of radiation enteritis (RE). The overexpressed Drsod and katE in AAEcN showed 5-fold ROS elimination efficiency compared to the wild EcN. Furthermore, the intestinal retention time of engineered EcN was prolonged through trefoil factor 3 gene (TFF3) modification of curli fibers on the bacterial surface, which contributed to the persistence of antioxidant enzyme activity. We found that AAEcN rapidly eliminated the intracellular ROS induced by radiation. Only a single oral dosing of AAEcN was satisfied to alleviate the radiation damage to the small intestine, colon, and spleen. Moreover, the homeostasis of pro-/anti-inflammatory cytokines was realized. The proliferation of the intestinal stem cells and spleen hematopoietic stem cells was enhanced, while the apoptosis of mucosal cells was inhibited. Our findings suggest valuable insights into the ROS scavenging way in RE, and establish an empirical basis for developing probiotics as an antioxidant enzyme vehicle for the bacteriotherapy of RE.
Collapse
Affiliation(s)
- Ke Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Feng Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Zhangyu Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Xueli Jia
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yadan Hu
- Beijing Institute of Radiation Medicine, Beijing, 100850, China; School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Ziyuan Chen
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jinyun Hong
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Lina Du
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
2
|
Meena SK, Joriya PR, Yadav SM, Kumar R, Meena P, Patel DD. Modulation of radiation-induced intestinal injury by radioprotective agents: a cellular and molecular perspectives. REVIEWS ON ENVIRONMENTAL HEALTH 2023; 38:295-311. [PMID: 35438851 DOI: 10.1515/reveh-2021-0108] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 03/11/2022] [Indexed: 06/02/2023]
Abstract
The gastrointestinal (GI) system has rapidly proliferating and differentiating cells, which make it one of the most radiosensitive organs in the body. Exposure to high dose of ionising radiation (IR) during radiotherapy may generate a variety of reactive oxygen species (ROS) and reactive nitrogen species (RNS) including radicals, cause some side effects such as nausea, vomiting, diarrhoea, pain, ulceration, mal-absorption etc. Irradiation disrupts GI system by damaging proliferating stem cells of the crypts that alters the histology and physiology of intestine. Radiation damage reflects the qualitative and quantitative changes in intestinal epithelial stem cells like enterocytes, enteroendocrine cells, goblet cells and Paneth cells. The damaging effects of radiation to bio-molecules and cellular structures can alter gene signalling cascades and grounds genomic instability, protein modifications, cell senescence and cell death. The signalling pathways of GI tract includes Wnt, BMP, Hedgehog, PTEN/PI3K and Notch plays an important role in self-renewal of intestinal stem cells (ISCs) and maintaining the balance between self-renewal and differentiation of ISCs. Various radiation countermeasures including radioprotectors and mitigators are under development phase globally but still not approved for clinical applications during any radiation emergencies. In view of above, present review highlights cellular and molecular interruptions of GI system due to acute and chronic GI radiation injury, role of radioprotectors in signalling cascade modulations in GI epithelium and involvement of ISC markers in radioprotection.
Collapse
Affiliation(s)
- Sunil Kumar Meena
- Department of Zoology, University of Rajasthan, Jaipur, Rajasthan, India
| | - Pukha Raj Joriya
- Department of Zoology, University of Rajasthan, Jaipur, Rajasthan, India
| | - Sanwar Mal Yadav
- Department of Zoology, University of Rajasthan, Jaipur, Rajasthan, India
| | - Raj Kumar
- Institute of Nuclear Medicine and Allied Science, DRDO, Delhi, India
| | - Priyadarshi Meena
- Department of Zoology, University of Rajasthan, Jaipur, Rajasthan, India
| | - Dev Dutt Patel
- Department of Zoology, University of Rajasthan, Jaipur, Rajasthan, India
| |
Collapse
|
3
|
Shen H, Han J, Liu C, Cao F, Huang Y. Grape Seed Proanthocyanidins Exert a Radioprotective Effect on the Testes and Intestines Through Antioxidant Effects and Inhibition of MAPK Signal Pathways. Front Med (Lausanne) 2022; 8:836528. [PMID: 35141259 PMCID: PMC8818786 DOI: 10.3389/fmed.2021.836528] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/29/2021] [Indexed: 11/15/2022] Open
Abstract
The testes and intestines are highly sensitive to ionizing radiation. Low-dose radiation can cause infertility and enteritis. However, there is a lack of safe and efficient radioprotective agents. This study aims to investigate the radioprotective effects of grape seed proanthocyanidins (GSPs) on testicular and intestinal damage induced by ionizing radiation. In vitro, GSPs reduced the apoptosis and proliferation inhibition of mouse testicular stromal cells TM3 and human small intestinal crypt epithelial cells HIEC induced by ionizing radiation, and alleviated DNA double-strand breaks. In vivo, GSPs ameliorated the pathological damage of the testes and intestines induced by ionizing radiation, and protected the endocrine function of the testes and the barrier function of the intestines. In addition, we preliminarily proved that the radioprotective effect of GSPs is related to its antioxidant effect and inhibition of MAPK signaling pathways. Our results indicate that GSPs are expected to be a safe and effective radioprotective drug.
Collapse
Affiliation(s)
- Hui Shen
- Department of Central Laboratory, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jun Han
- Department of Radiology, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chunlei Liu
- Department of Radiation Oncology, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng, China
| | - Fei Cao
- Department of Radiotherapy, Changhai Hospital of Shanghai, First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yijuan Huang
- Department of Radiology, First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
4
|
Khayyal MT, Abdel-Naby DH, El-Ghazaly MA. Propolis extract protects against radiation-induced intestinal mucositis through anti-apoptotic mechanisms. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:24672-24682. [PMID: 31240658 DOI: 10.1007/s11356-019-05782-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/17/2019] [Indexed: 06/09/2023]
Abstract
Intestinal mucositis is a common side effect during radiotherapy that could be largely prevented by compounds possessing anti-inflammatory or anti-oxidant properties, including extracts of propolis containing a high proportion of flavonoids. A specially formulated aqueous extract of propolis (PWE) has been prepared in such a way to preclude the inclusion of flavonoids but contain mostly organic aromatic acids to study whether it would still protect against radiation-induced intestinal mucositis and to study the possible involvement of apoptotic pathways. Rats were exposed to a gamma radiation dose of 8 Gy from a Cesium-137 source in order to inflict intestinal mucositis. Three days before exposure, rats were given PWE orally and treatment continued for 2 more days. Twenty-four hours later, rats were sacrificed, the small intestine was excised, and sections were examined histologically. Different parameters for apoptosis, inflammation, and oxidative stress were determined in the serum and in intestinal homogenates. Radiation exposure led to histological and biochemical signs of intestinal damage. This was associated with an increase in apoptotic indicators and derangement in oxidative stress parameters. All deranged parameters were largely prevented by PWE. The findings provide evidence that the protective effect of PWE against intestinal radiation damage involves not only its anti-inflammatory and anti-oxidant effects but also its anti-apoptotic properties as well.
Collapse
Affiliation(s)
- Mohamed T Khayyal
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt.
| | - Doaa H Abdel-Naby
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, 3 Ahmad El-Zomor Street, Nasr City, Cairo, Egypt
| | - Mona A El-Ghazaly
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, 3 Ahmad El-Zomor Street, Nasr City, Cairo, Egypt
| |
Collapse
|
5
|
Upregulated epithelial junction expression represents a novel parameter of the epithelial radiation response to fractionated irradiation in oral mucosa. Strahlenther Onkol 2018; 194:771-779. [PMID: 29675597 PMCID: PMC6061151 DOI: 10.1007/s00066-018-1302-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/06/2018] [Indexed: 02/06/2023]
Abstract
Purpose During head and neck cancer treatment, the radiation response of the oral mucosa represents a frequent early side effect. Besides radiation-induced inhibition of proliferation, various other cellular responses occur. The radiation response of adherens and tight junction proteins was so far mostly investigated with large single-dose irradiation protocols, in vivo and in vitro. Therefore, the current study was initiated to investigate the impact of daily fractionated irradiation on the expression of adherens and tight junction proteins in vivo. Materials and methods Fractionation with 5 × 3 Gy/week (days 0–4, 7–11) was given to the snouts of mice. Groups of 5 animals per day were euthanized every second day between day 0 (unirradiated controls) and day 14, and their tongues subjected to histological processing. Adherens junction marker (β-catenin and E‑cadherin) and tight junction marker (claudin-1 and occludin) expression was analysed in the oral mucosa of unirradiated controls and during two weeks of fractionated irradiation. Results Adherens as well as tight junction marker proteins were rapidly and consistently upregulated in both the germinal as well as the functional layer of the oral mucosa. This represents a previously unknown parameter of the epithelial radiation response to clinically relevant fractionation protocols. Conclusion Fractionated irradiation significantly enhanced the expression of all proteins investigated. This study revealed a new parameter of the epithelial radiation response to fractionated irradiation. Electronic supplementary material The online version of this article (10.1007/s00066-018-1302-6) contains supplementary material, which is available to authorized users.
Collapse
|
6
|
Liang Y, Zhou H, Yao Y, Deng A, Wang Z, Gao B, Zhou M, Cui Y, Wang L, Zhou L, Wang B, Wang L, Liu A, Qiu L, Qian K, Lu Y, Deng W, Zheng X, Han Z, Li Y, Sun J. 12-O-tetradecanoylphorbol-13-acetate (TPA) increases murine intestinal crypt stem cell survival following radiation injury. Oncotarget 2018; 8:45566-45576. [PMID: 28545017 PMCID: PMC5542208 DOI: 10.18632/oncotarget.17269] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 03/22/2017] [Indexed: 01/03/2023] Open
Abstract
Radiation enteropathy is a common complication in cancer patients following radiation therapy. Thus, there is a need for agents that can protect the intestinal epithelium against radiation. 12-O-tetradecanoylphorbol-13-acetate (TPA) has been shown to induce differentiation and/or apoptosis in multiple cell lines and primary cells. In the current report, we studied the function of TPA in radiation induced enteropathy in cultured rat intestinal epithelial cell line IEC-6 after ionizing radiation (IR) and in mice after high dose total-body gamma-IR (TBI). In IEC-6 cells, there were reduced apoptosis and cell cycle arrest in TPA treated cells after IR. We detected a four-fold increase in crypt cell survival and a two-fold increase in animal survival post TBI in TPA treated mice. The beneficial effects of TPA were accompanied by upregulation of stem cells markers and higher level of proteins that are involved in PKC signaling pathway. In addition, TPA also decreased the TBI-augmented levels of the DNA damage indicators. The effects were only observed when TPA was given before irradiation. These results suggest that TPA has the ability to modulate intestinal crypt stem cells survival and this may represent a promising countermeasure against radiation induced enteropathy.
Collapse
Affiliation(s)
- Yaojie Liang
- Department of Geriatric Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Hongwei Zhou
- Department of Geriatric Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Yibing Yao
- Department of Geriatric Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Ailing Deng
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhihong Wang
- Department of Geriatric Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Boning Gao
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Texas, USA
| | - Minhang Zhou
- Department of Geriatric Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Yu Cui
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Lili Wang
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Lei Zhou
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Bianhong Wang
- Department of Hematology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Li Wang
- Department of Hematology, Laoshan Branch, No.401 Hospital of Chinese PLA, Qingdao, China
| | - Anqi Liu
- Department of Critical Care Medicine, Beijing Electric Power Hospital, Capital Medical University, Beijing, China
| | - Lanlan Qiu
- Department of Hematology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Kun Qian
- Department of Hematology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Yejian Lu
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Wanping Deng
- Department of Geriatric Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Xi Zheng
- Department of Geriatric Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Zhengtao Han
- Henan Tumor Research Institute, Zheng Zhou, China
| | - Yonghui Li
- Department of Hematology, Chinese PLA General Hospital, Beijing, China
| | - Junzhong Sun
- Department of Geriatric Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Modeling radiation injury-induced cell death and countermeasure drug responses in a human Gut-on-a-Chip. Cell Death Dis 2018; 9:223. [PMID: 29445080 PMCID: PMC5833800 DOI: 10.1038/s41419-018-0304-8] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/13/2017] [Accepted: 01/12/2018] [Indexed: 12/24/2022]
Abstract
Studies on human intestinal injury induced by acute exposure to γ-radiation commonly rely on use of animal models because culture systems do not faithfully mimic human intestinal physiology. Here we used a human Gut-on-a-Chip (Gut Chip) microfluidic device lined by human intestinal epithelial cells and vascular endothelial cells to model radiation injury and assess the efficacy of radiation countermeasure drugs in vitro. Exposure of the Gut Chip to γ-radiation resulted in increased generation of reactive oxygen species, cytotoxicity, apoptosis, and DNA fragmentation, as well as villus blunting, disruption of tight junctions, and compromise of intestinal barrier integrity. In contrast, pre-treatment with a potential prophylactic radiation countermeasure drug, dimethyloxaloylglycine (DMOG), significantly suppressed all of these injury responses. Thus, the human Gut Chip may serve as an in vitro platform for studying radiation-induced cell death and associate gastrointestinal acute syndrome, in addition to screening of novel radio-protective medical countermeasure drugs.
Collapse
|
8
|
Castillo GM, Nishimoto-Ashfield A, Jones CC, Kabirov KK, Zakharov A, Lyubimov AV. Protected graft copolymer-formulated fibroblast growth factors mitigate the lethality of partial body irradiation injury. PLoS One 2017; 12:e0171703. [PMID: 28207794 PMCID: PMC5313194 DOI: 10.1371/journal.pone.0171703] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 01/23/2017] [Indexed: 12/11/2022] Open
Abstract
We evaluated the mitigating effects of fibroblast growth factor 4 and 7 (FGF4 and FGF7, respectively) in comparison with long acting protected graft copolymer (PGC)-formulated FGF4 and 7 (PF4 and PF7, respectively) administered to C57BL/6J mice a day after exposure to LD50/30 (15.7 Gy) partial body irradiation (PBI) which targeted the gastrointestinal (GI) system. The PGC that we developed increased the bioavailability of FGF4 and FGF7 by 5- and 250-fold compared to without PGC, respectively, and also sustained a 24 hr presence in the blood after a single subcutaneous administration. The dose levels tested for mitigating effects on radiation injury were 3 mg/kg for the PF4 and PF7 and 1.5 mg each for their combination (PF4/7). Amifostine administered prior to PBI was used as a positive control. The PF4, PF7, or PF4/7 mitigated the radiation lethality in mice. The mitigating effect of PF4 and PF7 was similar to the positive control and PF7 was better than other mitigators tested. The plasma citrulline levels and hematology parameters were early markers of recovery and survival. GI permeability function appeared to be a late or full recovery indicator. The villus length and crypt number correlated with plasma citrulline level, indicating that it can act as a surrogate marker for these histology evaluations. The IL-18 concentrations in jejunum as early as day 4 and TPO levels in colon on day 10 following PBI showed statistically significant changes in irradiated versus non-irradiated mice which makes them potential biomarkers of radiation exposure. Other colon and jejunum cytokine levels are potentially useful but require larger numbers of samples than in the present study before their full utility can be realized.
Collapse
Affiliation(s)
| | | | | | - Kasim K. Kabirov
- Toxicology Research Laboratory, Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Alexander Zakharov
- Toxicology Research Laboratory, Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Alexander V. Lyubimov
- Toxicology Research Laboratory, Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, United States of America
| |
Collapse
|
9
|
Zheng J, Wang J, Pouliot M, Authier S, Zhou D, Loose DS, Hauer-Jensen M. Gene expression profiling in non-human primate jejunum, ileum and colon after total-body irradiation: a comparative study of segment-specific molecular and cellular responses. BMC Genomics 2015; 16:984. [PMID: 26589571 PMCID: PMC4654820 DOI: 10.1186/s12864-015-2168-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/29/2015] [Indexed: 12/26/2022] Open
Abstract
Background Although extensive studies have investigated radiation-induced injuries in particular gastrointestinal (GI) segments, a systematic comparison among the different segments on the basis of mode, magnitude and mechanism has not been reported. Here, a comparative study of segment-specific molecular and cellular responses was performed on jejunum, ileum and colon obtained at three time points (4, 7 and 12 days after irradiation) from non-human primate (Rhesus macaque) models exposed to 6.7 Gy or 7.4 Gy total body irradiation (TBI). Results Pathway analysis on the gene expression profiles identified radiation-induced time-, dose- and segment-dependent activation of tumor necrosis factor α (TNFα) cascade, tight junction, apoptosis, cell cycle control/DNA damage repair and coagulation system signaling. Activation of these signaling pathways suggests that colon sustained the severest mucosal barrier disruption and inflammation, and jejunum the greatest DNA damage, apoptosis and endothelial dysfunction. These more pronounced alterations correlate with the high incidence of macroscopic pathologies that are observed in the colon after TBI. Compared to colon and jejunum, ileum was resistant to radiation injury. In addition to the identification a marked increase of TNFα cascade, this study also identified radiation induced strikingly up-regulated tight junction gene CLDN2 (196-fold after 7.4-Gy TBI), matrix degradation genes such as MMP7 (increased 11- and 41-fold after 6.7-Gy and 7.4-Gy TBI), and anoikis mediated gene EDA2R that mediate mucosal shedding and barrier disruption. Conclusions This is the first systematic comparative study of the molecular and cellular responses to radiation injury in jejunum, ileum and colon. The strongest activation of TNFα cascades and the striking up-regulation of its down-stream matrix-dissociated genes suggest that TNFα modulation could be a target for mitigating radiation-induced mucosal barrier disruption. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2168-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Junying Zheng
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA.
| | - Junru Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA.
| | | | - Simon Authier
- CiToxLAB North America, Laval, Quebec, Canada, H7V 4B3.
| | - Daohong Zhou
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA.
| | - David S Loose
- Integrative Biology and Pharmacology, University of Texas Medical School at Houston, Houston, TX, 77030, USA.
| | - Martin Hauer-Jensen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, 72205, USA. .,Surgical Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, 72205, USA.
| |
Collapse
|
10
|
Novel regenerative peptide TP508 mitigates radiation-induced gastrointestinal damage by activating stem cells and preserving crypt integrity. J Transl Med 2015; 95:1222-33. [PMID: 26280221 PMCID: PMC4626368 DOI: 10.1038/labinvest.2015.103] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/22/2015] [Accepted: 07/07/2015] [Indexed: 01/25/2023] Open
Abstract
In recent years, increasing threats of radiation exposure and nuclear disasters have become a significant concern for the United States and countries worldwide. Exposure to high doses of radiation triggers a number of potentially lethal effects. Among the most severe is the gastrointestinal (GI) toxicity syndrome caused by the destruction of the intestinal barrier, resulting in bacterial translocation, systemic bacteremia, sepsis, and death. The lack of effective radioprotective agents capable of mitigating radiation-induced damage has prompted a search for novel countermeasures that can mitigate the effects of radiation post exposure, accelerate tissue repair in radiation-exposed individuals, and prevent mortality. We report that a single injection of regenerative peptide TP508 (rusalatide acetate, Chrysalin) 24 h after lethal radiation exposure (9 Gy, LD100/15) appears to significantly increase survival and delay mortality by mitigating radiation-induced intestinal and colonic toxicity. TP508 treatment post exposure prevents the disintegration of GI crypts, stimulates the expression of adherens junction protein E-cadherin, activates crypt cell proliferation, and decreases apoptosis. TP508 post-exposure treatment also upregulates the expression of DCLK1 and LGR5 markers of stem cells that have been shown to be responsible for maintaining and regenerating intestinal crypts. Thus, TP508 appears to mitigate the effects of GI toxicity by activating radioresistant stem cells and increasing the stemness potential of crypts to maintain and restore intestinal integrity. These results suggest that TP508 may be an effective emergency nuclear countermeasure that could be delivered within 24 h post exposure to increase survival and delay mortality, giving victims time to reach clinical sites for advanced medical treatment.
Collapse
|
11
|
Datta K, Suman S, Fornace AJ. Radiation persistently promoted oxidative stress, activated mTOR via PI3K/Akt, and downregulated autophagy pathway in mouse intestine. Int J Biochem Cell Biol 2014; 57:167-76. [PMID: 25449263 DOI: 10.1016/j.biocel.2014.10.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 09/14/2014] [Accepted: 10/17/2014] [Indexed: 12/19/2022]
Abstract
While acute effects of toxic radiation doses on intestine are well established, we are yet to acquire a complete spectrum of sub-lethal radiation-induced chronic intestinal perturbations at the molecular level. We investigated persistent effects of a radiation dose (2 Gy) commonly used as a daily fraction in radiotherapy on oxidants and anti-oxidants, and autophagy pathways, which are interlinked processes affecting intestinal homeostasis. Six to eight weeks old C57BL/6J mice (n=10) were exposed to 2 Gy γ-ray. Mice were euthanized two or twelve months after radiation, intestine surgically removed, and flushed using sterile PBS. Parts of the intestine from jejunal-ilial region were fixed, frozen, or used for intestinal epithelial cell (IEC) isolation. While oxidant levels and mitochondrial status were assessed in isolated IEC, autophagy and oxidative stress related signaling pathways were probed in frozen and fixed samples using PCR-based expression arrays and immunoprobing. Radiation exposure caused significant alterations in the expression level of 26 autophagy and 17 oxidative stress related genes. Immunoblot results showed decreased Beclin1 and LC3-II and increased p62, PI3K/Akt, and mTOR. Flow cytometry data showed increased oxidant production and compromised mitochondrial integrity in irradiated samples. Immunoprobing of intestinal sections showed increased 8-oxo-dG and nuclear PCNA, and decreased autophagosome marker LC3-II in IEC after irradiation. We show that sub-lethal radiation could persistently downregulate anti-oxidants and autophagy signaling, and upregulate oxidant production and proliferative signaling. Radiation-induced promotion of oxidative stress and downregulation of autophagy could work in tandem to alter intestinal functions and have implications for post-radiation chronic gastrointestinal diseases.
Collapse
Affiliation(s)
- Kamal Datta
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Shubhankar Suman
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Albert J Fornace
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA; Center of Excellence In Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
12
|
Khayyal MT, Abdel-Naby DH, Abdel-Aziz H, El-Ghazaly MA. A multi-component herbal preparation, STW 5, shows anti-apoptotic effects in radiation induced intestinal mucositis in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:1390-1399. [PMID: 25022208 DOI: 10.1016/j.phymed.2014.04.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 02/25/2014] [Accepted: 04/18/2014] [Indexed: 06/03/2023]
Abstract
PURPOSE Intestinal mucositis is a common adverse effect in patients undergoing radiotherapy and constitutes a treatment-limiting condition. Since no agents are yet known that can adequately guard against its development, the search continues to find safe and effective measures. The present study was intended to investigate whether the herbal preparation, STW 5, could offer a potentially effective agent in this respect. METHODS Intestinal mucositis was induced in rats by exposing them to whole body gamma-irradiation (6 Gy). Rats were treated orally with STW 5 (5 or 10 ml/kg) for five days before and two days after irradiation. One day later, rats were sacrificed and segments of small intestine were examined histologically. Intestinal homogenates and serum samples were used to assess relevant parameters for apoptosis and different markers for inflammation and oxidative stress. RESULTS Exposure to radiation produced dose-dependent extents of intestinal injury associated with apoptotic changes with high radiation levels. Apoptosis was associated with an increase in cytosolic calcium, depletion of mitochondrial cytochrome c, B-cell lymphoma-2 and complex I. Oxidative stress parameters (reduced glutathione, thiobarbituric acid reactive substance and total nitrate/nitrite) were deranged. Inflammation markers (tumor necrosis factor and myeloperoxidase) and indices of intestinal damage (serum diamine oxidase) were increased. STW 5 protected to a large extent against histological changes and counteracted the deranged parameters. CONCLUSION The findings provide experimental evidence for the potential beneficial use of STW5 in protecting against the development of radiation-induced intestinal mucositis and associated changes in tissue biomarkers.
Collapse
Affiliation(s)
- Mohamed T Khayyal
- Department of Pharmacology, Faculty of Pharmacy, Cairo University, Egypt.
| | - Doaa H Abdel-Naby
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, Egypt
| | - Heba Abdel-Aziz
- Scientific Department, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Mona A El-Ghazaly
- Department of Drug Radiation Research, National Centre for Radiation Research and Technology, Atomic Energy Authority, Egypt
| |
Collapse
|
13
|
Shiou SR, Yu Y, Guo Y, He SM, Mziray-Andrew CH, Hoenig J, Sun J, Petrof EO, Claud EC. Synergistic protection of combined probiotic conditioned media against neonatal necrotizing enterocolitis-like intestinal injury. PLoS One 2013; 8:e65108. [PMID: 23717690 PMCID: PMC3663790 DOI: 10.1371/journal.pone.0065108] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 04/26/2013] [Indexed: 11/26/2022] Open
Abstract
Balance among the complex interactions of the gut microbial community is important for intestinal health. Probiotic bacteria can improve bacterial balance and have been used to treat gastrointestinal diseases. Neonatal necrotizing enterocolitis (NEC) is a life-threatening inflammatory bowel disorder primarily affecting premature infants. NEC is associated with extensive inflammatory NF-κB signaling activation as well as intestinal barrier disruption. Clinical studies have shown that probiotic administration may protect against NEC, however there are safety concerns associated with the ingestion of large bacterial loads in preterm infants. Bacteria-free conditioned media (CM) from certain probiotic organisms have been shown to retain bioactivity including anti-inflammatory and cytoprotective properties without the risks of live organisms. We hypothesized that the CM from Lactobacillus acidophilus (La), Bifidobacterium infantis (Bi), and Lactobacillus plantarum (Lp), used separately or together would protect against NEC. A rodent model with intestinal injury similar to NEC was used to study the effect of CM from Lp, La/Bi, and La/Bi/Lp on the pathophysiology of NEC. All the CM suppressed NF-κB activation via preserved IκBα expression and this protected IκBα was associated with decreased liver activity of the proteasome, which is the degrading machinery for IκBα. These CM effects also caused decreases in intestinal production of the pro-inflammatory cytokine TNF-α, a downstream target of the NF-κB pathway. Combined La/Bi and La/Bi/Lp CM in addition protected intestinal barrier function by maintaining tight junction protein ZO-1 levels and localization at the tight junction. Double combined La/Bi CM significantly reduced intestinal injury incidence from 43% to 28% and triple combined La/Bi/Lp CM further reduced intestinal injury incidence to 20%. Thus, this study demonstrates different protective mechanisms and synergistic bioactivity of the CM from different organisms in ameliorating NEC-like intestinal injury in an animal model.
Collapse
Affiliation(s)
- Sheng-Ru Shiou
- Department of Pediatrics, Section of Neonatology, The University of Chicago, Chicago, Illinois, United States of America
| | - Yueyue Yu
- Department of Pediatrics, Section of Neonatology, The University of Chicago, Chicago, Illinois, United States of America
| | - Yuee Guo
- Department of Pediatrics, Section of Neonatology, The University of Chicago, Chicago, Illinois, United States of America
| | - Shu-Mei He
- Department of Medicine, Section of Infectious Diseases and GIDRU, Queen's University, Kingston, Ontario, Canada
| | - C. Haikaeli Mziray-Andrew
- Department of Pediatrics, Section of Pediatric Gastroenterology and Nutrition, Southern Illinois University School of Medicine, Carbondale, Illinois, United States of America
| | - Jeanette Hoenig
- Edward Hospital, Naperville, Illinois, United States of America
| | - Jun Sun
- Department of Biochemistry, Rush University, Chicago, Illinois, United States of America
| | - Elaine O. Petrof
- Department of Medicine, Section of Infectious Diseases and GIDRU, Queen's University, Kingston, Ontario, Canada
| | - Erika C. Claud
- Department of Pediatrics, Section of Neonatology, The University of Chicago, Chicago, Illinois, United States of America
- Department of Medicine, Section of Gastroenterology, The University of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
14
|
Miró L, Pérez-Bosque A, Maijó M, Amat C, Naftalin RJ, Moretó M. Aldosterone induces myofibroblast EGF secretion to regulate epithelial colonic permeability. Am J Physiol Cell Physiol 2013; 304:C918-26. [DOI: 10.1152/ajpcell.00292.2012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In vivo studies show that raised aldosterone (Aldo) during low-Na adaptation regulates the growth of pericryptal myofibroblasts and reduces the permeability of the colonic epithelium. The aim of this study was to reproduce in vitro the in vivo condition of increased Aldo using human CCD-18Co myofibroblasts and T84 colonic epithelial cells to measure myofibroblast and epithelial proliferation and the expression of intercellular junction proteins. Proliferation was quantified by measuring 5-bromo-2′-deoxyuridine incorporation. The myofibroblast expression of EGF, VEGFa, and transforming growth factor-β1 (TGF-β1) was measured by real-time PCR and the expression of junctional complex proteins by Western blot. Aldo stimulated the proliferation of myofibroblasts by 70% ( P < 0.05) and increased EGF mRNA expression by 30% ( P < 0.05) without affecting VEGFa and TGF-β1. EGF concentration in the incubation medium increased by 30% ( P < 0.05) 24 h after Aldo addition, and these effects were prevented by the addition of spironolactone. Myofibroblast proliferation in response to Aldo was mediated by EGF receptor (EGFR) and involved both MAPKK and phosphatidylinositol 3-kinase pathways. When T84 cells were incubated with medium from myofibroblasts stimulated with Aldo (conditioned medium), the expression of β-catenin and claudin IV was increased by 30% ( P < 0.05) and proliferation by 40% ( P < 0.05). T84 proliferation decreased when α-EGF, or the EGFR antagonist AG1478, was present. Results in vivo indicate that rats fed a low-salt diet showed an increased expression of EGF and EGFR in the colonic mucosa. These results support the view that changes in colonic permeability during low-Na adaptation are mediated by the EGF secreted by myofibroblasts in response to raised Aldo.
Collapse
Affiliation(s)
- Lluïsa Miró
- Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, and Institut de Recerca en Nutrició i Seguretat Alimentària, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; and
| | - Anna Pérez-Bosque
- Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, and Institut de Recerca en Nutrició i Seguretat Alimentària, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; and
| | - Mònica Maijó
- Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, and Institut de Recerca en Nutrició i Seguretat Alimentària, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; and
| | - Concepció Amat
- Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, and Institut de Recerca en Nutrició i Seguretat Alimentària, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; and
| | - Richard J. Naftalin
- Physiology Department, King's College London, Franklin-Wilkins Building, Waterloo Campus, London, United Kingdom
| | - Miquel Moretó
- Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, and Institut de Recerca en Nutrició i Seguretat Alimentària, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; and
| |
Collapse
|
15
|
Shiou SR, Yu Y, Chen S, Ciancio MJ, Petrof EO, Sun J, Claud EC. Erythropoietin protects intestinal epithelial barrier function and lowers the incidence of experimental neonatal necrotizing enterocolitis. J Biol Chem 2011; 286:12123-32. [PMID: 21262973 PMCID: PMC3069416 DOI: 10.1074/jbc.m110.154625] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 12/28/2010] [Indexed: 11/06/2022] Open
Abstract
The impermeant nature of the intestinal barrier is maintained by tight junctions (TJs) formed between adjacent intestinal epithelial cells. Disruption of TJs and loss of barrier function are associated with a number of gastrointestinal diseases, including neonatal necrotizing enterocolitis (NEC), the leading cause of death from gastrointestinal diseases in preterm infants. Human milk is protective against NEC, and the human milk factor erythropoietin (Epo) has been shown to protect endothelial cell-cell and blood-brain barriers. We hypothesized that Epo may also protect intestinal epithelial barriers, thereby lowering the incidence of NEC. Our data demonstrate that Epo protects enterocyte barrier function by supporting expression of the TJ protein ZO-1. As immaturity is a key factor in NEC, Epo regulation of ZO-1 in the human fetal immature H4 intestinal epithelial cell line was examined and demonstrated Epo-stimulated ZO-1 expression in a dose-dependent manner through the PI3K/Akt pathway. In a rat NEC model, oral administration of Epo lowered the incidence of NEC from 45 to 23% with statistical significance. In addition, Epo treatment protected intestinal barrier function and prevented loss of ZO-1 at the TJs in vivo. These effects were associated with elevated Akt phosphorylation in the intestine. This study reveals a novel role of Epo in the regulation of intestinal epithelial TJs and barrier function and suggests the possible use of enteral Epo as a therapeutic agent for gut diseases.
Collapse
Affiliation(s)
- Sheng-Ru Shiou
- From the Department of Pediatrics, Section of Neonatology, and
| | - Yueyue Yu
- From the Department of Pediatrics, Section of Neonatology, and
| | - Sangzi Chen
- From the Department of Pediatrics, Section of Neonatology, and
| | - Mae J. Ciancio
- Biomedical Sciences, Midwestern University, Chicago, Illinois 60515
| | - Elaine O. Petrof
- the Department of Medicine, Queen's University, Kingston, Ontario K7L 3M6, Canada, and
| | - Jun Sun
- the Department of Medicine, University of Rochester, Rochester, New York 14627
| | - Erika C. Claud
- From the Department of Pediatrics, Section of Neonatology, and
- Department of Medicine, Section of Gastroenterology, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
16
|
Bamba S, Lee CY, Brittan M, Preston SL, Direkze NC, Poulsom R, Alison MR, Wright NA, Otto WR. Bone marrow transplantation ameliorates pathology in interleukin-10 knockout colitic mice. J Pathol 2006; 209:265-73. [PMID: 16550633 DOI: 10.1002/path.1967] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The authors have previously reported the derivation of colonic subepithelial myofibroblasts (SEMFs) in both humans and mice from bone marrow (BM). In the pathogenesis of inflammatory bowel disease (IBD), such as Crohn's disease and ulcerative colitis, colonic SEMFs mediate several types of inflammatory response. In the present study, interleukin (IL)-10-/- mice were used as a model of IBD to investigate the involvement of BM-derived cells in the inflamed mucosa. Male whole BM [either C57/BL10 (wild type: WT) or IL-10-/- donor mice] was used to perform bone marrow transplantation (BMT) into both WT and IL-10-/- female mice. Tissue samples were evaluated by immunohistochemistry for alpha-smooth muscle actin expression and by in situ hybridization using a Y-chromosome-specific probe to track the donor-derived colonic SEMFs. The mucosal expression of mRNA for pro-inflammatory cytokines was analysed by reverse transcriptase-polymerase chain reaction (RT-PCR). In addition, mRNA expression of matrix metalloproteinase (MMP)-7 and osteopontin in the inflamed mucosa was assessed using in situ hybridization. Body weights and histological scores showed that IL-10-/- mice that received WT BM had an improved course of colitis, decreased mucosal pro-inflammatory mRNA expression, and up to 30% of their SEMFs were of BM origin. Conversely, IL-10-/- mice receiving IL-10-/- BM progressed to extensive colitis, and Y probe analysis revealed that up to 45% of colonic SEMFs were of BM origin. WT mice receiving IL-10-/- or WT BM had no signs of colonic inflammation. The expression of MMP-7 and osteopontin was up-regulated in the inflamed mucosa. In conclusion, IL-10-/- mice displayed ameliorated disease activity after WT BMT, whilst colitis was not induced in WT mice by IL-10-/- BMT. The contribution of BM-derived cells to colonic SEMFs was significantly increased in the inflamed mucosa compared with non-inflamed mucosa.
Collapse
Affiliation(s)
- S Bamba
- Histopathology Unit, London Research Institute, Cancer Research UK, London WC2A 3PX, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Cristià E, Afzal-Ahmed I, Pérez-Bosque A, Amat C, Naftalin RJ, Moretó M. Pericryptal myofibroblast growth in rat descending colon induced by low-sodium diets is mediated by aldosterone and not by angiotensin II. J Membr Biol 2006; 206:53-9. [PMID: 16440181 DOI: 10.1007/s00232-005-0773-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2005] [Indexed: 10/25/2022]
Abstract
Pericryptal myofibroblast growth in descending colonic crypts correlates with the activation of the renin-angiotensin-aldosterone system. Earlier work showed that during the transition from a high-Na(+) (HS) to low-Na(+) (LS) diet there are changes in the colonic crypt wall and pericryptal sheath. As LS diet increases both aldosterone and angiotensin II, the aim here was to determine their individual contributions to the trophic changes in colonic crypts. Experiments were conducted on control and adrenalectomized Sprague-Dawley rats fed an HS diet and then switched to LS diet for 3 days and supplemented with aldosterone or angiotensin II. The actions of the angiotensin-converting enzyme inhibitor captopril, the angiotensin receptor antagonist losartan and the aldosterone antagonist spironolactone on extracellular matrix proteins, claudin 4 and E-cadherin myofibroblast proteins, alpha-smooth muscle actin (alpha-SMA) and OB-cadherin (cadherin 11), angiotensin type 1 and TGFbetar1 membrane receptors were determined by immunolocalization in fixed distal colonic mucosa. The LS diet or aldosterone supplementation following ADX in HS or LS increased extracellular matrix, membrane receptors and myofibroblast proteins, but angiotensin alone had no trophic effect on alpha-SMA. These results show that aldosterone stimulates myofibroblast growth in the distal colon independently of dietary Na(+) intake and of angiotensin levels. This stimulus could be a genomic response or secondary to stretch of the pericryptal sheath myofibroblasts accompanying enhanced rates of crypt fluid absorption.
Collapse
Affiliation(s)
- E Cristià
- Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
18
|
Andoh A, Bamba S, Fujiyama Y, Brittan M, Wright NA. Colonic subepithelial myofibroblasts in mucosal inflammation and repair: contribution of bone marrow-derived stem cells to the gut regenerative response. J Gastroenterol 2005; 40:1089-99. [PMID: 16378172 DOI: 10.1007/s00535-005-1727-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2005] [Accepted: 10/11/2005] [Indexed: 02/04/2023]
Affiliation(s)
- Akira Andoh
- Department of Gastroenterology, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Japan
| | | | | | | | | |
Collapse
|
19
|
A proteomics approach to identify changes in protein profiles in pre-cancerous colon. Biochem Biophys Res Commun 2005; 330:81-7. [PMID: 15781235 DOI: 10.1016/j.bbrc.2005.02.125] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Indexed: 01/04/2023]
Abstract
The development of colon cancer is characterised by alterations in multiple genetic and epigenetic pathways in colon tissue leading ultimately to deregulation of colon epithelial cells. Early detection is an important factor in decreasing colon cancer deaths. Proteomic techniques were used to identify potential early markers in colon tissue exhibiting pre-cancerous activity that may characterise pathological changes in a chemically induced colon cancer rat model. Protein profiles were assessed in soluble and insoluble fractions prepared from distal colon of rats treated with the colonotropic carcinogen, dimethylhydrazine. Alterations in protein profiles were associated with the presence of aberrant crypt foci, hyperplasia and dysplasia, microanatomical changes, and metabolic changes in rat colon. These changes may have a potential role in the identification of pre-pathological features preceding colon tumorigenesis.
Collapse
|
20
|
Dublineau I, Lebrun F, Grison S, Griffiths NM. Functional and structural alterations of epithelial barrier properties of rat ileum following X-irradiation. Can J Physiol Pharmacol 2005; 82:84-93. [PMID: 15052289 DOI: 10.1139/y03-129] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Irradiation of the digestive system leads to alterations of the small intestine. We have characterized the disruption of the barrier integrity in rat ileum from 1 to 14 days following irradiation ranging from 6 to 12 Gy. The intestinal permeability to 14C-mannitol and 3H-dextran 70 000 was measured in vitro in Ussing chambers. In parallel to these functional studies, immunohistochemical analyses of junctional proteins (ZO-1 and beta-catenin) of ileal epithelium were performed by confocal microscopy. Irradiation with 10 Gy induced a marked decrease in epithelial tissue resistance at three days and a fivefold increase in mannitol permeability, without modifications of dextran permeability. A disorganization of the localization for ZO-1 and beta-catenin was also observed. At 7 days after irradiation, we observed a recovery of the organization of junctional proteins in parallel to a return of intestinal permeability to control value. In addition to these time-dependent effects, a gradual effect on epithelial integrity of the radiation doses was observed 3 days after irradiation. This study shows a disruption of the integrity of the intestinal barrier in rat ileum following abdominal X-irradiation, depending on the time postirradiation and on the delivered dose. The loss of barrier integrity was characterized by a disorganization of proteins of tight and adherent junctions, leading to increased intestinal permeability to mannitol.
Collapse
Affiliation(s)
- I Dublineau
- Institut de Radioprotection et de Sûreté Nucléaire, Direction de la RadioProtection de l'Homme, Service de Radiobiologie et d'Epidemiologie, IRSN, BP 17, F-92262 Fontenay-aux-Roses, CEDEX, France.
| | | | | | | |
Collapse
|
21
|
Monti P, Wysocki J, van der Meeren A, Griffiths NM. The contribution of radiation-induced injury to the gastrointestinal tract in the development of multi-organ dysfunction syndrome or failure. Br J Radiol 2005. [DOI: 10.1259/bjr/53186341] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
22
|
Nagtegaal ID, Gaspar CGS, Peltenburg LTC, Marijnen CAM, Kapiteijn E, van de Velde CJH, Fodde R, van Krieken JHJM. Radiation induces different changes in expression profiles of normal rectal tissue compared with rectal carcinoma. Virchows Arch 2004; 446:127-35. [PMID: 15602683 DOI: 10.1007/s00428-004-1160-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2003] [Accepted: 08/20/2004] [Indexed: 01/21/2023]
Abstract
PURPOSE Radiotherapy is a very effective adjuvant treatment for rectal cancer with little side effects. Its killing effect on tumor cells seems to be more profound than the effect on normal tissue. The molecular events caused by irradiation are mainly analyzed in in vitro and animal models; investigations on human material are rare. In the current study, we analyzed the effects of irradiation on gene expression in normal and tumor tissue of rectal cancer patients. METHODS AND MATERIALS Normal and carcinoma tissue of patients from a randomized clinical trial of the benefits of preoperative radiotherapy were analyzed using the Affymetrix Human Cancer Gene Chip. Preoperative radiotherapy was given within 5 days prior to surgery. Results for normal tissue and tumor were compared to investigate the radiation-related differences between normal and tumor cells. We clustered the differentially expressed genes based on their functional annotation. Results were compared with immunohistochemical and literature data. RESULTS The majority of the investigated cancer-related genes remained unchanged by irradiation (92% in tumor tissue and 93% in normal tissue). The differentially expressed genes varied between tumor and normal tissue except for maspin and IL-8. Both in tumor and normal tissue, differentially expressed genes were present related to cell signaling and cycle control, apoptosis and cell survival and tissue response and repair. However, the spectrum of affected genes was totally different. CONCLUSION Pre-existing differences in gene expression between normal tissue and tumor tissue might explain the differences in their responses to radiation. This change in response may explain the clinical beneficial effect of radiotherapy on tumor cells (low local recurrence rate) and the less severe effects on normal tissue (minor side effects).
Collapse
Affiliation(s)
- I D Nagtegaal
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Scanff P, Souidi M, Grison S, Griffiths NM, Gourmelon P. Alteration of the enterohepatic recirculation of bile acids in rats after exposure to ionizing radiation. Can J Physiol Pharmacol 2004; 82:114-24. [PMID: 15052292 DOI: 10.1139/y03-131] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this work was to study acute alterations of the enterohepatic recirculation (EHR) of bile acids 3 days after an 8-Gy radiation exposure in vivo in the rat by a washout technique. Using this technique in association with HPLC analysis, the EHR of the major individual bile acids was determined in control and irradiated animals. Ex vivo ileal taurocholate absorption was also studied in Ussing chambers. Major hepatic enzyme activities involved in bile acid synthesis were also measured. Measurements of bile acid intestinal content and intestinal absorption efficiency calculation from washout showed reduced intestinal absorption with significant differences from one bile acid to another: absorption of taurocholate and tauromuricholate was decreased, whereas absorption of the more hydrophobic taurochenodeoxycholate was increased, suggesting that intestinal passive diffusion was enhanced, whereas ileal active transport might be reduced. Basal hepatic secretion was increased only for taurocholate, in accordance with the marked increase of CYP8B1 activity in the liver. The results are clearly demonstrate that concomitantly with radiation-induced intestinal bile acid malabsorption, hepatic bile acid synthesis and secretion are also changed. A current working model for pathophysiological changes in enterohepatic recycling after irradiation is thus proposed.Key words: irradiation, bile acids, intestine, liver, enterohepatic recycling.
Collapse
Affiliation(s)
- P Scanff
- Institut de Radioprotection et de Sûreté Nucléaire, Direction de la RadioProtection de l'Homme, Service de Radiobiologie et d'Epidemiologie, IRSN, B.P. n 17, F-92262 Fontenay-aux-Roses CEDEX, France.
| | | | | | | | | |
Collapse
|
24
|
Abstract
Chemotherapy and radiotherapy, whilst highly effective in the treatment of neoplasia, can also cause damage to healthy tissue. In particular, the alimentary tract may be badly affected. Severe inflammation, lesioning and ulceration can occur. Patients may experience intense pain, nausea and gastro-enteritis. They are also highly susceptible to infection. The disorder (mucositis) is a dose-limiting toxicity of therapy and affects around 500 000 patients world-wide annually. Oral and intestinal mucositis is multi-factorial in nature. The disruption or loss of rapidly dividing epithelial progenitor cells is a trigger for the onset of the disorder. However, the actual dysfunction that manifests and its severity and duration are greatly influenced by changes in other cell populations, immune responses and the effects of oral/gut flora. This complexity has hampered the development of effective palliative or preventative measures. Recent studies have concentrated on the use of bioactive/growth factors, hormones or interleukins to modify epithelial metabolism and reduce the susceptibility of the tract to mucositis. Some of these treatments appear to have considerable potential and are at present under clinical evaluation. This overview deals with the cellular changes and host responses that may lead to the development of mucositis of the oral cavity and gastrointestinal tract, and the potential of existing and novel palliative measures to limit or prevent the disorder. Presently available treatments do not prevent mucositis, but can limit its severity if used in combination. Poor oral health and existing epithelial damage predispose patients to mucositis. The elimination of dental problems or the minimization of existing damage to the alimentary tract, prior to the commencement of therapy, lowers their susceptibility. Measures that reduce the flora of the tract, before therapy, can also be helpful. Increased production of free radicals and the induction of inflammation are early events in the onset of mucositis. Prophylactic administration of scavengers or anti-inflammatories can partially counteract or limit some of these therapy-mediated effects, as can the use of cryotherapy. The regular use of mouthwashes, mouth coatings, antibiotics and analgesics is essential, prior to and during loss and ablation of the epithelial layer. Granulocyte-macrophage colony-stimulating factor/granulocyte colony-stimulating factor or the use of laser light therapy may aid restitution and repair. Glutamine supplements may be beneficial in the repair/recovery phase.
Collapse
Affiliation(s)
- M Duncan
- Rowett Research Institute, Bucksburn, Aberdeen, UK
| | | |
Collapse
|
25
|
Abstract
Intestinal barrier function regulates transport and host defense mechanisms at the mucosal interface with the outside world. Transcellular and paracellular fluxes are tightly controlled by membrane pumps, ion channels and tight junctions, adapting permeability to physiological needs. Food and microbial antigens are under constant surveillance of the mucosal immune system. Tolerance against commensals and immunity against pathogens require intact antigen uptake, recognition, processing and response mechanisms. Disturbance at any level, but particularly bacterial translocation due to increased permeability and breakdown of oral tolerance due to compromised epithelial and T cell interaction, can result in inflammation and tissue damage. New therapeutic approaches including probiotics and peptides to restore disrupted barrier function are evolving.
Collapse
Affiliation(s)
- Daniel C Baumgart
- Charité Medical Center - Virchow Hospital, Medical School of the Humboldt-University of Berlin, Department of Medicine, Division of Hepatology and Gastroenterology, Germany.
| | | |
Collapse
|
26
|
Brittan M, Hunt T, Jeffery R, Poulsom R, Forbes SJ, Hodivala-Dilke K, Goldman J, Alison MR, Wright NA. Bone marrow derivation of pericryptal myofibroblasts in the mouse and human small intestine and colon. Gut 2002; 50:752-7. [PMID: 12010874 PMCID: PMC1773238 DOI: 10.1136/gut.50.6.752] [Citation(s) in RCA: 187] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS In order to establish whether extraintestinal cells contribute to the turnover and repair of gastrointestinal tissues, we studied the colons and small intestines of female mice that had received a male bone marrow transplant, together with gastrointestinal biopsies from female patients that had developed graft versus host disease after receiving a bone marrow transplant from male donors. METHODS Using in situ hybridisation to detect Y chromosomes and immunohistochemistry, we demonstrated that cells derived from injected bone marrow frequently engrafted into the intestine and differentiated into pericryptal myofibroblasts. RESULTS In the human intestine, we confirmed by combining in situ hybridisation with immunostaining for smooth muscle actin that the bone marrow derived cells within the intestine exhibited a myofibroblast phenotype. In female mouse recipients of male bone marrow grafts, we observed colocalisation of Y chromosomes and clusters of newly formed marrow derived myofibroblasts. While few of these were present at seven days after bone marrow transplantation, they were numerous at 14 days, and by six weeks entire columns of pericryptal myofibroblasts could be seen running up the sides of crypts in both the small intestine and colon. These columns appeared to extend into the villi in the small intestine. Within the intestinal lamina propria, these Y chromosome positive cells were negative for the mouse macrophage marker F4/80 antigen and CD34. CONCLUSIONS Bone marrow derived pericryptal myofibroblasts were present in the mouse intestine following irradiation and bone marrow transplant, and in the intestines of human patients suffering graft versus host disease following a bone marrow transplant. Our data indicate that bone marrow cells contribute to the regeneration of intestinal myofibroblasts and epithelium after damage, and we suggest that this could be exploited therapeutically.
Collapse
Affiliation(s)
- M Brittan
- Histopathology Unit, Cancer Research UK, London, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Thiagarajah JR, Griffiths NM, Pedley KC, Naftalin RJ. Evidence for modulation of pericryptal sheath myofibroblasts in rat descending colon by transforming growth factor beta and angiotensin II. BMC Gastroenterol 2002; 2:4. [PMID: 11872151 PMCID: PMC65696 DOI: 10.1186/1471-230x-2-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2001] [Accepted: 02/12/2002] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Absorption of water and Na+ in descending colonic crypts is dependent on the barrier function of the surrounding myofibroblastic pericryptal sheath. Here the effects of high and low Na+ diets and exposure to whole body ionising radiation on the growth and activation of the descending colonic pericryptal myofibroblasts are evaluated. In addition the effect of a post-irradiation treatment with the angiotensin converting enzyme inhibitor Captopril was investigated. METHODS The levels of Angiotensin II type 1 receptor (AT1), ACE, collagen type IV, transforming growth factor-beta type 1 receptor (TGF-betaR1), OB cadherin and alpha-smooth muscle actin in both descending colon and caecum were evaluated, using immunocytochemistry and confocal microscopy, in rats fed on high and low Na+ diets (LS). These parameters were also determined during 3 months post-irradiation with 8Gy from a 60Co source in the presence and absence of the angiotensin converting enzyme inhibitor, Captopril. RESULTS Increases in AT1 receptor (135.6% +/- 18.3, P < 0.001); ACE (70.1% +/- 13.1, P < 0.001); collagen type IV (49.6% +/- 15.3, P < 0.001); TGF-+/-beta1 receptors (291.0% +/- 26.5, P < 0.001); OB-cadherin (26.3% +/- 13.8, P < 0.05) and alpha-smooth muscle actin (82.5% +/- 12.4, P < 0.001) were observed in the pericryptal myofibroblasts of the descending colon after LS diet. There are also increases in AT1 receptor and TGF-beta1 receptor, smooth muscle actin and collagen type IV after irradiation. Captopril reduced all these effects of irradiation on the pericryptal sheath and also decreased the amount of collagen and smooth muscle actin in control rats (P < 0.001). CONCLUSIONS These results demonstrate an activation of descending colonic myofibroblasts to trophic stimuli, or irradiation, which can be attenuated by Captopril, indicative of local trophic control by angiotensin II and TGF-beta release.
Collapse
Affiliation(s)
- Jay R Thiagarajah
- Division of Physiology, School of Biomedical Sciences, King's College London, Guys Campus, London, UK
| | - Nina M Griffiths
- Institut de Protection et de Sûreté Nucléaire, Fontenay aux Roses, France
| | - Kevin C Pedley
- Division of Physiology, School of Biomedical Sciences, King's College London, Guys Campus, London, UK
| | - Richard J Naftalin
- Division of Physiology, School of Biomedical Sciences, King's College London, Guys Campus, London, UK
| |
Collapse
|
28
|
Dublineau I, Morel E, Griffiths NM. Characterization of altered absorptive and secretory functions in the rat colon after abdominal irradiation: comparison with the effects of total-body irradiation. Radiat Res 2002; 157:52-61. [PMID: 11754642 DOI: 10.1667/0033-7587(2002)157[0052:coaaas]2.0.co;2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The aim of this work was to determine the alterations in the absorptive and secretory functions of the rat colon after abdominal irradiation and to compare the effects of abdominal and whole-body irradiation. Rats received an abdominal irradiation with 8 to 12 Gy and were studied at 1, 4 and 7 days after exposure. Water and electrolyte absorption was measured in vivo by insertion of an agarose cylinder into the colons of anesthetized rats. In vitro measurements of potential difference, short-circuit current and tissue conductance were performed in Ussing chambers under basal and agonist-stimulated conditions. Most of the changes appeared at 4 days after abdominal irradiation. At this time, a decrease in water and electrolyte absorption in the colon was observed for radiation doses > or = 9 Gy. The response to secretagogues (VIP, 5-HT and forskolin) was attenuated after 10 and 12 Gy. Epithelial integrity, estimated by potential difference and tissue conductance, was altered from 1 to 7 days after 12 Gy abdominal irradiation. These results show that the function of the colon was affected by abdominal irradiation. Comparison with earlier results for total-body irradiation demonstrated a difference of 2 Gy in the radiation dose needed to induce changes in the function of the colon.
Collapse
Affiliation(s)
- I Dublineau
- Institut de Protection et de Sûreté Nucléaire, Département de Protection de la santé de l'Homme et de Dosimétrie, IPSN, BP no. 6, F-92265 Fontenay-aux-Roses Cédex, France.
| | | | | |
Collapse
|
29
|
Thiagarajah JR, Jayaraman S, Naftalin RJ, Verkman AS. In vivo fluorescence measurement of Na(+) concentration in the pericryptal space of mouse descending colon. Am J Physiol Cell Physiol 2001; 281:C1898-903. [PMID: 11698248 DOI: 10.1152/ajpcell.2001.281.6.c1898] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A method involving surgical exposure of the colonic mucosa, fluorescent dye addition, and confocal microscopy has been developed for monitoring colonic crypt function in vivo in mice. Na(+) concentration in the extracellular pericryptal space of descending colon was measured using a low-affinity Na(+)-sensitive fluorescent indicator consisting of an Na(+)-sensitive chromophore (sodium red) and an Na(+)-insensitive chromophore (Bodipy-fl) immobilized on 200-nm-diameter polystyrene beads. The Na(+) indicator beads accumulated in the pericryptal spaces surrounding the colonic crypts after a 1-h exposure of the colonic luminal surface to the bead suspension. Na(+) concentration ([Na(+)]) in the pericryptal space was 491 +/- 62 mM (n = 4). After a 70-min exposure to amiloride (0.25 mM), pericryptal [Na(+)] was reduced to 152 +/- 21 mM. Blockage of the crypt lumen with mineral oil droplets reduced pericryptal [Na(+)] to 204 +/- 44 mM. Exposure of the colonic mucosa to FITC-dextran (4.5 kDa) led to rapid accumulation of the dye into the crypt lumen with a half time of 19.8 +/- 1.0 s, which was increased to 77.9 +/- 6.0 s after amiloride treatment. These results establish an in vivo fluorescence method to measure colonic crypt function and provide direct evidence for accumulation of a hypertonic absorbate in the pericryptal space of descending colon. The pericryptal space represents the first example of a hypertonic extracellular compartment in mammals that is not created by a countercurrent amplification mechanism.
Collapse
Affiliation(s)
- J R Thiagarajah
- Division of Physiology, Centre for Vascular Biology and Medicine, King's College London, Guy's Campus, London SE1 1UL, United Kingdom
| | | | | | | |
Collapse
|
30
|
Thiagarajah JR, Pedley KC, Naftalin RJ. Evidence of amiloride-sensitive fluid absorption in rat descending colonic crypts from fluorescence recovery of FITC-labelled dextran after photobleaching. J Physiol 2001; 536:541-53. [PMID: 11600688 PMCID: PMC2278881 DOI: 10.1111/j.1469-7793.2001.0541c.xd] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2001] [Accepted: 05/29/2001] [Indexed: 11/27/2022] Open
Abstract
1. Fluorescence recovery after photobleaching (FRAP) of fluorescein isothiocyanate (FITC)-labelled 10 and 250 kDa dextran (FITC dextran) in isolated rat descending colonic crypts was measured at 35 degrees C using laser scanning confocal microscopy. 2. FRAP of either 10 or 250 kDa FITC dextran in crypt lumens was almost complete within 2-3 min. 3. In the presence of amiloride (0.1 mM), or in the absence of Na+, the rate of FITC dextran uptake into the crypt lumens was reduced by 70-80 %. 4. The rate of fluid uptake into the crypt lumen, as estimated from the rate of total FITC dextran uptake into the crypt lumen and its adjacent pericryptal region after FRAP, was between 1.3 x 10(-3) and 1.7 x 10(-3) cm x s(-1). 5. Convective flow during FRAP was also determined from the initial rate of FITC dextran advance along the crypt lumen. This effect was almost completely blocked by amiloride (0.1 mM). 6. The permeability of 10 kDa FITC dextran across the descending colonic crypt wall was found to be higher than that of 250 kDa FITC dextran (3.7 (+/- 0.6) x 10(-5) and 1.8 (+/- 0.3) x 10(-6) cm x s(-1), respectively; n = 3 for both, P < 0.01). The permeability of the caecal crypt wall to 10 kDa dextran was higher than that of the descending crypt wall (2.03 (+/- 0.21) x 10(-5) cm x s(-1); n = 3, P < 0.025). 7. Simulation of the flow of Na+, water and FITC dextran into the crypt lumen and across the crypt wall and pericryptal sheath corroborates the observed parameters of water and Na+ flows.
Collapse
Affiliation(s)
- J R Thiagarajah
- Division of Physiology, Centre for Cardiovascular Biology and Medicine, King's College London, Guys Campus, London EC1 1UL, UK
| | | | | |
Collapse
|
31
|
Abstract
Apoptosis has an important role in carcinogenesis and cancer treatment. The end result of this complex pathway is the formation of apoptotic bodies. These can be difficult to quantify accurately, but quantitation is important if we wish to study this process. Several techniques are available which can help. 'TUNEL' is discussed, with its potential drawbacks, and newer antibody techniques, such as M30 and caspase 3, are then reviewed.
Collapse
Affiliation(s)
- J A Walker
- Department of Pathology, Algernon Firth Building, University of Leeds, Leeds LS2 9JT, UK
| | | |
Collapse
|