1
|
Ghosh S, Morita A, Nishiyama Y, Sakaue M, Fujiwara K, Morita D, Sonoyama Y, Higashi Y, Sasatani M. Rectal Epithelial Stem Cell Kinetics in Acute Radiation Proctitis. Int J Mol Sci 2024; 25:11252. [PMID: 39457033 PMCID: PMC11508457 DOI: 10.3390/ijms252011252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
The intestinal tract is a typical radiosensitive tissue, and radiation rectal injury is a severe side effect that limits the prescribed dose in radiotherapy of the abdominal and pelvic region. Understanding the post-irradiation kinetics of Lgr5-positive stem cells is crucial in comprehending this adverse process. In this study, we utilized Lgr5-EGFP knock-in mice expressing EGFP and LGR5 antibody fluorescence staining of wild-type mice. At the state of radiation injury, the qPCR analysis showed a significant decrease in the expression level of Lgr5 in the rectal epithelial tissue. The dose-response relationship analysis showed that at low to moderate doses up to 10 gray (Gy), Lgr5-clustered populations were observed at the base of the crypt, whereas at sublethal doses (20 Gy and 29 Gy), the cells exhibited a dot-like scatter pattern, termed Lgr5-dotted populations. During recovery, 30 days post-irradiation, Lgr5-clustered populations gradually re-emerged while Lgr5-dotted populations declined, implying that some of the Lgr5-dotted stem cell populations re-clustered, aiding regenerations. Based on statistical analysis of the dose-response relationship using wild-type mice, the threshold dose for destroying these stem cell structures is 18 Gy. These findings may help set doses in mouse abdominal irradiation experiments for radiation intestinal injury and for understanding the histological process of injury development.
Collapse
Affiliation(s)
- Sharmila Ghosh
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (S.G.); (Y.N.); (M.S.); (K.F.); (D.M.); (Y.S.); (Y.H.)
| | - Akinori Morita
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (S.G.); (Y.N.); (M.S.); (K.F.); (D.M.); (Y.S.); (Y.H.)
| | - Yuichi Nishiyama
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (S.G.); (Y.N.); (M.S.); (K.F.); (D.M.); (Y.S.); (Y.H.)
| | - Masahiro Sakaue
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (S.G.); (Y.N.); (M.S.); (K.F.); (D.M.); (Y.S.); (Y.H.)
| | - Ken Fujiwara
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (S.G.); (Y.N.); (M.S.); (K.F.); (D.M.); (Y.S.); (Y.H.)
| | - Daiki Morita
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (S.G.); (Y.N.); (M.S.); (K.F.); (D.M.); (Y.S.); (Y.H.)
| | - Yuichiro Sonoyama
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (S.G.); (Y.N.); (M.S.); (K.F.); (D.M.); (Y.S.); (Y.H.)
| | - Yuichi Higashi
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (S.G.); (Y.N.); (M.S.); (K.F.); (D.M.); (Y.S.); (Y.H.)
| | - Megumi Sasatani
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 754-8553, Japan;
| |
Collapse
|
2
|
Demyashkin G, Karakaeva E, Saakian S, Shchekin V, Elbuzdukaev E, Bamatgiraev U, Ashgalieva D, Evsultanova M, Kovalev D, Kabanova D, Shatunov O, Atiakshin D. Morphological aspects of small intestinal mucosal injury and repair after electron irradiation. Anat Cell Biol 2024; 57:384-391. [PMID: 38880781 PMCID: PMC11424566 DOI: 10.5115/acb.24.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/23/2024] [Accepted: 04/12/2024] [Indexed: 06/18/2024] Open
Abstract
Morphological evaluation of the small intestine mucosa and apoptosis activity (caspase-3) is necessary to assess the severity of damage to the small intestine. At the same time, proliferative index based on Ki-67 can be used to assess the regenerative potential of the small intestine. Fragments of small intestine of Wistar rats (n=60) of three groups: I) control (n=20); II) experimental group (n=20; local single electron irradiation at a dose of 2 Gy), III) experimental group (n=20; local single electron irradiation at a dose of 8 Gy) were studied by light microscopy using hematoxylin and eosin staining and immunohistochemical reactions with antibodies to Ki-67 and caspase-3. In all samples of the experimental groups, a decrease in all morphometric indices was observed on day 1 with a tendency to recover on day 3. Small intestinal electron irradiation led to disturbances in the histoarchitecture of varying severity, and an increase in cell apoptosis was observed (increased expression of caspase-3 and decrease in Ki-67). In addition, modulation of the PI3K/AKT and MAPK/ERK signaling pathways was detected. The most pronounced destructive changes were observed in the group of 8 Gy single electron irradiation. Local irradiation of the small intestine with electrons at a dose of 2 and 8 Gy results in a decrease in the number of enterocytes, mainly stem cells of the intestinal crypts.
Collapse
Affiliation(s)
- Grigory Demyashkin
- Laborant of the Labrotory of Histology and Immunohistochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Elza Karakaeva
- National Medical Research Centre of Radiology, Ministry of Health of Russia, Moscow, Russia
| | - Siuzanna Saakian
- National Medical Research Centre of Radiology, Ministry of Health of Russia, Moscow, Russia
| | - Vladimir Shchekin
- Laborant of the Labrotory of Histology and Immunohistochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Emir Elbuzdukaev
- Laborant of the Labrotory of Histology and Immunohistochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Umar Bamatgiraev
- Laborant of the Labrotory of Histology and Immunohistochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Daniia Ashgalieva
- Federal State Budget Educational Institution of Higher Education A.I. Yevdokimov Moscow State University of Medicine and Dentistry (MSUMD), Moscow, Russia
| | - Makka Evsultanova
- Laborant of the Labrotory of Histology and Immunohistochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Daniil Kovalev
- Laborant of the Labrotory of Histology and Immunohistochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Darya Kabanova
- Federal State Budget Educational Institution of Higher Education A.I. Yevdokimov Moscow State University of Medicine and Dentistry (MSUMD), Moscow, Russia
| | - Oleg Shatunov
- Laborant of the Labrotory of Histology and Immunohistochemistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Dmitrii Atiakshin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, RUDN University, Moscow, Russia
| |
Collapse
|
3
|
Li S, Feng W, Wu J, Cui H, Wang Y, Liang T, An J, Chen W, Guo Z, Lei H. A Narrative Review: Immunometabolic Interactions of Host-Gut Microbiota and Botanical Active Ingredients in Gastrointestinal Cancers. Int J Mol Sci 2024; 25:9096. [PMID: 39201782 PMCID: PMC11354385 DOI: 10.3390/ijms25169096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/03/2024] Open
Abstract
The gastrointestinal tract is where the majority of gut microbiota settles; therefore, the composition of the gut microbiota and the changes in metabolites, as well as their modulatory effects on the immune system, have a very important impact on the development of gastrointestinal diseases. The purpose of this article was to review the role of the gut microbiota in the host environment and immunometabolic system and to summarize the beneficial effects of botanical active ingredients on gastrointestinal cancer, so as to provide prospective insights for the prevention and treatment of gastrointestinal diseases. A literature search was performed on the PubMed database with the keywords "gastrointestinal cancer", "gut microbiota", "immunometabolism", "SCFAs", "bile acids", "polyamines", "tryptophan", "bacteriocins", "immune cells", "energy metabolism", "polyphenols", "polysaccharides", "alkaloids", and "triterpenes". The changes in the composition of the gut microbiota influenced gastrointestinal disorders, whereas their metabolites, such as SCFAs, bacteriocins, and botanical metabolites, could impede gastrointestinal cancers and polyamine-, tryptophan-, and bile acid-induced carcinogenic mechanisms. GPRCs, HDACs, FXRs, and AHRs were important receptor signals for the gut microbial metabolites in influencing the development of gastrointestinal cancer. Botanical active ingredients exerted positive effects on gastrointestinal cancer by influencing the composition of gut microbes and modulating immune metabolism. Gastrointestinal cancer could be ameliorated by altering the gut microbial environment, administering botanical active ingredients for treatment, and stimulating or blocking the immune metabolism signaling molecules. Despite extensive and growing research on the microbiota, it appeared to represent more of an indicator of the gut health status associated with adequate fiber intake than an autonomous causative factor in the prevention of gastrointestinal diseases. This study detailed the pathogenesis of gastrointestinal cancers and the botanical active ingredients used for their treatment in the hope of providing inspiration for research into simpler, safer, and more effective treatment pathways or therapeutic agents in the field.
Collapse
Affiliation(s)
- Shanlan Li
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China; (S.L.); (J.W.); (Y.W.); (T.L.); (J.A.); (W.C.); (Z.G.)
| | - Wuwen Feng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China;
| | - Jiaqi Wu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China; (S.L.); (J.W.); (Y.W.); (T.L.); (J.A.); (W.C.); (Z.G.)
| | - Herong Cui
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China; (S.L.); (J.W.); (Y.W.); (T.L.); (J.A.); (W.C.); (Z.G.)
| | - Yiting Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China; (S.L.); (J.W.); (Y.W.); (T.L.); (J.A.); (W.C.); (Z.G.)
| | - Tianzhen Liang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China; (S.L.); (J.W.); (Y.W.); (T.L.); (J.A.); (W.C.); (Z.G.)
| | - Jin An
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China; (S.L.); (J.W.); (Y.W.); (T.L.); (J.A.); (W.C.); (Z.G.)
| | - Wanling Chen
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China; (S.L.); (J.W.); (Y.W.); (T.L.); (J.A.); (W.C.); (Z.G.)
| | - Zhuoqian Guo
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China; (S.L.); (J.W.); (Y.W.); (T.L.); (J.A.); (W.C.); (Z.G.)
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 102488, China; (S.L.); (J.W.); (Y.W.); (T.L.); (J.A.); (W.C.); (Z.G.)
| |
Collapse
|
4
|
Zhao Z, He D, Wang J, Xiao Y, Gong L, Tang C, Peng H, Qiu X, Liu R, Zhang T, Li J. Swertiamarin relieves radiation-induced intestinal injury by limiting DNA damage. Mol Cell Biochem 2024:10.1007/s11010-024-05030-z. [PMID: 38795212 DOI: 10.1007/s11010-024-05030-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 05/04/2024] [Indexed: 05/27/2024]
Abstract
Radiotherapy is the conventional treatment for pelvic abdominal tumors. However, it can cause some damage to the small intestine and colorectal, which are very sensitive to radiation. Radiation-induced intestinal injury (RIII) affects the prognosis of radiotherapy, causing sequelae of loss of function and long-term damage to patients' quality of life. Swertiamarin is a glycoside that has been reported to prevent a variety of diseases including but not limited to diabetes, hypertension, atherosclerosis, arthritis, malaria, and abdominal ulcers. However, its therapeutic effect and mechanism of action on RIII have not been established. We investigated whether swertiamarin has a protective effect against RIII. In this article, we use irradiator to create cellular and mouse models of radiation damage. Preventive administration of swertiamarin could reduce ROS and superoxide anion levels to mitigate the cellular damage caused by radiation. Swertiamarin also attenuated RIII in mice, as evidenced by longer survival, less weight loss and more complete intestinal barrier. We also found an increase in the relative abundance of primary bile acids in irradiated mice, which was reduced by both FXR agonists and swertiamarin, and a reduction in downstream interferon and inflammatory factors via the cGAS-STING pathway to reduce radiation-induced damage.
Collapse
Affiliation(s)
- Zhe Zhao
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Dan He
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China
| | - Jinyu Wang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Yu Xiao
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China
| | - Lixin Gong
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China
| | - Can Tang
- School of Biological Science and Technology, Chengdu Medical College, Chengdu, China
| | - Haibo Peng
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xuemei Qiu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Tao Zhang
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China.
- School of Biological Science and Technology, Chengdu Medical College, Chengdu, China.
| | - Jingyi Li
- The Second Affiliated Hospital of Chengdu Medical College Nuclear Industry 416 Hospital, Chengdu, China.
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China.
| |
Collapse
|
5
|
McDonagh PR, Gobalakrishnan S, Rabender C, Vijayaragavan V, Zweit J. Molecular Imaging Investigations of Polymer-Coated Cerium Oxide Nanoparticles as a Radioprotective Therapeutic Candidate. Pharmaceutics 2023; 15:2144. [PMID: 37631358 PMCID: PMC10457862 DOI: 10.3390/pharmaceutics15082144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Cerium oxide nanoparticles (CONPs) have a unique surface redox chemistry that appears to selectively protect normal tissues from radiation induced damage. Our prior research exploring the biocompatibility of polymer-coated CONPs found further study of poly-acrylic acid (PAA)-coated CONPs was warranted due to improved systemic biodistribution and rapid renal clearance. This work further explores PAA-CONPs' radioprotective efficacy and mechanism of action related to tumor microenvironment pH. An ex vivo TUNEL assay was used to measure PAA-CONPs' protection of the irradiated mouse colon in comparison to the established radioprotector amifostine. [18F]FDG PET imaging of spontaneous colon tumors was utilized to determine the effects of PAA-CONPs on tumor radiation response. In vivo MRI and an ex vivo clonogenic assay were used to determine pH effects on PAA-CONPs' radioprotection in irradiated tumor-bearing mice. PAA-CONPs showed excellent radioprotective efficacy in the normal colon that was equivalent to uncoated CONPs and amifostine. [18F]FDG PET imaging showed PAA-CONPs do not affect tumor response to radiation. Normalization of tumor pH allowed some radioprotection of tumors by PAA-CONPs, which may explain their lack of tumor radioprotection in the acidic tumor microenvironment. Overall, PAA-CONPs meet the criteria for clinical application as a radioprotective therapeutic agent and are an excellent candidate for further study.
Collapse
Affiliation(s)
- Philip Reed McDonagh
- Department of Radiation Oncology, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
- Center for Molecular Imaging, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
| | - Sundaresan Gobalakrishnan
- Center for Molecular Imaging, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
- Department of Radiology, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
| | - Christopher Rabender
- Department of Radiation Oncology, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
| | - Vimalan Vijayaragavan
- Center for Molecular Imaging, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
- Department of Radiology, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
| | - Jamal Zweit
- Department of Radiation Oncology, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
- Center for Molecular Imaging, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
- Department of Radiology, Virginia Commonwealth University Health System, Richmond, VA 23219, USA
| |
Collapse
|
6
|
Devarakonda S, Thorsell A, Hedenström P, Rezapour A, Heden L, Banerjee S, Johansson MEV, Birchenough G, Toft Morén A, Gustavsson K, Skokic V, Pettersson VL, Sjöberg F, Kalm M, Al Masri M, Ekh M, Fagman H, Wolving M, Perkins R, Morales RA, Castillo F, Villablanca EJ, Yrlid U, Bergmark K, Steineck G, Bull C. Low-grade intestinal inflammation two decades after pelvic radiotherapy. EBioMedicine 2023; 94:104691. [PMID: 37480626 PMCID: PMC10393618 DOI: 10.1016/j.ebiom.2023.104691] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND Radiotherapy is effective in the treatment of cancer but also causes damage to non-cancerous tissue. Pelvic radiotherapy may produce chronic and debilitating bowel symptoms, yet the underlying pathophysiology is still undefined. Most notably, although pelvic radiotherapy causes an acute intestinal inflammation there is no consensus on whether the late-phase pathophysiology contains an inflammatory component or not. To address this knowledge gap, we examined the potential presence of a chronic inflammation in mucosal biopsies from irradiated pelvic cancer survivors. METHODS We biopsied 24 cancer survivors two to 20 years after pelvic radiotherapy, and four non-irradiated controls. Using tandem mass tag (TMT) mass spectrometry and mRNA sequencing (mRNA-seq), we charted proteomic and transcriptomic profiles of the mucosal tissue previously exposed to a high or a low/no dose of radiation. Changes in the immune cell populations were determined with flow cytometry. The integrity of the protective mucus layers were determined by permeability analysis and 16S rRNA bacterial detection. FINDINGS 942 proteins were differentially expressed in mucosa previously exposed to a high radiation dose compared to a low radiation dose. The data suggested a chronic low-grade inflammation with neutrophil activity, which was confirmed by mRNA-seq and flow cytometry and further supported by findings of a weakened mucus barrier with bacterial infiltration. INTERPRETATION Our results challenge the idea that pelvic radiotherapy causes an acute intestinal inflammation that either heals or turns fibrotic without progression to chronic inflammation. This provides a rationale for exploring novel strategies to mitigate chronic bowel symptoms in pelvic cancer survivors. FUNDING This study was supported by the King Gustav V Jubilee Clinic Cancer Foundation (CB), The Adlerbertska Research Foundation (CB), The Swedish Cancer Society (GS), The Swedish State under the ALF agreement (GS and CB), Mary von Sydow's foundation (MA and VP).
Collapse
Affiliation(s)
- Sravani Devarakonda
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Annika Thorsell
- Proteomics Core Facility, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Hedenström
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Gastroenterology and Hepatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Azar Rezapour
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lisen Heden
- Pelvic Cancer Rehabilitation, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sanghita Banerjee
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Malin E V Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - George Birchenough
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Amelie Toft Morén
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Gustavsson
- Pelvic Cancer Rehabilitation, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Viktor Skokic
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Molecular Medicine and Surgery and Department of Pelvic Cancer, Karolinska Institute, Stockholm, Sweden
| | - Victor L Pettersson
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fei Sjöberg
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Marie Kalm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Mohammad Al Masri
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Michaela Ekh
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Fagman
- Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg and Department of Clinical Patology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mats Wolving
- Department of Pathology, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rodrigo A Morales
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Francisca Castillo
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Eduardo J Villablanca
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Ulf Yrlid
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karin Bergmark
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar Steineck
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Bull
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
7
|
Wang X, Yuan R, Miao L, Li X, Guo Y, Tian H. Protective mechanism of a novel aminothiol compound on radiation-induced intestinal injury. Int J Radiat Biol 2023; 99:259-269. [PMID: 35583501 DOI: 10.1080/09553002.2022.2074163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE With the development of nuclear technology and radiotherapy, the risk of radiation injury has been increasing. Therefore, it is important to find an effective radiation-protective agent. In this study, we designed and synthesized a novel compound called compound 8, of which the radioprotective effect and mechanism were studied. MATERIALS AND METHODS Before being exposed to ionizing radiation, mice were pretreated with compound 8. The 30-day mortality assay, hematoxylin-eosin staining, and immunohistochemistry staining assay were performed to evaluate the anti-radiation effect of the compound 8. TUNEL and immunofluorescence assays were conducted to study the anti-radiation mechanism of compound 8. RESULTS Compared to the IR + vehicle group, the 30-day survival rate of mice treated with 25 mg/kg of compound 8 was significantly improved after 8 Gy total body irradiation. In the morphological study of the small intestine, we found that compound 8 could maintain crypt-villus structures in the irradiated mice. Further immunohistochemical staining displayed that compound 8 could improve the survival of Lgr5+ cells, ki67+ cells, and lysozyme+ cells. The results of TUNEL and immunofluorescence assays showed that compound 8 could decrease the expression of apoptosis-related caspase-8/-9, γ-H2AX, Bax, and p53. CONCLUSIONS These results indicate that compound 8 exerts its effects by maintaining structure and function of small intestine. It also reduces DNA damage, promotes crypt proliferation and differentiation. Moreover, it may enhance the anti-apoptotic ability of small intestinal tissue by inhibiting the activation of p53 and blocking the caspase cascade reaction. Compound 8 can protect the intestinal tract from post-radiation damage, it is thus a new and effective protective agent of radiation.
Collapse
Affiliation(s)
- Xinxin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Renbin Yuan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Longfei Miao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Xuejiao Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Yuying Guo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| |
Collapse
|
8
|
Livanova AA, Fedorova AA, Zavirsky AV, Krivoi II, Markov AG. Dose- and Segment-Dependent Disturbance of Rat Gut by Ionizing Radiation: Impact of Tight Junction Proteins. Int J Mol Sci 2023; 24:ijms24021753. [PMID: 36675266 PMCID: PMC9863103 DOI: 10.3390/ijms24021753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/30/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
The damaging effect of ionizing radiation (IR) exposure results in the disturbance of the gut natural barrier, followed by the development of severe gastrointestinal injury. However, the dose and application segment are known to determine the effects of IR. In this study, we demonstrated the dose- and segment-specificity of tight junction (TJ) alteration in IR-induced gastrointestinal injury in rats. Male Wistar rats were subjected to a total-body X-ray irradiation at doses of 2 or 10 Gy. Isolated jejunum and colon segments were tested in an Ussing chamber 72 h after exposure. In the jejunum, 10-Gy IR dramatically altered transepithelial resistance, short-circuit current and permeability for sodium fluorescein. These changes were accompanied by severe disturbance of histological structure and total rearrangement of TJ content (increased content of claudin-1, -2, -3 and -4; multidirectional changes in tricellulin and occludin). In the colon of 10-Gy irradiated rats, lesions of barrier and transport functions were less pronounced, with only claudin-2 and -4 altered among TJ proteins. The 2-Gy IR did not change electrophysiological characteristics or permeability in the colon or jejunum, although slight alterations in jejunum histology were noted, emphasized with claudin-3 increase. Considering that TJ proteins are critical for maintaining epithelial barrier integrity, these findings may have implications for countermeasures in gastrointestinal acute radiation injury.
Collapse
Affiliation(s)
- Alexandra A. Livanova
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Arina A. Fedorova
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Alexander V. Zavirsky
- Department of Military Toxicology and Radiation Defense, S. M. Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - Igor I. Krivoi
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Alexander G. Markov
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia
- Correspondence:
| |
Collapse
|
9
|
Protective Effect of Bojungikki-Tang against Radiation-Induced Intestinal Injury in Mice: Experimental Verification and Compound-Target Prediction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:5417813. [PMID: 36644439 PMCID: PMC9833920 DOI: 10.1155/2023/5417813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023]
Abstract
Bojungikki-tang (BJIT) is a traditional herbal medicine used in Korea, Japan, and China to treat gastrointestinal disorders. In this study, we aimed to investigate whether BJIT has protective effects against radiation-induced intestinal injury and to predict the underlying therapeutic mechanisms and related pathways via network pharmacological analyses. BJIT was injected intraperitoneally (50 mg/kg body weight) to C3H/HeN mice at 36 and 12 h before exposure to partial abdominal irradiation (5 Gy and 13 Gy) to evaluate the apoptotic changes and the histological changes and variations in inflammatory cytokine mRNA levels in the jejunum, respectively. Through in silico network analysis, we predicted the mechanisms underlying BJIT-mediated regulation of radiation-induced intestinal injury. BJIT reduced the level of apoptosis in the jejunal crypts 12 h post 5-Gy irradiation. Histological assessment revealed intestinal morphological changes in irradiated mice 3.5 days post 13-Gy irradiation. Furthermore, BJIT decreased inflammatory cytokine levels following radiation exposure. Apoptosis, TNF, p53, VEGF, toll-like receptor, PPAR, PI3K-Akt, and MAPK signaling pathways, as well as inflammatory bowel disease (IBD), were found to be linked to the radioprotective effects of BJIT against intestinal injury. According to our results, BJIT exerted its potential protective effects by attenuating histopathological changes in jejunal crypts and suppressing inflammatory mediator levels. Therefore, BJIT is a potential therapeutic agent that can treat radiation-induced intestinal injury and its associated symptoms.
Collapse
|
10
|
Sminia P, Guipaud O, Viktorsson K, Ahire V, Baatout S, Boterberg T, Cizkova J, Dostál M, Fernandez-Palomo C, Filipova A, François A, Geiger M, Hunter A, Jassim H, Edin NFJ, Jordan K, Koniarová I, Selvaraj VK, Meade AD, Milliat F, Montoro A, Politis C, Savu D, Sémont A, Tichy A, Válek V, Vogin G. Clinical Radiobiology for Radiation Oncology. RADIOBIOLOGY TEXTBOOK 2023:237-309. [DOI: 10.1007/978-3-031-18810-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
AbstractThis chapter is focused on radiobiological aspects at the molecular, cellular, and tissue level which are relevant for the clinical use of ionizing radiation (IR) in cancer therapy. For radiation oncology, it is critical to find a balance, i.e., the therapeutic window, between the probability of tumor control and the probability of side effects caused by radiation injury to the healthy tissues and organs. An overview is given about modern precision radiotherapy (RT) techniques, which allow optimal sparing of healthy tissues. Biological factors determining the width of the therapeutic window are explained. The role of the six typical radiobiological phenomena determining the response of both malignant and normal tissues in the clinic, the 6R’s, which are Reoxygenation, Redistribution, Repopulation, Repair, Radiosensitivity, and Reactivation of the immune system, is discussed. Information is provided on tumor characteristics, for example, tumor type, growth kinetics, hypoxia, aberrant molecular signaling pathways, cancer stem cells and their impact on the response to RT. The role of the tumor microenvironment and microbiota is described and the effects of radiation on the immune system including the abscopal effect phenomenon are outlined. A summary is given on tumor diagnosis, response prediction via biomarkers, genetics, and radiomics, and ways to selectively enhance the RT response in tumors. Furthermore, we describe acute and late normal tissue reactions following exposure to radiation: cellular aspects, tissue kinetics, latency periods, permanent or transient injury, and histopathology. Details are also given on the differential effect on tumor and late responding healthy tissues following fractionated and low dose rate irradiation as well as the effect of whole-body exposure.
Collapse
|
11
|
Saldi S, Fulcheri CPL, Zucchetti C, Abdelhamid AMH, Carotti A, Pierini A, Ruggeri L, Tricarico S, Chiodi M, Ingrosso G, Bini V, Velardi A, Martelli MF, Hui SK, Aristei C. Impact of total marrow/lymphoid irradiation dose to the intestine on graft-versus-host disease in allogeneic hematopoietic stem cell transplantation for hematologic malignancies. Front Oncol 2022; 12:1035375. [PMID: 36568236 PMCID: PMC9773831 DOI: 10.3389/fonc.2022.1035375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022] Open
Abstract
Background and purpose Graft-versus-host disease (GvHD) is a leading cause of non-relapse mortality in patients undergoing allogeneic hematopoietic stem cell transplantation. The Perugia Bone Marrow Transplantation Unit designed a new conditioning regimen with total marrow/lymphoid irradiation (TMLI) and adaptive immunotherapy. The present study investigated the impact of radiotherapy (RT) doses on the intestine on the incidence of acute GvHD (aGvHD) in transplant recipients, analyzing the main dosimetric parameters. Materials and methods Between August 2015 and April 2021, 50 patients with hematologic malignancies were enrolled. All patients underwent conditioning with TMLI. Dosimetric parameters (for the whole intestine and its segments) were assessed as risk factors for aGvHD. The RT dose that was received by each intestinal area with aGvHD was extrapolated from the treatment plan for each patient. Doses were compared with those of the whole intestine minus the affected area. Results Eighteen patients (36%) developed grade ≥2 aGvHD (G2 in 5, G3 in 11, and G4 in 2). Median time to onset was 41 days (range 23-69 days). The skin was involved in 11 patients, the intestine in 16, and the liver in 5. In all 50 TMLI patients, the mean dose to the whole intestine was 7.1 Gy (range 5.07-10.92 Gy). No patient developed chronic GvHD (cGvHD). No dosimetric variable emerged as a significant risk factor for aGvHD. No dosimetric parameter of the intestinal areas with aGvHD was associated with the disease. Conclusion In our clinical setting and data sample, we have found no clear evidence that current TMLI dosages to the intestine were linked to the development of aGvHD. However, due to some study limitations, this investigation should be considered as a preliminary assessment. Findings need to be confirmed in a larger cohort and in preclinical models.
Collapse
Affiliation(s)
- Simonetta Saldi
- Section of Radiation Oncology, Hospital of Santa Maria della Misericordia, Perugia, Italy
| | | | - Claudio Zucchetti
- Medical Physics, Hospital of Santa Maria della Misericordia, Perugia, Italy
| | - Amr Mohamed Hamed Abdelhamid
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia and Perugia General Hospital, Perugia, Italy,Department of Oncology and Nuclear Medicine, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Alessandra Carotti
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Antonio Pierini
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Loredana Ruggeri
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Sara Tricarico
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Marino Chiodi
- Radiology Unit, S. Maria Della Misericordia Hospital, Perugia, Italy
| | - Gianluca Ingrosso
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia and Perugia General Hospital, Perugia, Italy
| | - Vittorio Bini
- Internal Medicine, Endocrine and Metabolic Science Section, University of Perugia, Perugia, Italy
| | - Andrea Velardi
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Massimo Fabrizio Martelli
- Division of Hematology and Clinical Immunology, Department of Medicine, University of Perugia, Perugia, Italy
| | - Susanta Kumar Hui
- Department of Radiation Oncology, City of Hope National Medical Center, CA, United States
| | - Cynthia Aristei
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia and Perugia General Hospital, Perugia, Italy,*Correspondence: Cynthia Aristei,
| |
Collapse
|
12
|
Elmas O, Sahin HHK, Keskin E, Guven B, Uslu Erdemir R, ALMisned G, Zakaly HMH, Ene A, Tekin HO. Clinical comprehensive and experimental assessment of the radioprotective effect of Annona muricata leaf extract to prevent cellular damage in the ileum tissue. OPEN CHEM 2022. [DOI: 10.1515/chem-2022-0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
We report the radioprotective attitude of Annona muricata (AM) leaf extract as antioxidant material to prevent cellular damage in the ileum tissue. The protective effects of an ethyl acetate extract of AM leaves are comprehensively investigated against radiation-induced ileal damage in numerous rats. Thirty-two adult female rats were separated into 4 groups (3 intervention groups and 1 control) as follows: controls received 0.01 mL/kg distilled water, the AM group received 300 mg/kg AM leaf extract, the ionizing radiation (IR) group received a single dose of whole body radiation (8.3 Gy) after 0.01 mL/kg saline treatment, and the AM + IR group received 300 mg/kg AM leaf extract treatment and were subjected to whole body radiation (8.3 Gy) 1 h after the last gavage. All treatments are administered by oral gavage once a day for 9 days. At the end of the experiment, biochemical total oxidant status (TOS, interleukin-6, and caspase) and histological examinations are performed on blood samples as well as ileum tissue. TOS levels are found to be significantly high in rats, which received irradiation, and those in the AM group when compared to controls. These findings suggest that AM has radioprotective effects on ileum tissue, likely because of its antioxidative properties. The findings of this research may contribute to the minimizing of major side effects induced by excessive radiation exposure in patients undergoing radiotherapy and may serve as a significant impetus for further assessments. However, future studies are highly recommended to confirm safety and to determine extraction technique and dosage before human use can be considered.
Collapse
Affiliation(s)
- Ozlem Elmas
- Department of Radiation Oncology, Bulent Ecevit University Practice and Research Hospital , Zonguldak , Turkey
| | - Havva Hande Keser Sahin
- Department of Pathology, Hitit University Corum Training and Research Hospital , Corum , Turkey
| | - Emrah Keskin
- Department of Neurosurgery, Bulent Ecevit University Practice and Research Hospital , Zonguldak , Turkey
| | - Berrak Guven
- Department of Biochemistry, Bulent Ecevit University Practice and Research Hospital , Zonguldak , Turkey
| | - Rabiye Uslu Erdemir
- Department of Nuclear Medicine, Zonguldak Bülent Ecevit University, Faculty of Medicine , Zonguldak , Turkey
| | - Ghada ALMisned
- Department of Physics, College of Science, Princess Nourah Bint Abdulrahman University , P.O. Box 84428 , Riyadh 11671 , Saudi Arabia
| | - Hesham M. H. Zakaly
- Institute of Physics and Technology, Ural Federal University , 620002 Ekaterinburg , Russia
- Physics Department, Faculty of Science, Al-Azhar University , Assiut 71524 , Egypt
| | - Antoaneta Ene
- Department of Chemistry, INPOLDE Research Center, Physics and Environment, Faculty of Sciences and Environment, Dunarea de Jos University of Galati , 47 Domneasca Street , 800008 Galati , Romania
| | - Huseyin Ozan Tekin
- Department of Medical Diagnostic Imaging, College of Health Sciences, University of Sharjah , 27272 , Sharjah , United Arab Emirates
- Computer Engineering Department, Faculty of Engineering and Natural Sciences, Istinye University , Istanbul 34396 , Turkey
| |
Collapse
|
13
|
Pejchal J, Tichy A, Kmochova A, Fikejzlova L, Kubelkova K, Milanova M, Lierova A, Filipova A, Muckova L, Cizkova J. Mitigation of Ionizing Radiation-Induced Gastrointestinal Damage by Insulin-Like Growth Factor-1 in Mice. Front Pharmacol 2022; 13:663855. [PMID: 35847048 PMCID: PMC9277384 DOI: 10.3389/fphar.2022.663855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose: Insulin-like growth factor-1 (IGF-1) stimulates epithelial regeneration but may also induce life-threatening hypoglycemia. In our study, we first assessed its safety. Subsequently, we examined the effect of IGF-1 administered in different dose regimens on gastrointestinal damage induced by high doses of gamma radiation. Material and methods: First, fasting C57BL/6 mice were injected subcutaneously with IGF-1 at a single dose of 0, 0.2, 1, and 2 mg/kg to determine the maximum tolerated dose (MTD). The glycemic effect of MTD (1 mg/kg) was additionally tested in non-fasting animals. Subsequently, a survival experiment was performed. Animals were irradiated (60Co; 14, 14.5, or 15 Gy; shielded head), and IGF-1 was administered subcutaneously at 1 mg/kg 1, 24, and 48 h after irradiation. Simultaneously, mice were irradiated (60Co; 12, 14, or 15 Gy; shielded head), and IGF-1 was administered subcutaneously under the same regimen. Jejunum and lung damage were assessed 84 h after irradiation. Finally, we evaluated the effect of six different IGF-1 dosage regimens administered subcutaneously on gastrointestinal damage and peripheral blood changes in mice 6 days after irradiation (60Co; 12 and 14 Gy; shielded head). The regimens differed in the number of doses (one to five doses) and the onset of administration (starting at 1 [five regimens] or 24 h [one regimen] after irradiation). Results: MTD was established at 1 mg/kg. MTD mitigated lethality induced by 14 Gy and reduced jejunum and lung damage caused by 12 and 14 Gy. However, different dosing regimens showed different efficacy, with three and four doses (administered 1, 24, and 48 h and 1, 24, 48, and 72 h after irradiation, respectively) being the most effective. The three-dose regimens supported intestinal regeneration even if the administration started at 24 h after irradiation, but its potency decreased. Conclusion: IGF-1 seems promising in the mitigation of high-dose irradiation damage. However, the selected dosage regimen affects its efficacy.
Collapse
Affiliation(s)
- Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| | - Ales Tichy
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| | - Adela Kmochova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| | - Lenka Fikejzlova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| | - Klara Kubelkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| | - Marcela Milanova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| | - Anna Lierova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| | - Alzbeta Filipova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| | - Lubica Muckova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| | - Jana Cizkova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Brno, Czechia
| |
Collapse
|
14
|
Xi C, Zhao H, Liu HX, Xiang JQ, Lu X, Cai TJ, Li S, Gao L, Tian XL, Liu KH, Tian M, Liu QJ. Screening of radiation gastrointestinal injury biomarkers in rat plasma by high-coverage targeted lipidomics. Biomarkers 2022; 27:448-460. [PMID: 35315697 DOI: 10.1080/1354750x.2022.2056920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
INTRODUCTION In the event of radiological accidents and cancer radiotherapies in clinic, the gastrointestinal (GI) system is vulnerable to ionizing radiation and shows GI injury. Accessible biomarkers may provide means to predict, evaluate, and treat GI tissue damage. The current study investigated radiation GI injury biomarkers in rat plasma. MATERIAL AND METHODS High-coverage targeted lipidomics was employed to profile lipidome perturbations at 72 h after 0, 1, 2, 3, 5 and 8 Gy (60Co γ-rays at 1 Gy/min) total-body irradiation in male rat jejunum. The results were correlated with previous plasma screening outcomes. RESULTS In total, 93 differential metabolites and 28 linear dose-responsive metabolites were screened in the jejunum. Moreover, 52 lipid species with significant differences both in jejunum and plasma were obtained. Three lipid species with linear dose-response relationship both in jejunum and plasma were put forth, which exhibited good to excellent sensitivity and specificity in triaging different exposure levels. DISCUSSION The linear dose-effect relationship of lipid metabolites in the jejunum and the triage performance of radiation GI injury biomarkers in plasma were studied for the first time. CONCLUSION The present study can provide insights into expanded biomarkers of IR-mediated GI injury and minimally invasive assays for evaluation.
Collapse
Affiliation(s)
- Cong Xi
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hua Zhao
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hai-Xiang Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jia-Qi Xiang
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xue Lu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Tian-Jing Cai
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shuang Li
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ling Gao
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xue-Lei Tian
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ke-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mei Tian
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qing-Jie Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
15
|
Fujimichi Y, Otsuka K, Tomita M, Iwasaki T. Ionizing radiation alters organoid forming potential and replenishment rate in a dose/dose-rate dependent manner. JOURNAL OF RADIATION RESEARCH 2022; 63:166-173. [PMID: 34977948 PMCID: PMC8944312 DOI: 10.1093/jrr/rrab120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/21/2021] [Indexed: 06/14/2023]
Abstract
Intestinal organoids are an in vitro cultured tissue model generated from intestinal stem cells, and they contain a mixture of epithelial cell types. We previously established an efficient 'one cell/well' sorting method, and defined organoid-forming potential (OFP) as a useful index to evaluate the stemness of individual cells. In this study, we assessed the response to radiation dose and dose-rate by measuring both OFP and the percentage of stem cells in the crypts. After high-dose-rate (HDR, 0.5 Gy/min) irradiation in vivo, the percentage of stem cells in the harvested crypt cells decreased, and the replenishment of cycling stem cells originating from dormant cells was enhanced, but OFP increased in cells irradiated with a total dose of >1 Gy. In contrast, at a total dose of 0.1 Gy the percentage of stem cells reduced slightly, but neither replenishment rate nor OFP changed. Furthermore, the response to 1 Gy of low-dose-rate (LDR) irradiation was similar to the response to 0.1 Gy HDR irradiation. These results suggest that 0.1 Gy HDR irradiation or 1 Gy LDR irradiation does not alter stemness. Additionally, the OFP increase in the colon in response to irradiation was smaller than that in the duodenum, similar to the percentage of stem cells. Understanding the differences in the response of stem cells between the colon and the duodenum to radiation is important to clarify the mechanisms underlying the development of radiation-associated intestinal cancers.
Collapse
Affiliation(s)
- Yuki Fujimichi
- Corresponding Author. Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry, 2-11-1 Iwado kita, Komae-shi, Tokyo 201-8511 Japan, Tel: +81-70-5457-2909 Fax: +81-3-3480-3113,
| | - Kensuke Otsuka
- Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry, 2-11-1 Iwado kita, Komae-shi, Tokyo 201-8511 Japan
| | - Masanori Tomita
- Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry, 2-11-1 Iwado kita, Komae-shi, Tokyo 201-8511 Japan
| | - Toshiyasu Iwasaki
- Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry, 2-11-1 Iwado kita, Komae-shi, Tokyo 201-8511 Japan
| |
Collapse
|
16
|
Malipatlolla DK, Devarakonda S, Patel P, Sjöberg F, Rascón A, Grandér R, Skokic V, Kalm M, Danial J, Mehdin E, Warholm M, Norling H, Stringer A, Johansson MEV, Nyman M, Steineck G, Bull C. A Fiber-Rich Diet and Radiation-Induced Injury in the Murine Intestinal Mucosa. Int J Mol Sci 2021; 23:439. [PMID: 35008864 PMCID: PMC8745769 DOI: 10.3390/ijms23010439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Dietary fiber is considered a strong intestinal protector, but we do not know whether dietary fiber protects against the long-lasting mucosal damage caused by ionizing radiation. To evaluate whether a fiber-rich diet can ameliorate the long-lasting pathophysiological hallmarks of the irradiated mucosa, C57BL/6J mice on a fiber-rich bioprocessed oat bran diet or a fiber-free diet received 32 Gray in four fractions to the distal colorectum using a linear accelerator and continued on the diets for one, six or 18 weeks. We quantified degenerating crypts, crypt fission, cell proliferation, crypt survival, macrophage density and bacterial infiltration. Crypt loss through crypt degeneration only occurred in the irradiated mice. Initially, it was most frequent in the fiber-deprived group but declined to levels similar to the fiber-consuming group by 18 weeks. The fiber-consuming group had a fast response to irradiation, with crypt fission for growth or healing peaking already at one week post-irradiation, while crypt fission in the fiber-deprived group peaked at six weeks. A fiber-rich diet allowed for a more intense crypt cell proliferation, but the recovery of crypts was eventually lost by 18 weeks. Bacterial infiltration was a late phenomenon, evident in the fiber-deprived animals and intensified manyfold after irradiation. Bacterial infiltration also coincided with a specific pro-inflammatory serum cytokine profile. In contrast, mice on a fiber-rich diet were completely protected from irradiation-induced bacterial infiltration and exhibited a similar serum cytokine profile as sham-irradiated mice on a fiber-rich diet. Our findings provide ample evidence that dietary fiber consumption modifies the onset, timing and intensity of radiation-induced pathophysiological processes in the intestinal mucosa. However, we need more knowledge, not least from clinical studies, before this finding can be introduced to a new and refined clinical practice.
Collapse
Affiliation(s)
- Dilip Kumar Malipatlolla
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| | - Sravani Devarakonda
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| | - Piyush Patel
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
- Department of Infectious Diseases at the Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Fei Sjöberg
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
- Department of Infectious Diseases at the Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
| | - Ana Rascón
- Department of Food Technology, Engineering and Nutrition, Lund University, 221 00 Lund, Sweden; (A.R.); (M.N.)
| | - Rita Grandér
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| | - Viktor Skokic
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| | - Marie Kalm
- Department of Pharmacology at the Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden;
| | - Jolie Danial
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| | - Eva Mehdin
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| | - Malin Warholm
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| | - Henrietta Norling
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| | - Andrea Stringer
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Malin E. V. Johansson
- Department of Medical Biochemistry and Cell Biology at the Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden;
| | - Margareta Nyman
- Department of Food Technology, Engineering and Nutrition, Lund University, 221 00 Lund, Sweden; (A.R.); (M.N.)
| | - Gunnar Steineck
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| | - Cecilia Bull
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden; (D.K.M.); (S.D.); (P.P.); (F.S.); (R.G.); (V.S.); (J.D.); (E.M.); (M.W.); (H.N.); (G.S.)
| |
Collapse
|
17
|
Tang H, Wang J, Luo X, Wang Q, Chen J, Zhang X, Li Q, Gao C, Li Y, Han S. An Apoptosis-Related Gene Prognostic Index for Colon Cancer. Front Cell Dev Biol 2021; 9:790878. [PMID: 34957118 PMCID: PMC8692577 DOI: 10.3389/fcell.2021.790878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/11/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose: To construct an apoptosis-related gene prognostic index (ARGPI) for colon cancer, and clarify the molecular and immune characteristics of the risk subgroup as defined by the prognostic index and the benefits of adjuvant chemotherapy. Integrating the prognostic index and clinicopathological risk factors to better evaluate the prognosis of patients with colon cancer. Methods: Based on the colon adenocarcinoma data in the TCGA database, 20 apoptosis-related hub genes were screened by weighted gene co-expression network analysis (WGCNA). Five genes constituting the prognosis model were determined by Cox regression and verified by the Gene Expression Omnibus (GEO) dataset. Then the molecular and immune characteristics of risk subgroups defined by the prognostic index and the benefits of adjuvant chemotherapy were analyzed. Finally, nomograms integrating ARGPI and four clinicopathological risk factors were used to evaluate the prognosis of patients with colon cancer. Results: The ARGPI was constructed based on the FAS, VWA5A, SPTBN2, PCK1, and TIMP1 genes. In the TCGA cohort, patients in the low-risk subgroup had a longer progression-free interval (PFI) than patients in the high-risk subgroup, which coincided with the results of the GEO cohort. The comprehensive results showed that the high-risk score was related to the enrichment of the cell cycle pathway, high mutation rate of TP53 and KRAS, high infiltration of T regulatory cells (Tregs), immunosuppressive state, and less chemotherapeutic benefit. However, low-risk scores are related to drug metabolism-related pathways, low TP53 and KRAS mutation rates, high infiltration of plasma cells, more resting CD4 memory cells and eosinophils, active immune function, and better chemotherapeutic benefits. Receiver operating characteristic curve of two-year progress prediction evaluation showed that the ARGPI had higher prognostic accuracy than TNM staging. Nomograms integrating ARGPI and clinicopathological risk factors can better evaluate the prognosis of patients with colon cancer. Conclusions: The ARGPI is a promising biomarker for determining risk of colon cancer progression, molecular and immune characteristics, and chemotherapeutic benefit. This is a reliable method to predict the prognosis of colon cancer patients. It also can assist doctors in formulating more effective treatment strategies.
Collapse
Affiliation(s)
- Hanmin Tang
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Wang
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xuehui Luo
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qi Wang
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jie Chen
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xinyue Zhang
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiuting Li
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chengyi Gao
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuesen Li
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Suxia Han
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
18
|
Leibowitz BJ, Zhao G, Wei L, Ruan H, Epperly M, Chen L, Lu X, Greenberger JS, Zhang L, Yu J. Interferon b drives intestinal regeneration after radiation. SCIENCE ADVANCES 2021; 7:eabi5253. [PMID: 34613772 PMCID: PMC8494436 DOI: 10.1126/sciadv.abi5253] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/16/2021] [Indexed: 05/14/2023]
Abstract
The cGAS-STING cytosolic DNA sensing pathway is critical for host defense. Here, we report that cGAS-STING–dependent type 1 interferon (IFN) response drives intestinal regeneration and animal recovery from radiation injury. STING deficiency has no effect on radiation-induced DNA damage or crypt apoptosis but abrogates epithelial IFN-β production, local inflammation, innate transcriptional response, and subsequent crypt regeneration. cGAS KO, IFNAR1 KO, or CCR2 KO also abrogates radiation-induced acute crypt inflammation and regeneration. Impaired intestinal regeneration and survival in STING-deficient mice are fully rescued by a single IFN-β treatment given 48 hours after irradiation but not by wild-type (WT) bone marrow. IFN-β treatment remarkably improves the survival of WT mice and Lgr5+ stem cell regeneration through elevated compensatory proliferation and more rapid DNA damage removal. Our findings support that inducible IFN-β production in the niche couples ISC injury and regeneration and its potential use to treat acute radiation injury.
Collapse
Affiliation(s)
- Brian J. Leibowitz
- Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Guangyi Zhao
- Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Liang Wei
- Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Hang Ruan
- Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Michael Epperly
- Department of Radiation Oncology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Lujia Chen
- Department of Medical Informatics, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Xinghua Lu
- Department of Medical Informatics, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Joel S. Greenberger
- Department of Radiation Oncology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Lin Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Jian Yu
- Department of Pathology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Radiation Oncology, University of Pittsburgh School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
19
|
Guillermin O, Angelis N, Sidor CM, Ridgway R, Baulies A, Kucharska A, Antas P, Rose MR, Cordero J, Sansom O, Li VSW, Thompson BJ. Wnt and Src signals converge on YAP-TEAD to drive intestinal regeneration. EMBO J 2021; 40:e105770. [PMID: 33950519 PMCID: PMC8246259 DOI: 10.15252/embj.2020105770] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Wnt signalling induces a gradient of stem/progenitor cell proliferation along the crypt-villus axis of the intestine, which becomes expanded during intestinal regeneration or tumour formation. The YAP transcriptional co-activator is known to be required for intestinal regeneration, but its mode of regulation remains controversial. Here we show that the YAP-TEAD transcription factor is a key downstream effector of Wnt signalling in the intestine. Loss of YAP activity by Yap/Taz conditional knockout results in sensitivity of crypt stem cells to apoptosis and reduced cell proliferation during regeneration. Gain of YAP activity by Lats1/2 conditional knockout is sufficient to drive a crypt hyperproliferation response. In particular, Wnt signalling acts transcriptionally to induce YAP and TEAD1/2/4 expression. YAP normally localises to the nucleus only in crypt base stem cells, but becomes nuclear in most intestinal epithelial cells during intestinal regeneration after irradiation, or during organoid growth, in a Src family kinase-dependent manner. YAP-driven crypt expansion during regeneration involves an elongation and flattening of the Wnt signalling gradient. Thus, Wnt and Src-YAP signals cooperate to drive intestinal regeneration.
Collapse
Affiliation(s)
- Oriane Guillermin
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUK
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Nikolaos Angelis
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Clara M Sidor
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Rachel Ridgway
- Colorectal Cancer and Wnt signalling LaboratoryCancer Research UK Beatson InstituteGlasgowUK
| | - Anna Baulies
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Anna Kucharska
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Pedro Antas
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Melissa R Rose
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Julia Cordero
- Institute of Cancer SciencesWolfson Wohl Cancer Research CentreBearsdenUK
| | - Owen Sansom
- Colorectal Cancer and Wnt signalling LaboratoryCancer Research UK Beatson InstituteGlasgowUK
| | - Vivian S W Li
- Stem Cell and Cancer Biology LaboratoryFrancis Crick InstituteLondonUK
| | - Barry J Thompson
- Epithelial Biology LaboratoryFrancis Crick InstituteLondonUK
- EMBL Australia ACRF Department of Cancer Biology & TherapeuticsJohn Curtin School of Medical ResearchThe Australian National UniversityActonACTAustralia
| |
Collapse
|
20
|
Sprangers J, Zaalberg IC, Maurice MM. Organoid-based modeling of intestinal development, regeneration, and repair. Cell Death Differ 2021; 28:95-107. [PMID: 33208888 PMCID: PMC7852609 DOI: 10.1038/s41418-020-00665-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
The intestinal epithelium harbors a remarkable adaptability to undergo injury-induced repair. A key part of the regenerative response is the transient reprogramming of epithelial cells into a fetal-like state, which drives uniform proliferation, tissue remodeling, and subsequent restoration of the homeostatic state. In this review, we discuss how Wnt and YAP signaling pathways control the intestinal repair response and the transitioning of cell states, in comparison with the process of intestinal development. Furthermore, we highlight how organoid-based applications have contributed to the characterization of the mechanistic principles and key players that guide these developmental and regenerative events.
Collapse
Affiliation(s)
- Joep Sprangers
- Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Irene C Zaalberg
- Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Madelon M Maurice
- Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
21
|
Prasad B, Grimm D, Strauch SM, Erzinger GS, Corydon TJ, Lebert M, Magnusson NE, Infanger M, Richter P, Krüger M. Influence of Microgravity on Apoptosis in Cells, Tissues, and Other Systems In Vivo and In Vitro. Int J Mol Sci 2020; 21:E9373. [PMID: 33317046 PMCID: PMC7764784 DOI: 10.3390/ijms21249373] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023] Open
Abstract
All life forms have evolved under the constant force of gravity on Earth and developed ways to counterbalance acceleration load. In space, shear forces, buoyance-driven convection, and hydrostatic pressure are nullified or strongly reduced. When subjected to microgravity in space, the equilibrium between cell architecture and the external force is disturbed, resulting in changes at the cellular and sub-cellular levels (e.g., cytoskeleton, signal transduction, membrane permeability, etc.). Cosmic radiation also poses great health risks to astronauts because it has high linear energy transfer values that evoke complex DNA and other cellular damage. Space environmental conditions have been shown to influence apoptosis in various cell types. Apoptosis has important functions in morphogenesis, organ development, and wound healing. This review provides an overview of microgravity research platforms and apoptosis. The sections summarize the current knowledge of the impact of microgravity and cosmic radiation on cells with respect to apoptosis. Apoptosis-related microgravity experiments conducted with different mammalian model systems are presented. Recent findings in cells of the immune system, cardiovascular system, brain, eyes, cartilage, bone, gastrointestinal tract, liver, and pancreas, as well as cancer cells investigated under real and simulated microgravity conditions, are discussed. This comprehensive review indicates the potential of the space environment in biomedical research.
Collapse
Affiliation(s)
- Binod Prasad
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, Staudtstraße 5, 91058 Erlangen, Germany; (B.P.); (M.L.)
| | - Daniela Grimm
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus C, Denmark; (D.G.); (T.J.C.)
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.I.); (M.K.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Sebastian M. Strauch
- Postgraduate Program in Health and Environment, University of Joinville Region, Rua Paulo Malschitzki, 10 - Zona Industrial Norte, Joinville, SC 89219-710, Brazil; (S.M.S.); (G.S.E.)
| | - Gilmar Sidnei Erzinger
- Postgraduate Program in Health and Environment, University of Joinville Region, Rua Paulo Malschitzki, 10 - Zona Industrial Norte, Joinville, SC 89219-710, Brazil; (S.M.S.); (G.S.E.)
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, 8000 Aarhus C, Denmark; (D.G.); (T.J.C.)
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Blvd. 99, 8200 Aarhus N, Denmark
| | - Michael Lebert
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, Staudtstraße 5, 91058 Erlangen, Germany; (B.P.); (M.L.)
- Space Biology Unlimited SAS, 24 Cours de l’Intendance, 33000 Bordeaux, France
| | - Nils E. Magnusson
- Diabetes and Hormone Diseases, Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark;
| | - Manfred Infanger
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.I.); (M.K.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Peter Richter
- Gravitational Biology Group, Department of Biology, Friedrich-Alexander University, Staudtstraße 5, 91058 Erlangen, Germany; (B.P.); (M.L.)
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.I.); (M.K.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| |
Collapse
|
22
|
Huang W, Yu J, Liu T, Tudor G, Defnet AE, Zalesak S, Kumar P, Booth C, Farese AM, MacVittie TJ, Kane MA. Proteomic Evaluation of the Natural History of the Acute Radiation Syndrome of the Gastrointestinal Tract in a Non-human Primate Model of Partial-body Irradiation with Minimal Bone Marrow Sparing Includes Dysregulation of the Retinoid Pathway. HEALTH PHYSICS 2020; 119:604-620. [PMID: 32947489 PMCID: PMC7541663 DOI: 10.1097/hp.0000000000001351] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Exposure to ionizing radiation results in injuries of the hematopoietic, gastrointestinal, and respiratory systems, which are the leading causes responsible for morbidity and mortality. Gastrointestinal injury occurs as an acute radiation syndrome. To help inform on the natural history of the radiation-induced injury of the partial body irradiation model, we quantitatively profiled the proteome of jejunum from non-human primates following 12 Gy partial body irradiation with 2.5% bone marrow sparing over a time period of 3 wk. Jejunum was analyzed by liquid chromatography-tandem mass spectrometry, and pathway and gene ontology analysis were performed. A total of 3,245 unique proteins were quantified out of more than 3,700 proteins identified in this study. Also a total of 289 proteins of the quantified proteins showed significant and consistent responses across at least three time points post-irradiation, of which 263 proteins showed strong upregulations while 26 proteins showed downregulations. Bioinformatic analysis suggests significant pathway and upstream regulator perturbations post-high dose irradiation and shed light on underlying mechanisms of radiation damage. Canonical pathways altered by radiation included GP6 signaling pathway, acute phase response signaling, LXR/RXR activation, and intrinsic prothrombin activation pathway. Additionally, we observed dysregulation of proteins of the retinoid pathway and retinoic acid, an active metabolite of vitamin A, as quantified by liquid chromatography-tandem mass spectrometry. Correlation of changes in protein abundance with a well-characterized histological endpoint, corrected crypt number, was used to evaluate biomarker potential. These data further define the natural history of the gastrointestinal acute radiation syndrome in a non-human primate model of partial body irradiation with minimal bone marrow sparing.
Collapse
Affiliation(s)
- Weiliang Huang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | - Jianshi Yu
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | - Tian Liu
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | | | - Amy E Defnet
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | - Stephanie Zalesak
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | - Praveen Kumar
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
| | | | - Ann M. Farese
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Maureen A Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, USA
- Correspondence: Maureen A. Kane, University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, 20 N. Pine Street, Room N731, Baltimore, MD 21201, Phone: (410) 706-5097, Fax: (410) 706-0886,
| |
Collapse
|
23
|
Al Zouabi L, Bardin AJ. Stem Cell DNA Damage and Genome Mutation in the Context of Aging and Cancer Initiation. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a036210. [PMID: 31932318 DOI: 10.1101/cshperspect.a036210] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Adult stem cells fuel tissue homeostasis and regeneration through their unique ability to self-renew and differentiate into specialized cells. Thus, their DNA provides instructions that impact the tissue as a whole. Since DNA is not an inert molecule, but rather dynamic, interacting with a myriad of chemical and physical factors, it encounters damage from both endogenous and exogenous sources. Damage to DNA introduces deviations from its normal intact structure and, if left unrepaired, may result in a genetic mutation. In turn, mutant genomes of stem and progenitor cells are inherited in cells of the lineage, thus eroding the genetic information that maintains homeostasis of the somatic cell population. Errors arising in stem and progenitor cells will have a substantially larger impact on the tissue in which they reside than errors occurring in postmitotic differentiated cells. Therefore, maintaining the integrity of genomic DNA within our stem cells is essential to protect the instructions necessary for rebuilding healthy tissues during homeostatic renewal. In this review, we will first discuss DNA damage arising in stem cells and cell- and tissue-intrinsic mechanisms that protect against harmful effects of this damage. Secondly, we will examine how erroneous DNA repair and persistent DNA damage in stem and progenitor cells impact stem cells and tissues in the context of cancer initiation and aging. Finally, we will discuss the use of invertebrate and vertebrate model systems to address unanswered questions on the role that DNA damage and mutation may play in aging and precancerous conditions.
Collapse
Affiliation(s)
- Lara Al Zouabi
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, 75248 Paris, France.,Sorbonne Universités, UPMC University, Paris 6, France
| | - Allison J Bardin
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, 75248 Paris, France.,Sorbonne Universités, UPMC University, Paris 6, France
| |
Collapse
|
24
|
Matsuu-Matsuyama M, Shichijo K, Tsuchiya T, Kondo H, Miura S, Matsuda K, Sekine I, Nakashima M. Protective effects of a cystine and theanine mixture against acute radiation injury in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 78:103395. [PMID: 32325407 DOI: 10.1016/j.etap.2020.103395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 06/11/2023]
Abstract
This study aims to examine the effects of cystine and theanine (CT), which increases glutathione biosynthesis, on the survival rate and acute radiation injury of the small intestine and bone marrow using a rat model. CT pre-treatment (280 mg/kg for 5 days) significantly improved weight loss and survival rate of rats as compared with the control group after 5 Gy. CT pre-treatment significantly increased the rate of mucosa and crypt length, and decreased the number of apoptotic cells, TUNEL and cleaved caspase-3 positive cells, while increasing the number of mitotic cells and Ki-67 positive cells in jejunal crypts and villi compared to control rats post-irradiation. CT also suppressed bone marrow cell loss and reduced the number of apoptotic cells in bone marrow. These results suggest a protective effect of CT pre-treatment for acute injury after irradiation through apoptosis inhibition and increased proliferative activity in jejunal crypt cells and bone marrow cells.
Collapse
Affiliation(s)
- Mutsumi Matsuu-Matsuyama
- Tissue and Histopathology Section, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| | - Kazuko Shichijo
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| | - Takashi Tsuchiya
- Sendai City Medical Center, 5-22-1 Tsurugaya, Miyagino, Miyagi 983-0824, Japan.
| | - Hisayoshi Kondo
- Biostatistics Section, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| | - Shiro Miura
- Department of Pathology, National Hospital Organization Nagasaki Medical Center, 2-1001-1 Kubara, Omura, Nagasaki 856-8562, Japan.
| | - Katsuya Matsuda
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| | - Ichiro Sekine
- Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| | - Masahiro Nakashima
- Tissue and Histopathology Section, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| |
Collapse
|
25
|
Jardé T, Chan WH, Rossello FJ, Kaur Kahlon T, Theocharous M, Kurian Arackal T, Flores T, Giraud M, Richards E, Chan E, Kerr G, Engel RM, Prasko M, Donoghue JF, Abe SI, Phesse TJ, Nefzger CM, McMurrick PJ, Powell DR, Daly RJ, Polo JM, Abud HE. Mesenchymal Niche-Derived Neuregulin-1 Drives Intestinal Stem Cell Proliferation and Regeneration of Damaged Epithelium. Cell Stem Cell 2020; 27:646-662.e7. [PMID: 32693086 DOI: 10.1016/j.stem.2020.06.021] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/13/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022]
Abstract
Epidermal growth factor (EGF) maintains intestinal stem cell (ISC) proliferation and is a key component of organoid growth media yet is dispensable for intestinal homeostasis, suggesting roles for multiple EGF family ligands in ISC function. Here, we identified neuregulin 1 (NRG1) as a key EGF family ligand that drives tissue repair following injury. NRG1, but not EGF, is upregulated upon damage and is expressed in mesenchymal stromal cells, macrophages, and Paneth cells. NRG1 deletion reduces proliferation in intestinal crypts and compromises regeneration capacity. NRG1 robustly stimulates proliferation in crypts and induces budding in organoids, in part through elevated and sustained activation of mitogen-activated protein kinase (MAPK) and AKT. Consistently, NRG1 treatment induces a proliferative gene signature and promotes organoid formation from progenitor cells and enhances regeneration following injury. These data suggest mesenchymal-derived NRG1 is a potent mediator of tissue regeneration and may inform the development of therapies for enhancing intestinal repair after injury.
Collapse
Affiliation(s)
- Thierry Jardé
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia; Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia.
| | - Wing Hei Chan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Fernando J Rossello
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; University of Melbourne Centre for Cancer Research, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Tanvir Kaur Kahlon
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Mandy Theocharous
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia; Cancer Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Teni Kurian Arackal
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Tracey Flores
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Mégane Giraud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Elizabeth Richards
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Eva Chan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Genevieve Kerr
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Rebekah M Engel
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia; Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern, VIC 3144, Australia
| | - Mirsada Prasko
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Jacqueline F Donoghue
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; Department of Obstetrics and Gynaecology, Royal Women's Hospital, Melbourne University, Melbourne, VIC 3052, Australia
| | - Shin-Ichi Abe
- Center for Education, Kumamoto Health Science University, Kumamoto 861-5598, Japan
| | - Toby J Phesse
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK; Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Christian M Nefzger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Paul J McMurrick
- Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern, VIC 3144, Australia
| | - David R Powell
- Monash Bioinformatics Platform, Monash University, Clayton, VIC 3800, Australia
| | - Roger J Daly
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia; Cancer Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia.
| |
Collapse
|
26
|
Liju VB, Thomas A, Sivadasan SD, Kuttan R, Maliakel B, Im K. Amelioration of Radiation-Induced Damages in Mice by Curcuminoids: The Role of Bioavailability. Nutr Cancer 2020; 73:617-629. [PMID: 32449395 DOI: 10.1080/01635581.2020.1766092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE The present study investigated the role of free curcuminoids bioavailability on the relative radioprotective efficacy of natural unformulated curcuminoids. MATERIALS AND METHODS A food-grade bioavailable formulation of curcuminoids as curcumagalactomannosides (CGM) and unformulated curcuminoids (UC) were employed for the study. Swiss albino mice were randomized into Normal control, Radiation control, Radiation + UC, and Radiation + CGM groups and irradiated with γ-radiation of 6, 8, 10 and 12 Gy. Survival rate, hematological and biochemical parameters, bone marrow cellularity, chromosomal aberrations and histopathology of intestine were followed as a measure of the relative efficacy.Results and Discussion: Oral administration with both UC and CGM at 100 mg/kg. b.wt. produced significant radioprotective effect over the untreated control group of animals. However, CGM treatment was found to provide better clastogenic and genotoxic potential as compared to UC. Further, the histopathology analysis of intestine confirmed the better protective effect of CGM over UC-treated animals. CONCLUSION The present study demonstrated the positive role of the bioavailability of curcuminoids in the amelioration of radiation-induced damages in mice since CGM treatment exerted better survival rate and radioprotective effect as compared with UC, despite the relatively low concentrations of curcuminoids in CGM (39% w/w).
Collapse
Affiliation(s)
| | - Angel Thomas
- Amala Cancer Research Center, Thrissur, Kerala, India
| | | | | | - Balu Maliakel
- R&D Centre, Akay Natural Ingredients Pvt. Ltd., Kerala, India
| | - Krishnakumar Im
- R&D Centre, Akay Natural Ingredients Pvt. Ltd., Kerala, India
| |
Collapse
|
27
|
Akpolat M, Oz ZS, Gulle K, Hamamcioglu AC, Bakkal BH, Kececi M. X irradiation induced colonic mucosal injury and the detection of apoptosis through PARP-1/p53 regulatory pathway. Biomed Pharmacother 2020; 127:110134. [PMID: 32361637 DOI: 10.1016/j.biopha.2020.110134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022] Open
Abstract
This study aimed to explore whether PARP-1 regulatory pathway mediated X irradiation induced cell cycle arrest and apoptosis or not. In this regard, colonic mucosal injury caused by whole-body X-irradiation induced apoptosis through PARP-1, caspase 3 and p53 regulatory pathway were evaluated in experimental rat models. Eighteen Wistar albino rats were divided into three groups. Two radiation groups received 8.3 Gy dose of whole-body X-irradiation as a single dose and the control group received physiological saline intraperitoneally. Radiation groups were sacrificed after 6 h and 4 days of irradiation. PARP-1 and caspase 3 expression in the nuclei of colonic crypt cells significantly increased 6 h after irradiation, and declined 4 days after irradiation. In conflict with other studies that reported p53 as not being expressed widely in colonic mucosa, in our study the expressions of p53 were elevated both in the cytoplasm and in the nucleus of the crypt cells, especially 6 h after irradiation. In the radiation groups, colonic mucosal injury score was significantly elevated compared with that of the control group. Our data demonstrated that PARP-1, caspase-3 and p53 expression increased in colonic mucosa 6 h after irradiation.
Collapse
Affiliation(s)
- Meryem Akpolat
- Zonguldak Bulent Ecevit University, Faculty of Medicine, Department of Histology and Embryology, Zonguldak, Turkey.
| | - Zehra Safi Oz
- Zonguldak Bulent Ecevit University Faculty of Medicine, Department of Medical Biology, Zonguldak, Turkey.
| | - Kanat Gulle
- Suleyman Demirel University, Faculty of Medicine, Department of Histology and Embryology, Isparta, Turkey.
| | - Ayse C Hamamcioglu
- Zonguldak Bulent Ecevit University, Faculty of Pharmacy, Department of Biochemistry, Zonguldak, Turkey.
| | - Bekir H Bakkal
- Zonguldak Bulent Ecevit University, Faculty of Medicine, Department of Radiation Oncology, Zonguldak, Turkey.
| | - Mete Kececi
- Zonguldak Bulent Ecevit University, Faculty of Medicine, Department of Histology and Embryology, Zonguldak, Turkey
| |
Collapse
|
28
|
Li K, Zhang J, Cao J, Li X, Tian H. 1,4-Dithiothreitol treatment ameliorates hematopoietic and intestinal injury in irradiated mice: Potential application of a treatment for acute radiation syndrome. Int Immunopharmacol 2019; 76:105913. [PMID: 31627170 DOI: 10.1016/j.intimp.2019.105913] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/29/2019] [Accepted: 09/12/2019] [Indexed: 01/07/2023]
Abstract
Radiation exposure poses a significant threat to public health, which can lead to acute hematopoietic system and intestinal system injuries due to their higher radiation sensitivity. Hence, antioxidants and thiol-reducing agents could have a potential protective effect against this complication. The dithiol compound 1,4-dithiothreitol (DTT) has been used in biochemistry, peptide/protein chemistry and clinical medicine. However, the effect of DTT on ionizing radiation (IR)-induced hematopoietic injury and intestinal injury are unknown. The current investigation was designed to evaluate the effect of DTT as a safe and clinically applicable thiol-radioprotector in irradiated mice. DTT treatment improved the survival of irradiated mice and ameliorated whole body irradiation (WBI)-induced hematopoietic injury by attenuating myelosuppression and myeloid skewing, increasing self-renewal and differentiation of hematopoietic progenitor cells/hematopoietic stem cells (HPCs/HSCs). In addition, DTT treatment protected mice from abdominal irradiation (ABI)-induced changes in crypt-villus structures and function. Furthermore, treatment with DTT significantly enhanced the ABI-induced reduction in Olfm4 positive cells and offspring cells of Lgr5+ stem cells, including lysozyme+ Paneth cells and Ki67+ cells. Moreover, IR-induced DNA strand break damage, and the expression of proapoptotic-p53, Bax, Bak protein and antiapoptotic-Bcl-2 protein were reversed in DTT treated mice, and DTT also promoted small intestine repair after radiation exposure via the p53 intrinsic apoptotic pathway. In general, these results demonstrated the potential of DTT for protection against hematopoietic injury and intestinal injury after radiation exposure, suggesting DTT as a novel effective agent for radioprotection.
Collapse
Affiliation(s)
- Kui Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Junling Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Jian Cao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Xuejiao Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
29
|
Malipatlolla DK, Patel P, Sjöberg F, Devarakonda S, Kalm M, Angenete E, Lindskog EB, Grandér R, Persson L, Stringer A, Wilderäng U, Swanpalmer J, Kuhn HG, Steineck G, Bull C. Long-term mucosal injury and repair in a murine model of pelvic radiotherapy. Sci Rep 2019; 9:13803. [PMID: 31551503 PMCID: PMC6760522 DOI: 10.1038/s41598-019-50023-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 09/04/2019] [Indexed: 01/02/2023] Open
Abstract
Chronic intestinal injury after pelvic radiotherapy affects countless cancer survivors worldwide. A comprehensive understanding of the long-term injury dynamics is prevented in available animal models. With linear accelerators that are used to treat cancer in patients, we irradiated a small volume encompassing the colorectum in mice with four fractions of 8 Gy per fraction. We then determined the long-term dynamics of mucosal injury, repair, and the duration of inflammation. We show that crypt fission, not cell proliferation, is the main long-term mechanism for rescuing crypt density after irradiation, and provides a potentially wide window for clinical interventions. Persisting macrophage aggregations indicate a chronic mucosal inflammation. A better understanding as to how crypt fission is triggered and why it fails to repair fully the mucosa may help restore bowel health after pelvic radiotherapy. Moreover, anti-inflammatory interventions, even if implemented long after completed radiotherapy, could promote bowel health in pelvic cancer survivors.
Collapse
Affiliation(s)
- Dilip K Malipatlolla
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Piyush Patel
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Infectious Diseases at the Institute of Biomedicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Fei Sjöberg
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Infectious Diseases at the Institute of Biomedicine, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sravani Devarakonda
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marie Kalm
- Department of Pharmacology at the Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Eva Angenete
- The Department of Surgery at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elinor Bexe Lindskog
- The Department of Surgery at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Rita Grandér
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Linda Persson
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Andrea Stringer
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Ulrica Wilderäng
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - John Swanpalmer
- Department of Radiation Physics at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Hans Georg Kuhn
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Gunnar Steineck
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cecilia Bull
- The Division of Clinical Cancer Epidemiology, Department of Oncology at the Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
30
|
Preclinical murine platform to evaluate therapeutic countermeasures against radiation-induced gastrointestinal syndrome. Proc Natl Acad Sci U S A 2019; 116:20672-20678. [PMID: 31551264 PMCID: PMC6789742 DOI: 10.1073/pnas.1906611116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Currently, there are no therapies available to mitigate intestinal damage after radiation injury. Efforts to study and design new therapies are hampered by a lack of models that can be readily adopted to study therapeutic targets. Here we describe a preclinical platform to evaluate therapeutic countermeasures against intestinal radiation injury in vivo in a mouse model that permits inducible and reversible gene suppression following radiation exposure. We demonstrate that transient intestinal Apc suppression stimulates intestinal regeneration and mitigates lethality after radiation intestinal injury, thus validating pulsed Wnt pathway agonism as a therapeutic strategy. This platform can be readily adopted to study theoretically any gene of interest associated with the biology and treatment of intestinal radiation injury. Radiation-induced gastrointestinal syndrome (RIGS) is a limiting factor for therapeutic abdominopelvic radiation and is predicted to be a major source of morbidity in the event of a nuclear accident or radiological terrorism. In this study, we developed an in vivo mouse-modeling platform that enables spatial and temporal manipulation of potential RIGS targets in mice following whole-abdomen irradiation without the confounding effects of concomitant hematopoietic syndrome that occur following whole-body irradiation. We then tested the utility of this platform to explore the effects of transient Wnt pathway activation on intestinal regeneration and animal recovery following induction of RIGS. Our results demonstrate that intestinal epithelial suppression of adenomatous polyposis coli (Apc) mitigates RIGS lethality in vivo after lethal ionizing radiation injury-induced intestinal epithelial damage. These results highlight the potential of short-term Wnt agonism as a therapeutic target and establish a platform to evaluate other strategies to stimulate intestinal regeneration after ionizing radiation damage.
Collapse
|
31
|
Trosko JE. What Can Chemical Carcinogenesis Shed Light on the LNT Hypothesis in Radiation Carcinogenesis? Dose Response 2019; 17:1559325819876799. [PMID: 31565039 PMCID: PMC6755642 DOI: 10.1177/1559325819876799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 08/27/2019] [Indexed: 12/14/2022] Open
Abstract
To protect the public’s health from exposure to physical, chemical, and microbiological
agents, it is important that any policy be based on rigorous scientifically based
research. The concept of “linear no-threshold” (LNT) has been implemented to provide
guideline exposures to these agents. The practical limitation to testing this hypothesis
is to provide sufficient samples for experimental or epidemiological studies. While there
is no universally accepted understanding of most human diseases, there seems to be better
understanding of cancer that might help resolve the “LNT” model. The public’s concern,
after being exposed to radiation, is the potential of producing cancer. The most rigorous
hypothesis of human carcinogenesis is the “multistage, multimechanism” chemical
carcinogenesis model. The radiation carcinogenesis LNT model, rarely, if ever, built it
into their support. It will be argued that this multistage, multimechanism model of
carcinogenesis, involving the “initiation” of a single cell by a mutagen event, followed
by chronic exposure to threshold levels of epigenetic agents or conditions that stimulate
the clonal expansion of the “initiated” cell, can convert these benign cells to become
invasive and metastatic. This “promotion” process can be interrupted, thereby preventing
these initiated cells from transitioning to the “progression” process of invasion and
metastasis.
Collapse
Affiliation(s)
- James E Trosko
- Department Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
32
|
Duhachek-Muggy S, Bhat K, Medina P, Cheng F, He L, Alli C, Saki M, Muthukrishnan SD, Ruffenach G, Eghbali M, Vlashi E, Pajonk F. Radiation mitigation of the intestinal acute radiation injury in mice by 1-[(4-nitrophenyl)sulfonyl]-4-phenylpiperazine. Stem Cells Transl Med 2019; 9:106-119. [PMID: 31464098 PMCID: PMC6954722 DOI: 10.1002/sctm.19-0136] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/19/2019] [Indexed: 12/29/2022] Open
Abstract
The objective of the study was to identify the mechanism of action for a radiation mitigator of the gastrointestinal (GI) acute radiation syndrome (ARS), identified in an unbiased high‐throughput screen. We used mice irradiated with a lethal dose of radiation and treated with daily injections of the radiation mitigator 1‐[(4‐nitrophenyl)sulfonyl]‐4‐phenylpiperazine to study its effects on key pathways involved in intestinal stem cell (ISC) maintenance. RNASeq, quantitative reverse transcriptase‐polymerase chain reaction, and immunohistochemistry were performed to identify pathways engaged after drug treatment. Target validation was performed with competition assays, reporter cells, and in silico docking. 1‐[(4‐Nitrophenyl)sulfonyl]‐4‐phenylpiperazine activates Hedgehog signaling by binding to the transmembrane domain of Smoothened, thereby expanding the ISC pool, increasing the number of regenerating crypts and preventing the GI‐ARS. We conclude that Smoothened is a target for radiation mitigation in the small intestine that could be explored for use in radiation accidents as well as to mitigate normal tissue toxicity during and after radiotherapy of the abdomen.
Collapse
Affiliation(s)
- Sara Duhachek-Muggy
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Paul Medina
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Fei Cheng
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Ling He
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Claudia Alli
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Mohammad Saki
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Sree Deepthi Muthukrishnan
- Department of Psychiatry, Semel Institute of Neuroscience and Human Behavior, UCLA, Los Angeles, California
| | - Gregoire Ruffenach
- Department of Anesthesiology, Division of Molecular Medicine, Cardiovascular Research Laboratory, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Mansoureh Eghbali
- Department of Anesthesiology, Division of Molecular Medicine, Cardiovascular Research Laboratory, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
33
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Potes Y, Shabeeb D, Musa AE. Modulation of apoptosis by melatonin for improving cancer treatment efficiency: An updated review. Life Sci 2019; 228:228-241. [DOI: 10.1016/j.lfs.2019.05.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022]
|
34
|
Kiang JG, Smith JT, Anderson MN, Umali MV, Ho C, Zhai M, Lin B, Jiang S. A novel therapy, using Ghrelin with pegylated G-CSF, inhibits brain hemorrhage from ionizing radiation or combined radiation injury. PHARMACY & PHARMACOLOGY INTERNATIONAL JOURNAL 2019; 7:133-145. [PMID: 34368440 PMCID: PMC8341084 DOI: 10.15406/ppij.2019.07.00243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Medical treatment becomes challenging when complicated injuries arise from secondary reactive metabolic and inflammatory products induced by initial acute ionizing radiation injury (RI) or when combined with subsequent trauma insult(s) (CI). With such detrimental effects on many organs, CI exacerbates the severity of primary injuries and decreases survival. Previously, in a novel study, we reported that ghrelin therapy significantly improved survival after CI. This study aimed to investigate whether brain hemorrhage induced by RI and CI could be inhibited by ghrelin therapy with pegylated G-CSF (i.e., Neulasta®, an FDA-approved drug). B6D2F1 female mice were exposed to 9.5 Gy 60Co-γ-radiation followed by 15% total-skin surface wound. Several endpoints were measured at several days. Brain hemorrhage and platelet depletion were observed in RI and CI mice. Brain hemorrhage severity was significantly higher in CI mice than in RI mice. Ghrelin therapy with pegylated G-CSF reduced the severity in brains of both RI and CI mice. RI and CI did not alter PARP and NF-κB but did significantly reduce PGC-1α and ghrelin receptors; the therapy, however, was able to partially recover ghrelin receptors. RI and CI significantly increased IL-6, KC, Eotaxin, G-CSF, MIP-2, MCP-1, MIP-1α, but significantly decreased IL-2, IL-9, IL-10, MIG, IFN-γ, and PDGF-bb; the therapy inhibited these changes. RI and CI significantly reduced platelet numbers, cellular ATP levels, NRF1/2, and AKT phosphorylation. The therapy significantly mitigated these CI-induced changes and reduced p53-mdm2 mediated caspase-3 activation. Our data are the first to support the view that Ghrelin therapy with pegylated G-CSF is potentially a novel therapy for treating brain hemorrhage after RI and CI.
Collapse
Affiliation(s)
- J G Kiang
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, USA
| | - J T Smith
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - M N Anderson
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - M V Umali
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - C Ho
- Department of Biochemistry, University of California, USA
| | - M Zhai
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - B Lin
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| | - S Jiang
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, USA
| |
Collapse
|
35
|
Chuah LO, Foo HL, Loh TC, Mohammed Alitheen NB, Yeap SK, Abdul Mutalib NE, Abdul Rahim R, Yusoff K. Postbiotic metabolites produced by Lactobacillus plantarum strains exert selective cytotoxicity effects on cancer cells. Altern Ther Health Med 2019; 19:114. [PMID: 31159791 PMCID: PMC6547513 DOI: 10.1186/s12906-019-2528-2] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/23/2019] [Indexed: 12/31/2022]
Abstract
Background Lactobacillus plantarum, a major species of Lactic Acid Bacteria (LAB), are capable of producing postbiotic metabolites (PM) with prominent probiotic effects that have been documented extensively for rats, poultry and pigs. Despite the emerging evidence of anticancer properties of LAB, very limited information is available on cytotoxic and antiproliferative activity of PM produced by L. plantarum. Therefore, the cytotoxicity of PM produced by six strains of L. plantarum on various cancer and normal cells are yet to be evaluated. Methods Postbiotic metabolites (PM) produced by six strains of L. plantarum were determined for their antiproliferative and cytotoxic effects on normal human primary cells, breast, colorectal, cervical, liver and leukemia cancer cell lines via MTT assay, trypan blue exclusion method and BrdU assay. The toxicity of PM was determined for human and various animal red blood cells via haemolytic assay. The cytotoxicity mode was subsequently determined for selected UL4 PM on MCF-7 cells due to its pronounced cytotoxic effect by fluorescent microscopic observation using AO/PI dye reagents and flow cytometric analyses. Results UL4 PM exhibited the lowest IC50 value on MCF-7, RG14 PM on HT29 and RG11 and RI11 PM on HL60 cell lines, respectively from MTT assay. Moreover, all tested PM did not cause haemolysis of human, dog, rabbit and chicken red blood cells and demonstrated no cytotoxicity on normal breast MCF-10A cells and primary cultured cells including human peripheral blood mononuclear cells, mice splenocytes and thymocytes. Antiproliferation of MCF-7 and HT-29 cells was potently induced by UL4 and RG 14 PM respectively after 72 h of incubation at the concentration of 30% (v/v). Fluorescent microscopic observation and flow cytometric analyses showed that the pronounced cytotoxic effect of UL4 PM on MCF-7 cells was mediated through apoptosis. Conclusion In conclusion, PM produced by the six strains of L. plantarum exhibited selective cytotoxic via antiproliferative effect and induction of apoptosis against malignant cancer cells in a strain-specific and cancer cell type-specific manner whilst sparing the normal cells. This reveals the vast potentials of PM from L. plantarum as functional supplement and as an adjunctive treatment for cancer.
Collapse
|
36
|
Radiation-induced oxidative injury of the ileum and colon is alleviated by glucagon-like peptide-1 and -2. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2019. [DOI: 10.1016/j.jrras.2015.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
37
|
Carter CL, Hankey KG, Booth C, Tudor GL, Parker GA, Jones JW, Farese AM, MacVittie TJ, Kane MA. Characterizing the Natural History of Acute Radiation Syndrome of the Gastrointestinal Tract: Combining High Mass and Spatial Resolution Using MALDI-FTICR-MSI. HEALTH PHYSICS 2019; 116:454-472. [PMID: 30681424 PMCID: PMC6384159 DOI: 10.1097/hp.0000000000000948] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The acute radiation syndrome of the gastrointestinal tract has been histologically characterized, but the molecular and functional mechanisms that lead to these cellular alterations remain enigmatic. Mass spectrometry imaging is the only technique that enables the simultaneous detection and cellular or regional localization of hundreds of biomolecules in a single experiment. This current study utilized matrix-assisted laser desorption/ionization mass spectrometry imaging for the molecular characterization of the first natural history study of gastrointestinal acute radiation syndrome in the nonhuman primate. Jejunum samples were collected at days 4, 8, 11, 15, and 21 following 12-Gy partial-body irradiation with 2.5% bone marrow sparing. Mass spectrometry imaging investigations identified alterations in lipid species that further understanding of the functional alterations that occur over time in the different cellular regions of the jejunum following exposure to high doses of irradiation. Alterations in phosphatidylinositol species informed on dysfunctional epithelial cell differentiation and maturation. Differences in glycosphingolipids of the villi epithelium that would influence the absorptive capacity and functional structure of the brush border membrane were detected. Dichotomous alterations in cardiolipins indicated altered structural and functional integrity of mitochondria. Phosphatidylglycerol species, known regulators of toll-like receptors, were detected and localized to regions in the lamina propria that contained distinct immune cell populations. These results provide molecular insight that can inform on injury mechanism in a nonhuman primate model of the acute radiation syndrome of the gastrointestinal tract. Findings may contribute to the identification of therapeutic targets and the development of new medical countermeasures.
Collapse
Affiliation(s)
- Claire L. Carter
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD USA
| | - Kim G. Hankey
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD USA
| | | | | | - George A. Parker
- Charles River Laboratories, Pathology Associates, Raleigh-Durham, North Carolina, USA
| | - Jace W. Jones
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD USA
| | - Ann M. Farese
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD USA
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD USA
| | - Maureen A. Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD USA
| |
Collapse
|
38
|
Cheng Y, Dong Y, Hou Q, Wu J, Zhang W, Tian H, Li D. The protective effects of XH-105 against radiation-induced intestinal injury. J Cell Mol Med 2019; 23:2238-2247. [PMID: 30663222 PMCID: PMC6378229 DOI: 10.1111/jcmm.14159] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/17/2018] [Accepted: 12/24/2018] [Indexed: 12/26/2022] Open
Abstract
Radiation-induced intestinal injury is one of the major side effects in patients receiving radiation therapy. There is no specific treatment for radiation enteritis in the clinic. We designed and synthesized a new compound named XH-105, which is expected to cleave into polyphenol and aminothiol in vivo to mitigate radiation injury. In the following study, we describe the beneficial effects of XH-105 against radiation-induced intestinal injury. C57BL/6J mice were treated by gavage with XH-105 1 hour before total body irradiation (TBI), and the survival rate was monitored. Histological changes were examined, and survival of Lgr5+ intestinal stem cells Ki67+ cells, villi+ enterocytes and lysozymes was determined by immunohistochemistry. DNA damage and cellular apoptosis in intestinal tissue were also evaluated. Compared to vehicle-treated mice after TBI, XH-105 treatment significantly enhanced the survival rate, attenuated structural damage of the small intestine, decreased the apoptotic rate, reduced DNA damage, maintained cell regeneration and promoted crypt proliferation and differentiation. XH-105 also reduced the expression of Bax and p53 in the small intestine. These data suggest that XH-105 is beneficial for the protection of radiation-induced intestinal injury by inhibiting the p53-dependent apoptosis signalling pathway.
Collapse
Affiliation(s)
- Ying Cheng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.,Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Qinlian Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Jing Wu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Wei Zhang
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| |
Collapse
|
39
|
Doggett K, Williams BB, Markmiller S, Geng FS, Coates J, Mieruszynski S, Ernst M, Thomas T, Heath JK. Early developmental arrest and impaired gastrointestinal homeostasis in U12-dependent splicing-defective Rnpc3-deficient mice. RNA (NEW YORK, N.Y.) 2018; 24:1856-1870. [PMID: 30254136 PMCID: PMC6239176 DOI: 10.1261/rna.068221.118] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/20/2018] [Indexed: 05/10/2023]
Abstract
Splicing is an essential step in eukaryotic gene expression. While the majority of introns is excised by the U2-dependent, or major class, spliceosome, the appropriate expression of a very small subset of genes depends on U12-dependent, or minor class, splicing. The U11/U12 65K protein (hereafter 65K), encoded by RNPC3, is one of seven proteins that are unique to the U12-dependent spliceosome, and previous studies including our own have established that it plays a role in plant and vertebrate development. To pinpoint the impact of 65K loss during mammalian development and in adulthood, we generated germline and conditional Rnpc3-deficient mice. Homozygous Rnpc3-/- embryos died prior to blastocyst implantation, whereas Rnpc3+/- mice were born at the expected frequency, achieved sexual maturity, and exhibited a completely normal lifespan. Systemic recombination of conditional Rnpc3 alleles in adult (Rnpc3lox/lox ) mice caused rapid weight loss, leukopenia, and degeneration of the epithelial lining of the entire gastrointestinal tract, the latter due to increased cell death and a reduction in cell proliferation. Accompanying this, we observed a loss of both 65K and the pro-proliferative phospho-ERK1/2 proteins from the stem/progenitor cells at the base of intestinal crypts. RT-PCR analysis of RNA extracted from purified preparations of intestinal epithelial cells with recombined Rnpc3lox alleles revealed increased frequency of U12-type intron retention in all transcripts tested. Our study, using a novel conditional mouse model of Rnpc3 deficiency, establishes that U12-dependent splicing is not only important during development but is indispensable throughout life.
Collapse
Affiliation(s)
- Karen Doggett
- Development and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Ben B Williams
- Development and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Sebastian Markmiller
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Fan-Suo Geng
- Development and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Janine Coates
- Development and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Stephen Mieruszynski
- Development and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria 3050, Australia
| | - Tim Thomas
- Development and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Joan K Heath
- Development and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
- Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria 3050, Australia
| |
Collapse
|
40
|
Koren E, Yosefzon Y, Ankawa R, Soteriou D, Jacob A, Nevelsky A, Ben-Yosef R, Bar-Sela G, Fuchs Y. ARTS mediates apoptosis and regeneration of the intestinal stem cell niche. Nat Commun 2018; 9:4582. [PMID: 30389919 PMCID: PMC6214937 DOI: 10.1038/s41467-018-06941-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023] Open
Abstract
Stem cells (SCs) play a pivotal role in fueling homeostasis and regeneration. While much focus has been given to self-renewal and differentiation pathways regulating SC fate, little is known regarding the specific mechanisms utilized for their elimination. Here, we report that the pro-apoptotic protein ARTS (a Septin4 isoform) is highly expressed in cells comprising the intestinal SC niche and that its deletion protects Lgr5+ and Paneth cells from undergoing apoptotic cell death. As a result, the Sept4/ARTS−/− crypt displays augmented proliferation and, in culture, generates massive cystic-like organoids due to enhanced Wnt/β-catenin signaling. Importantly, Sept4/ARTS−/− mice exhibit resistance against intestinal damage in a manner dependent upon Lgr5+ SCs. Finally, we show that ARTS interacts with XIAP in intestinal crypt cells and that deletion of XIAP can abrogate Sept4/ARTS−/−-dependent phenotypes. Our results indicate that intestinal SCs utilize specific apoptotic proteins for their elimination, representing a unique target for regenerative medicine. The mechanisms regulating intestinal stem cell elimination remain unclear. Here, the authors identify that the pro-apoptotic protein ARTS (a Septin4 isoform) interacts with XIAP in the intestinal stem cell niche to regulate stem cell survival during intestinal homeostasis and regeneration.
Collapse
Affiliation(s)
- Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.,Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, 3200003, Israel.,Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Yahav Yosefzon
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.,Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, 3200003, Israel.,Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Roi Ankawa
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.,Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, 3200003, Israel.,Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Despina Soteriou
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel.,Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, 3200003, Israel.,Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Avi Jacob
- Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel
| | - Alexander Nevelsky
- Oncology Division, Rambam Health Care Campus, P.O.B. 9602, Haifa, 31096, Israel
| | - Rahamim Ben-Yosef
- Oncology Division, Rambam Health Care Campus, P.O.B. 9602, Haifa, 31096, Israel
| | - Gil Bar-Sela
- Oncology Division, Rambam Health Care Campus, P.O.B. 9602, Haifa, 31096, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel. .,Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, 3200003, Israel. .,Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
41
|
Association between radiation-induced cell death and clinically relevant radioresistance. Histochem Cell Biol 2018; 150:649-659. [DOI: 10.1007/s00418-018-1728-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2018] [Indexed: 02/06/2023]
|
42
|
The Protective Effect of New Compound XH-103 on Radiation-Induced GI Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3920147. [PMID: 30116481 PMCID: PMC6079366 DOI: 10.1155/2018/3920147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/01/2018] [Accepted: 05/21/2018] [Indexed: 12/13/2022]
Abstract
Background Radiation-induced intestinal injury is one of the side effects in patients receiving radiotherapy. The aim of the present study was to investigate the protective effect of XH-103 on radiation-induced small intestinal injury and to explore its mechanism. Methods C57BL/6N mice were irradiated and treated with XH-103. Firstly, the survival rate of mice exposed to 9.0 Gy and 11.0 Gy total body irradiation (TBI) was examined. Subsequently, at 3.5 d after IR, the small intestinal morphological changes were examined by HE. The numbers of crypt cells, the villus height, the expression of Ki67 and Lgr5, and the apoptotic cells in the intestinal crypts were examined by immunohistochemistry. Furthermore, the expression of p53 and Bax was analyzed by WB. Results Compared to the irradiation group, XH-103 improved the mice survival rate, protected the intestinal morphology of mice, decreased the apoptotic rate of intestinal crypt cells, maintained cell regeneration, and promoted crypt proliferation and differentiation. XH-103 also reduced the expression of p53 and Bax in the small intestine compared to the IR group. Conclusion These data demonstrate that XH-103 can prevent radiation-induced intestinal injury, which is beneficial for the protection of radiation injuries.
Collapse
|
43
|
Wang J, Li T, Feng J, Li L, Wang R, Cheng H, Yuan Y. Kaempferol protects against gamma radiation-induced mortality and damage via inhibiting oxidative stress and modulating apoptotic molecules in vivo and vitro. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:128-137. [PMID: 29705372 DOI: 10.1016/j.etap.2018.04.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/12/2018] [Accepted: 04/16/2018] [Indexed: 05/02/2023]
Abstract
To investigate the potential protective effect of kaempferol, a representative flavonoid, against radiation induced mortality and injury in vivo and vitro.C57BL/6 male mice and human umbilical venous endothelial cells (HUVECs) were pretreated with kaempferol before radiation. We found that kaempferol can effectively increase 30-day survival rate after 8.5 Gy lethal total body irradiation (TBI). Mice were sacrificed at 7th day after 7 Gy TBI, we found kaempferol against radiation-induced tissues damage, by inhibiting the oxidative stress, and attenuating morphological changes and cell apoptosis. In vitro, kaempferol increased HUVECs cell viability and decrease apoptosis. It also mitigated oxidative stress and restored the abnormal expression of prx-5, Cyt-c, Caspase9 and Caspase3 in mRNA and protein level in HUVECs after radiation. Taken together, it suggests kaempferol can protect against gamma-radiation induced tissue damage and mortality. The present study is the first report of the radioprotective role of kaempferol in vivo and vitro.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mo He Rd, Shanghai 201999, China; Department of Pharmacy, Punan Hospital, Shanghai 200125, China; Department of Pharmacology, College of Pharmacy, Second Millitary Medical University, Shanghai 200433, China
| | - Tiejun Li
- Department of Pharmacy, Punan Hospital, Shanghai 200125, China
| | - Jingjing Feng
- Department of Pharmacology, College of Pharmacy, Second Millitary Medical University, Shanghai 200433, China
| | - Li Li
- Department of Pharmacology, College of Pharmacy, Second Millitary Medical University, Shanghai 200433, China
| | - Rong Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mo He Rd, Shanghai 201999, China
| | - Hao Cheng
- Department of Pharmacology, College of Pharmacy, Second Millitary Medical University, Shanghai 200433, China
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 280 Mo He Rd, Shanghai 201999, China.
| |
Collapse
|
44
|
Suzuki F, Loucas BD, Ito I, Asai A, Suzuki S, Kobayashi M. Survival of Mice with Gastrointestinal Acute Radiation Syndrome through Control of Bacterial Translocation. THE JOURNAL OF IMMUNOLOGY 2018; 201:77-86. [PMID: 29743312 DOI: 10.4049/jimmunol.1701515] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/20/2018] [Indexed: 11/19/2022]
Abstract
Macrophages (Mϕ) with the M2b phenotype (Pheno2b-Mϕ) in bacterial translocation sites have been described as cells responsible for the increased susceptibility of mice with gastrointestinal acute radiation syndrome to sepsis caused by gut bacteria. In this study, we tried to reduce the mortality of mice exposed to 7-10 Gy of gamma rays by controlling Pheno2b-Mϕ polarization in bacterial translocation sites. MicroRNA-222 was induced in association with gamma irradiation. Pheno2b-Mϕ polarization was promoted and maintained in gamma-irradiated mice through the reduction of a long noncoding RNA growth arrest-specific transcript 5 (a CCL1 gene silencer) influenced by this microRNA. Therefore, the host resistance of 7-9-Gy gamma-irradiated mice to sepsis caused by bacterial translocation was improved after treatment with CCL1 antisense oligodeoxynucleotide. However, the mortality of 10-Gy gamma-irradiated mice was not alleviated by this treatment. The crypts and villi in the ileum of 10-Gy gamma-irradiated mice were severely damaged, but these were markedly improved after transplantation of intestinal lineage cells differentiated from murine embryonic stem cells. All 10-Gy gamma-irradiated mice given both of the oligodeoxynucleotide and intestinal lineage cells survived, whereas all of the same mice given either of them died. These results indicate that high mortality rates of mice irradiated with 7-10 Gy of gamma rays are reducible by depleting CCL1 in combination with the intestinal lineage cell transplantation. These findings support the novel therapeutic possibility of victims who have gastrointestinal acute radiation syndrome for the reduction of their high mortality rates.
Collapse
Affiliation(s)
- Fujio Suzuki
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX 77555;
| | - Bradford D Loucas
- Department of Radiation Oncology, The University of Texas Medical Branch, Galveston, TX 77555
| | - Ichiaki Ito
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX 77555
| | - Akira Asai
- The Second Department of Internal Medicine, Osaka Medical College, Takatsuki 569-8686, Japan; and
| | - Sumihiro Suzuki
- Department of Biostatistics and Epidemiology, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Makiko Kobayashi
- Division of Infectious Diseases, Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
45
|
Noguchi H, Miyagi-Shiohira C, Nakashima Y. Induced Tissue-Specific Stem Cells and Epigenetic Memory in Induced Pluripotent Stem Cells. Int J Mol Sci 2018; 19:E930. [PMID: 29561778 PMCID: PMC5979574 DOI: 10.3390/ijms19040930] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/09/2018] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
Induced pluripotent stem (iPS) cells have significant implications for overcoming most of the ethical issues associated with embryonic stem (ES) cells. The pattern of expressed genes, DNA methylation, and covalent histone modifications in iPS cells are very similar to those in ES cells. However, it has recently been shown that, following the reprogramming of mouse/human iPS cells, epigenetic memory is inherited from the parental cells. These findings suggest that the phenotype of iPS cells may be influenced by their cells of origin and that their skewed differentiation potential may prove useful in the generation of differentiated cell types that are currently difficult to produce from ES/iPS cells for the treatment of human diseases. Our recent study demonstrated the generation of induced tissue-specific stem (iTS) cells by transient overexpression of the reprogramming factors combined with tissue-specific selection. iTS cells are cells that inherit numerous components of epigenetic memory from donor tissue and acquire self-renewal potential. This review describes the "epigenetic memory" phenomenon in iPS and iTS cells and the possible clinical applications of these stem cells.
Collapse
Affiliation(s)
- Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| | - Yoshiki Nakashima
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan.
| |
Collapse
|
46
|
Abstract
The transcription factor MafB is expressed by monocytes and macrophages. Efferocytosis (apoptotic cell uptake) by macrophages is important for inhibiting the development of autoimmune diseases, and is greatly reduced in Mafb-deficient macrophages. Here, we show the expression of the first protein in the classical complement pathway C1q is important for mediating efferocytosis and is reduced in Mafb-deficient macrophages. The efferocytosis defect in Mafb-deficient macrophages can be rescued by adding serum from wild-type mice, but not by adding serum from C1q-deficient mice. By hemolysis assay we also show that activation of the classical complement pathway is decreased in Mafb-deficient mice. In addition, MafB overexpression induces C1q-dependent gene expression and signals that induce C1q genes are less effective in the absence of MafB. We also show that Mafb-deficiency can increase glomerular autoimmunity, including anti-nuclear antibody deposition. These results show that MafB is an important regulator of C1q.
Collapse
|
47
|
Bull C, Malipatlolla D, Kalm M, Sjöberg F, Alevronta E, Grandér R, Sultanian P, Persson L, Boström M, Eriksson Y, Swanpalmer J, Wold AE, Blomgren K, Björk-Eriksson T, Steineck G. A novel mouse model of radiation-induced cancer survivorship diseases of the gut. Am J Physiol Gastrointest Liver Physiol 2017; 313:G456-G466. [PMID: 28729245 DOI: 10.1152/ajpgi.00113.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/06/2017] [Accepted: 07/16/2017] [Indexed: 01/31/2023]
Abstract
A deeper understanding of the radiation-induced pathophysiological processes that develop in the gut is imperative to prevent, alleviate, or eliminate cancer survivorship diseases after radiotherapy to the pelvic area. Most rodent models of high-dose gastrointestinal radiation injury are limited by high mortality. We therefore established a model that allows for the delivering of radiation in fractions at high doses while maintaining long-term survival. Adult male C57/BL6 mice were exposed to small-field irradiation, restricted to 1.5 cm of the colorectum using a linear accelerator. Each mouse received 6 or 8 Gy, two times daily in 12-h intervals in two, three, or four fractions. Acute cell death was examined at 4.5 h postirradiation and histological changes at 6 wk postirradiation. Another group was given four fractions of 8 Gy and followed over time for development of visible symptoms. Irradiation caused immediate cell death, mainly limited to the colorectum. At 6 wk postirradiation, several crypts displayed signs of radiation-induced degeneration. The degenerating crypts were seen alongside crypts that appeared perfectly healthy. Crypt survival was reduced after the fourth fraction regardless of dose, whereas the number of macrophages increased. Angiogenesis was induced, likely as a compensatory mechanism for hypoxia. Four months postirradiation, mice began to show radiation-induced symptoms, and histological examination revealed an extensive crypt loss and fibrosis. Our model is uniquely suitable for studying the long-term trajectory and underlying mechanisms of radiation-induced gastrointestinal injury.NEW & NOTEWORTHY A novel mouse model for studying the long-term trajectory of radiation-induced gut injury. The method allows for the use of high doses and multiple fractions, with minor impact on animal health for at least 3 mo. Crypt loss and a slow progression of fibrosis is observed. Crypt degeneration is a process restricted to isolated crypts. Crypt degeneration is presented as a convenient proxy endpoint for long-term radiation-induced gut injury.
Collapse
Affiliation(s)
- Cecilia Bull
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dilip Malipatlolla
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Marie Kalm
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fei Sjöberg
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Eleftheria Alevronta
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rita Grandér
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pedram Sultanian
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linda Persson
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Martina Boström
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yohanna Eriksson
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - John Swanpalmer
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Agnes E Wold
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Thomas Björk-Eriksson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Gunnar Steineck
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden;
| |
Collapse
|
48
|
Zhang H, Yan H, Zhou X, Wang H, Yang Y, Zhang J, Wang H. The protective effects of Resveratrol against radiation-induced intestinal injury. Altern Ther Health Med 2017; 17:410. [PMID: 28814292 PMCID: PMC5559783 DOI: 10.1186/s12906-017-1915-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 08/08/2017] [Indexed: 11/21/2022]
Abstract
Background Intestinal injury is a potential cause of death after high-dose radiation exposure. The aim of the present study was to investigate the protective effects of resveratrol against radiation-induced small intestine injury. Methods C57BL/6 N mice were irradiated and treated with resveratrol and/or Ex527 (a potent Sirt1 inhibitor), and subsequent examining intestinal morphological changes, and crypt cell apoptosis. Then, the expression and enzyme activity of SOD2 in the small intestine were examined. Furthermore, Sirt1 and acetylated p53 expression was analysed. Results Compared to the vehicle control, treatment with resveratrol improved intestinal morphology, decreased apoptosis of crypt cells, maintained cell regeneration, and ameliorated SOD2 expression and activity. Resveratrol also regulated Sirt1 and acetylated p53 expression perturbed by irradiation in the small intestine. The protective effect of resveratrol against ionizing radiation induced small intestine injury was significantly inhibited by Ex527. Conclusion Our results suggest that resveratrol decreases the effects of radiation on intestinal injury at least partly via activation of Sirt1. Electronic supplementary material The online version of this article (doi:10.1186/s12906-017-1915-9) contains supplementary material, which is available to authorized users.
Collapse
|
49
|
Ghrelin Therapy Decreases Incidents of Intracranial Hemorrhage in Mice after Whole-Body Ionizing Irradiation Combined with Burn Trauma. Int J Mol Sci 2017; 18:ijms18081693. [PMID: 28771181 PMCID: PMC5578083 DOI: 10.3390/ijms18081693] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/17/2017] [Accepted: 07/27/2017] [Indexed: 12/30/2022] Open
Abstract
Nuclear industrial accidents and the detonation of nuclear devices cause a variety of damaging factors which, when their impacts are combined, produce complicated injuries challenging for medical treatment. Thus, trauma following acute ionizing irradiation (IR) can deteriorate the IR-induced secondary reactive metabolic and inflammatory impacts to dose-limiting tissues, such as bone marrow/lymphatic, gastrointestinal tissues, and vascular endothelial tissues, exacerbating the severity of the primary injury and decreasing survival from the exposure. Previously we first reported that ghrelin therapy effectively improved survival by mitigating leukocytopenia, thrombocytopenia, and bone-marrow injury resulting from radiation combined with burn trauma. This study was aimed at investigating whether radiation combined with burn trauma induced the cerebro-vascular impairment and intracranial hemorrhage that could be reversed by ghrelin therapy. When B6D2F1 female mice were exposed to 9.5 Gy Cobalt-60 γ-radiation followed by 15% total skin surface burn, cerebro-vascular impairment and intracranial hemorrhage as well as platelet depletion were observed. Ghrelin treatment after irradiation combined with burn trauma significantly decreased platelet depletion and brain hemorrhage. The results suggest that ghrelin treatment is an effective therapy for ionizing radiation combined with burn trauma.
Collapse
|
50
|
Anwar M, Ahmad S, Akhtar R, Mahmood A, Mahmood S. Antioxidant Supplementation: A Linchpin in Radiation-Induced Enteritis. Technol Cancer Res Treat 2017; 16:676-691. [PMID: 28532242 PMCID: PMC5762044 DOI: 10.1177/1533034617707598] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Radiation enteritis is one of the most feared complications of abdominal and pelvic regions. Thus, radiation to abdominal or pelvic malignancies unavoidably injures the intestine. Because of rapid cell turnover, the intestine is highly sensitive to radiation injury, which is the limiting factor in the permissible dosage of irradiation. Bowel injuries such as fistulas, strictures, and chronic malabsorption are potentially life-threatening complications and have an impact on patient quality of life. The incidence of radiation enteritis is increasing because of the current trend of combined chemotherapy and radiation. The consequences of radiation damage to the intestine may result in considerable morbidity and even mortality. The observed effects of ionizing radiation are mediated mainly by oxygen-free radicals that are generated by its action on water and are involved in several steps of signal transduction cascade, leading to apoptosis. The oxyradicals also induce DNA strand breaks and protein oxidation. An important line of defense against free radical damage is the presence of antioxidants. Therefore, administration of antioxidants may ameliorate the radiation-induced damage to the intestine.
Collapse
Affiliation(s)
- Mumtaz Anwar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shabeer Ahmad
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Reyhan Akhtar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Akhtar Mahmood
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Safrun Mahmood
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|