1
|
Gkikoudi A, Manda G, Beinke C, Giesen U, Al-Qaaod A, Dragnea EM, Dobre M, Neagoe IV, Sangsuwan T, Haghdoost S, Vasilopoulos SN, Triantopoulou S, Georgakopoulou A, Tremi I, Koutsoudaki PN, Havaki S, Gorgoulis VG, Kokkoris M, Krasniqi F, Terzoudi GI, Georgakilas AG. Synergistic Effects of UVB and Ionizing Radiation on Human Non-Malignant Cells: Implications for Ozone Depletion and Secondary Cosmic Radiation Exposure. Biomolecules 2025; 15:536. [PMID: 40305266 PMCID: PMC12024869 DOI: 10.3390/biom15040536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
The ozone layer in the Earth's atmosphere filters solar radiation and limits the unwanted effects on humans. A depletion of this ozone shield would permit hazardous levels of UV solar radiation, especially in the UVB range, to bombard Earth's surface, resulting in potentially significant effects on human health. The concern for these adverse effects intensifies if we consider that the UVB solar radiation is combined with secondary cosmic radiation (SCR) components, such as protons and muons, as well as terrestrial gamma rays. This research aims to delve into the intricate interplay between cosmic and solar radiation on earth at the cellular level, focusing on their synergistic effects on human cell biology. Through a multidisciplinary approach integrating radiobiology and physics, we aim to explore key aspects of biological responses, including cell viability, DNA damage, stress gene expression, and finally, genomic instability. To assess the impact of the combined exposure, normal i.e., non-malignant human cells (skin fibroblasts, keratinocytes, monocytes, and lymphocytes) were exposed to high-energy protons or gamma rays in combination with UVB. Cellular molecular and cytogenetic biomarkers of radiation exposure, such as DNA damage (γH2AΧ histone protein and dicentric chromosomes), as well as the expression pattern of various stress genes, were analyzed. In parallel, the MTS reduction and lactate dehydrogenase assays were used as indicators of cell viability, proliferation, and cytotoxicity. Results reveal remaining DNA damage for the co-exposed samples compared to samples exposed to only one type of radiation in all types of cells, accompanied by increased genomic instability and distinct stress gene expression patterns detected at 24-48 h post-exposure. Understanding the impact of combined radiation exposures is crucial for assessing the health risks posed to humans if the ozone layer is partially depleted, with structural and functional damages inflicted by combined cosmic and UVB exposure.
Collapse
Affiliation(s)
- Angeliki Gkikoudi
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campus, 15780 Athens, Greece; (A.G.); (S.N.V.); (A.G.); (I.T.)
- Laboratory of Health Physics, Radiobiology & Cytogenetics, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, 15341 Agia Paraskevi, Greece; (S.T.); (G.I.T.)
| | - Gina Manda
- Radiobiology Laboratory, “Victor Babeș” National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (G.M.); (E.-M.D.); (M.D.); (I.V.N.)
| | - Christina Beinke
- Bundeswehr Institute of Radiobiology, University of Ulm, Neuherbergstraβe 11, 80937 Munich, Germany;
| | - Ulrich Giesen
- Physikalisch-Technische Bundesanstalt (PTB), Bundesallee 100, 38116 Braunschweig, Germany (A.A.-Q.); (F.K.)
| | - Amer Al-Qaaod
- Physikalisch-Technische Bundesanstalt (PTB), Bundesallee 100, 38116 Braunschweig, Germany (A.A.-Q.); (F.K.)
| | - Elena-Mihaela Dragnea
- Radiobiology Laboratory, “Victor Babeș” National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (G.M.); (E.-M.D.); (M.D.); (I.V.N.)
| | - Maria Dobre
- Radiobiology Laboratory, “Victor Babeș” National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (G.M.); (E.-M.D.); (M.D.); (I.V.N.)
| | - Ionela Victoria Neagoe
- Radiobiology Laboratory, “Victor Babeș” National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (G.M.); (E.-M.D.); (M.D.); (I.V.N.)
| | - Traimate Sangsuwan
- ABTE/ToxEMAC Laboratory, University of Caen Normandy, F-14050 Caen, France; (T.S.); (S.H.)
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden
| | - Siamak Haghdoost
- ABTE/ToxEMAC Laboratory, University of Caen Normandy, F-14050 Caen, France; (T.S.); (S.H.)
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden
| | - Spyridon N. Vasilopoulos
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campus, 15780 Athens, Greece; (A.G.); (S.N.V.); (A.G.); (I.T.)
| | - Sotiria Triantopoulou
- Laboratory of Health Physics, Radiobiology & Cytogenetics, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, 15341 Agia Paraskevi, Greece; (S.T.); (G.I.T.)
| | - Anna Georgakopoulou
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campus, 15780 Athens, Greece; (A.G.); (S.N.V.); (A.G.); (I.T.)
| | - Ioanna Tremi
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campus, 15780 Athens, Greece; (A.G.); (S.N.V.); (A.G.); (I.T.)
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.K.); (S.H.); (V.G.G.)
| | - Paraskevi N. Koutsoudaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.K.); (S.H.); (V.G.G.)
| | - Sophia Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.K.); (S.H.); (V.G.G.)
| | - Vassilis G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.N.K.); (S.H.); (V.G.G.)
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Ninewells Hospital and Medical School, University of Dundee, Dundee DD2 1SG, UK
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7YH, UK
| | - Michael Kokkoris
- Group of Nuclear Physics, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), 15780 Zografou, Greece;
| | - Faton Krasniqi
- Physikalisch-Technische Bundesanstalt (PTB), Bundesallee 100, 38116 Braunschweig, Germany (A.A.-Q.); (F.K.)
| | - Georgia I. Terzoudi
- Laboratory of Health Physics, Radiobiology & Cytogenetics, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, 15341 Agia Paraskevi, Greece; (S.T.); (G.I.T.)
| | - Alexandros G. Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campus, 15780 Athens, Greece; (A.G.); (S.N.V.); (A.G.); (I.T.)
| |
Collapse
|
2
|
Galal SM, El Kiki SM, Elgazzar EM. The Potential Therapeutic Approach of Ursodeoxycholic Acid as a Potent Activator of ACE-2 on Cerebral Disorders Induced by γ-irradiation in Rats. Cell Biochem Funct 2024; 42:e70024. [PMID: 39660593 DOI: 10.1002/cbf.70024] [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: 06/12/2024] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
The present investigation assesses ursodeoxycholic acid's efficacy (UDCA) as an ACE2 activator against gamma irradiation through activating the renin-angiotensin system's (RAS) beneficial axis, ACE2/Ang-(1-7)/Mas1 via its profitable influence on inflammation, oxidative stress, and neuronal damage caused by irradiation (IRR). Four groups of rats were treated as follows: control group, group receiving UDCA (100 mg/kg/day) for 14 days by gavage, group irradiated at 6 Gy, and group receiving UDCA post-irradiation for 14 days. The results revealed that gamma-irradiation (6 Gy) caused a substantial drop in the cerebral ACE2/Ang-(1-7)/Mas1 axis and remarkably increased the expression of cerebral inflammatory mediators: tumor necrosis factor-α (TNF-α), nuclear factor kappa-B (NF-κB), interleukin-6 (IL-6) and interleukin-1β (IL-1β) combined with significant elevation in cyclooxygenase-II (COX-II), (NADPH) oxidases (NOX4), lipooxygenase (LOX) activities and nitric oxide (NO) content. Moreover, it greatly enhanced the reduction in N-methyl-d-aspartate (NMDA) level, while dramatically increasing gamma-aminobutyric acid (GABA) level and neuronal nitric oxide synthases (nNOS) enzyme activity in cerebral tissue homogenate. Irradiated rats' brain sections underwent histological investigation using hematoxylin and eosin staining, which revealed cellular damage and a pathological appearance. The administration of UDCA inverts these unusual alterations. In conclusion, UDCA treatment efficiently normalizes the above-mentioned pathological abnormalities and avoids the development of IRR-associated neurological dysfunction by upregulating the beneficial axis of RAS in the brain. Hence, ursodeoxycholic acid presents a novel option for patient care during radiotherapy.
Collapse
Affiliation(s)
- Shereen Mohamed Galal
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Shereen Mohamed El Kiki
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Eman Mahmoud Elgazzar
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
3
|
Cui J, Wang TJ, Zhang YX, She LZ, Zhao YC. Molecular biological mechanisms of radiotherapy-induced skin injury occurrence and treatment. Biomed Pharmacother 2024; 180:117470. [PMID: 39321513 DOI: 10.1016/j.biopha.2024.117470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024] Open
Abstract
Radiotherapy-Induced Skin Injury (RISI) is radiation damage to normal skin tissue that primarily occurs during tumor Radiotherapy and occupational exposure. The risk of RISI is high due to the fact that the skin is not only the first body organ that ionizing radiation comes into contact with, but it is also highly sensitive to it, especially the basal cell layer and capillaries. Typical clinical manifestations of RISI include erythema, dry desquamation, moist desquamation, and ulcers, which have been established to significantly impact patient care and cancer treatment. Notably, our current understanding of RISI's pathological mechanisms and signaling pathways is inadequate, and no standard treatments have been established. Radiation-induced oxidative stress, inflammatory responses, fibrosis, apoptosis, and cellular senescence are among the known mechanisms that interact and promote disease progression. Additionally, radiation can damage all cellular components and induce genetic and epigenetic changes, which play a crucial role in the occurrence and progression of skin injury. A deeper understanding of these mechanisms and pathways is crucial for exploring the potential therapeutic targets for RISI. Therefore, in this review, we summarize the key mechanisms and potential treatment methods for RISI, offering a reference for future research and development of treatment strategies.
Collapse
Affiliation(s)
- Jie Cui
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| | - Tie-Jun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| | - Yu-Xuan Zhang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| | - Li-Zhen She
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| | - Yue-Chen Zhao
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin 130022, China.
| |
Collapse
|
4
|
Li S, Chen K, Sun Z, Chen M, Pi W, Zhou S, Yang H. Radiation drives tertiary lymphoid structures to reshape TME for synergized antitumour immunity. Expert Rev Mol Med 2024; 26:e30. [PMID: 39438247 PMCID: PMC11505612 DOI: 10.1017/erm.2024.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 04/24/2024] [Accepted: 07/15/2024] [Indexed: 10/25/2024]
Abstract
Radiotherapy (RT) plays a key role in the tumour microenvironment (TME), impacting the immune response via cellular and humoral immunity. RT can induce local immunity to modify the TME. It can stimulate dendritic cell maturation and T-cell infiltration. Moreover, B cells, macrophages and other immune cells may also be affected. Tertiary lymphoid structure (TLS) is a unique structure within the TME and a class of aggregates containing T cells, B cells and other immune cells. The maturation of TLS is determined by the presence of mature dendritic cells, the density of TLS is determined by the number of immune cells. TLS maturation and density both affect the antitumour immune response in the TME. This review summarized the recent research on the impact and the role of RT on TLS, including the changes of TLS components and formation conditions and the mechanism of how RT affects TLS and transforms the TME. RT may promote TLS maturation and density to modify the TME regarding enhanced antitumour immunity.
Collapse
Affiliation(s)
- Shuling Li
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Kuifei Chen
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Zhenwei Sun
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Meng Chen
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Haihua Yang
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
5
|
Saleh DS, Hussain HS, Al-Haidari HN, Abbas SK, Zgair AK, Ali SM. Serum Level Alteration of IL-6, IL-1 β, and IFN- γ in Groups of Healthy Adults with Oxidative DNA Damage in Najaf Governorate. ScientificWorldJournal 2024; 2024:9048536. [PMID: 39257966 PMCID: PMC11383644 DOI: 10.1155/2024/9048536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024] Open
Abstract
Background Najaf governorate was recorded as one of the most polluted Iraqi governorates with increased cancer, autoimmune, and abortion cases. Study Groups. A total of 88 adult volunteers from three test groups were divided based on their inhabitance in different geographical regions in Najaf governorate. Group 1 (G1; n, 29) inhabitants of Al-Ansar, Al-Abbaseyeh, and Al-Manathera districts, Group 2 (G2; n, 27) inhabitants of 22 different scattered districts of the governorate, Group 3 (G3; n, 32) inhabitants of Kufa city and center districts in the old Najaf city. According to previous authors' findings, all participants had uranium contamination in their urine and blood samples, and also, they had DNA damage according to the level of urinary 8-OHdG compound. The control group 4 (G4; n, 25) were adult healthy Iraqi volunteers who were residents of the Sulaimaniyah governorate, which has low-level uranium pollution. The present study aims to determine the effect of uranium pollution and DNA damage on the immune system function in terms of estimating the levels of serum interleukin (IL)-6, interferon-gamma (IFN-γ), and IL-1 beta (β). Method Enzyme-linked immunosorbent assay (ELISA) (Sandwich method technique) was used for estimating the serum cytokines levels in test and control groups. Results A significant elevation of cytokines levels was reported as compared with the control groups (p ≤ 0.01). The level of IL-6 was 764.64 ± 24.12 pg/ml, 768.87 ± 19.64 pg/ml, and 735.62 ± 18.47 in G1, G2, and G3, respectively. The level of IFN-γ was 264.55 ± 19.17 pg/ml, 259 ± 18.76 pg/ml, and 261.20 ± 12.99 pg/ml for G1, G2, and G3, respectively. The level of IL-1β was 99.85 ± 10.81 pg/ml, 116.8 ± 10.71 pg/ml, and 83 ± 19.24 pg/ml in G1, G2, and G3, respectively. The levels of IL-6, IFN-γ, and IL-1β were 86.5 ± 22.9 pg/ml, 19.4 ± 2.8 pg/ml, and 16.1 ± 3.2 pg/ml in the sera of control (G4). The results showed significant statistical elevation with the corresponding p value cut-off p ≤ 0.01 in IL-6, IFN-γ, and IL-1β in the sera of three test groups as compared with the results of the control group. Conclusion The change in the proinflammatory cytokines (IL-6, IFN-γ, and IL-1β) levels indicates a persistent inflammatory response in the participants and may reflect immune system impairment as a consequence of exposure to long-term low-dose ionizing radiation.
Collapse
Affiliation(s)
- Dhuha S. Saleh
- Department of BiologyCollege of ScienceUniversity of Baghdad, Baghdad, Iraq
| | - Hayder S. Hussain
- Department of PhysicsCollege of ScienceUniversity of Baghdad, Baghdad, Iraq
| | - Hasan N. Al-Haidari
- Department of RadiologyKing Hussien Medical CenterJordanian Royal Medical Services, Amman, Jordan
| | - Samia K. Abbas
- General Directorate of Education in NajafMinistry of Education, Dahuk, Iraq
| | - Ayaid K. Zgair
- Department of BiologyCollege of ScienceUniversity of Baghdad, Baghdad, Iraq
| | - Seenaa M. Ali
- Laboratory DepartmentCollege of Health and Medical TechnologySulaimani Polytechnic University, Sulaymaniyah, Iraq
| |
Collapse
|
6
|
Rostami Nejad M, Bandarian F, Razi F, Razzaghi Z, Robati RM, Rezaei M, Arjmand B, Rezaei-Tavirani M, Hamzeloo-Moghadam M. Evaluation of Laser Intensity Effect on Photodynamic Therapy Efficacy. J Lasers Med Sci 2024; 15:e33. [PMID: 39193105 PMCID: PMC11348446 DOI: 10.34172/jlms.2024.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/08/2024] [Indexed: 08/29/2024]
Abstract
Introduction: Intensity is one of the important parameters of laser radiation in photodynamic therapy. Effective treatment requires the selection of a suitable power of laser. This study aimed to evaluate laser effectiveness in photodynamic therapy via high and low intensity by the analysis of the gene expression profiles of the treated cells. Methods: The gene expression profiles of human SK-ChA-1 cells which are treated by 500mW and 50mW laser radiation were retrieved from the Gene Expression Omnibus (GEO) database. Data were assessed by the GEO2R program, and the significant differentially expressed genes (DEGs) were investigated via expression examination and protein-protein interaction (PPI) network analysis. Results: Analyses revealed that the higher intensity of radiation is associated with wide gene expression changes relative to the lower mode. 196 significant DEGs were identified and assessed. The extremely dysregulated DEGs except MMP1 were down-regulated. STAT1, IRF7, IL1B, DDX58, ISG15, RSAD2, DHX58, OASL, OAS1, STAT2, DDX60, OAS2, USP18, and IFI44L were introduced as hubs of the main component of the PPI network. Final analysis showed that STAT1, IRF7, IL1B, DDX58, and STAT2 are the critical DEGs. Conclusion: Compared to the 50 mW mode of radiation, 500 mW laser intensity effectively changed apoptosis, differentiation, cell proliferation and angiogenesis, regulation of other inflammation-related molecules, innate immunity, and maintaining immune homeostasis.
Collapse
Affiliation(s)
- Mohammad Rostami Nejad
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bandarian
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Razi
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Razzaghi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza M Robati
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mitra Rezaei
- Genomic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Iranian Cancer Control Center (MACSA), Tehran, Iran
| | | | - Maryam Hamzeloo-Moghadam
- Traditional Medicine and Materia Medica Research Center, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Ran B, Ren X, Lin X, Teng Y, Xin F, Ma W, Zhao X, Li M, Wang J, Wang C, Sun L, Zhang J. Glycyrrhetinic acid loaded in milk-derived extracellular vesicles for inhalation therapy of idiopathic pulmonary fibrosis. J Control Release 2024; 370:811-820. [PMID: 38754632 DOI: 10.1016/j.jconrel.2024.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and life-threatening lung disease for which treatment options are limited. Glycyrrhetinic acid (GA) is a triterpenoid with multiple biological effects, such as anti-inflammatory and anti-fibrotic properties. Herein, inhalable milk-derived extracellular vesicles (mEVs) encapsulating GA (mEVs@GA) were screened and evaluated for IPF treatment. The results indicated that the loading efficiency of GA in mEVs@GA was 8.65%. Therapeutic effects of inhalable mEVs@GA were investigated in vitro and in vivo. The mEVs@GA demonstrated superior anti-inflammatory effects on LPS-stimulated MHS cells. Furthermore, repeated noninvasive inhalation delivery of mEVs@GA in bleomycin-induced IPF mice could decrease the levels of transforming growth factors β1 (TGF-β1), Smad3 and inflammatory cytokines IL-6, IL-1β and TNF-α. The mEVs@GA effectively diminished the development of fibrosis and improved pulmonary function in the IPF mice model at a quarter of the dose compared with the pirfenidone oral administration group. Additionally, compared to pirfenidone-loaded mEVs, mEVs@GA demonstrated superior efficacy at the same drug concentration in the pharmacodynamic study. Overall, inhaled mEVs@GA have the potential to serve as an effective therapeutic option in the treatment of IPF.
Collapse
Affiliation(s)
- Bo Ran
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Xiaohong Ren
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Xueyuan Lin
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Yupu Teng
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Fangyuan Xin
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Wuzhen Ma
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiangyu Zhao
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Mingwei Li
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Jinghuang Wang
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Caifen Wang
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Lixin Sun
- Shenyang Pharmaceutiacal University, Shenyang 110016, China.
| | - Jiwen Zhang
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China.
| |
Collapse
|
8
|
Miaskowski C, Conley YP, Cooper BA, Paul SM, Smoot BJ, Hammer MJ, Fu M, Levine JD. Identification Of A Higher Risk Lymphedema Phenotype And Associations With Cytokine Gene Polymorphisms. J Pain Symptom Manage 2024; 67:375-383.e3. [PMID: 38307372 DOI: 10.1016/j.jpainsymman.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/04/2024]
Abstract
CONTEXT Breast cancer-related lymphedema (BCRL) is chronic condition that occurs in 5% to 75% of women following treatment for breast cancer. However, little is known about the risk factors and mechanisms associated with a worse BCRL profile. OBJECTIVES Identify distinct BCRL profiles in women with the condition (i.e., lower vs. higher risk phenotype) and evaluate for associations with pro- and anti-inflammatory genes. METHODS Latent class profile analysis (LCPA) was used to identify the BCRL profiles using phenotypic characteristics evaluated prior to surgery. Candidate gene analyses were done to identify cytokine genes associated with the two BCRL profiles. RESULTS Of the 155 patients evaluated, 35.5% (n = 55) were in the Lower and 64.5% (n = 100) were in the Higher Risk classes. Risk factors for membership in the Higher class included: lower functional status, having sentinel lymph node biopsy, axillary lymph node dissection, mastectomy, higher number of positive lymph nodes, and receipt of chemotherapy. Polymorphisms for interleukin (IL)1-beta and IL6 were associated with membership in the Higher Risk class. CONCLUSION The readily available and clinically relevant phenotypic characteristics associated with a worse BCRL profile can be used by clinicians to identify higher risk patients. If confirmed, these characteristics can be tested in predictive risk models. In addition, the candidate gene findings may guide the development of mechanistically-based interventions to decrease the risk of BCRL.
Collapse
Affiliation(s)
- Christine Miaskowski
- School of Nursing, University of California, San Francisco, CA, USA; School of Medicine, University of California, San Francisco, CA, USA.
| | | | - Bruce A Cooper
- School of Nursing, University of California, San Francisco, CA, USA
| | - Steven M Paul
- School of Nursing, University of California, San Francisco, CA, USA
| | - Betty J Smoot
- School of Medicine, University of California, San Francisco, CA, USA
| | | | - Mei Fu
- School of Nursing and Health Studies, University of Missouri, Kansas City, MO, USA
| | - Jon D Levine
- School of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
9
|
Farr NTH, Workman VL, Saad S, Roman S, Hearnden V, Chapple CR, Murdoch C, Rodenburg C, MacNeil S. Uncovering the relationship between macrophages and polypropylene surgical mesh. BIOMATERIALS ADVANCES 2024; 159:213800. [PMID: 38377947 DOI: 10.1016/j.bioadv.2024.213800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/06/2024] [Accepted: 02/11/2024] [Indexed: 02/22/2024]
Abstract
Currently, in vitro testing examines the cytotoxicity of biomaterials but fails to consider how materials respond to mechanical forces and the immune response to them; both are crucial for successful long-term implantation. A notable example of this failure is polypropylene mid-urethral mesh used in the treatment of stress urinary incontinence (SUI). The mesh was largely successful in abdominal hernia repair but produced significant complications when repurposed to treat SUI. Developing more physiologically relevant in vitro test models would allow more physiologically relevant data to be collected about how biomaterials will interact with the body. This study investigates the effects of mechanochemical distress (a combination of oxidation and mechanical distention) on polypropylene mesh surfaces and the effect this has on macrophage gene expression. Surface topology of the mesh was characterised using SEM and AFM; ATR-FTIR, EDX and Raman spectroscopy was applied to detect surface oxidation and structural molecular alterations. Uniaxial mechanical testing was performed to reveal any bulk mechanical changes. RT-qPCR of selected pro-fibrotic and pro-inflammatory genes was carried out on macrophages cultured on control and mechanochemically distressed PP mesh. Following exposure to mechanochemical distress the mesh surface was observed to crack and craze and helical defects were detected in the polymer backbone. Surface oxidation of the mesh was seen after macrophage attachment for 7 days. These changes in mesh surface triggered modified gene expression in macrophages. Pro-fibrotic and pro-inflammatory genes were upregulated after macrophages were cultured on mechanochemically distressed mesh, whereas the same genes were down-regulated in macrophages exposed to control mesh. This study highlights the relationship between macrophages and polypropylene surgical mesh, thus offering more insight into the fate of an implanted material than existing in vitro testing.
Collapse
Affiliation(s)
- Nicholas T H Farr
- Department of Materials Science and Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK; Insigneo Institute for in silico Medicine, The Pam Liversidge Building, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK.
| | - Victoria L Workman
- Department of Materials Science and Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK; Insigneo Institute for in silico Medicine, The Pam Liversidge Building, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK
| | - Sanad Saad
- Department of Materials Science and Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK; Department of Urology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Sabiniano Roman
- Department of Materials Science and Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK
| | - Vanessa Hearnden
- Department of Materials Science and Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK; Insigneo Institute for in silico Medicine, The Pam Liversidge Building, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK
| | | | - Craig Murdoch
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield, UK
| | - Cornelia Rodenburg
- Department of Materials Science and Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK; Insigneo Institute for in silico Medicine, The Pam Liversidge Building, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK
| | - Sheila MacNeil
- Department of Materials Science and Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, UK
| |
Collapse
|
10
|
Fijardo M, Kwan JYY, Bissey PA, Citrin DE, Yip KW, Liu FF. The clinical manifestations and molecular pathogenesis of radiation fibrosis. EBioMedicine 2024; 103:105089. [PMID: 38579363 PMCID: PMC11002813 DOI: 10.1016/j.ebiom.2024.105089] [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: 01/08/2024] [Revised: 02/25/2024] [Accepted: 03/12/2024] [Indexed: 04/07/2024] Open
Abstract
Advances in radiation techniques have enabled the precise delivery of higher doses of radiotherapy to tumours, while sparing surrounding healthy tissues. Consequently, the incidence of radiation toxicities has declined, and will likely continue to improve as radiotherapy further evolves. Nonetheless, ionizing radiation elicits tissue-specific toxicities that gradually develop into radiation-induced fibrosis, a common long-term side-effect of radiotherapy. Radiation fibrosis is characterized by an aberrant wound repair process, which promotes the deposition of extensive scar tissue, clinically manifesting as a loss of elasticity, tissue thickening, and organ-specific functional consequences. In addition to improving the existing technologies and guidelines directing the administration of radiotherapy, understanding the pathogenesis underlying radiation fibrosis is essential for the success of cancer treatments. This review integrates the principles for radiotherapy dosimetry to minimize off-target effects, the tissue-specific clinical manifestations, the key cellular and molecular drivers of radiation fibrosis, and emerging therapeutic opportunities for both prevention and treatment.
Collapse
Affiliation(s)
- Mackenzie Fijardo
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Yin Yee Kwan
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | | - Deborah E Citrin
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, United States of America
| | - Kenneth W Yip
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Fei-Fei Liu
- Research Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Hannon G, Lesch ML, Gerber SA. Harnessing the Immunological Effects of Radiation to Improve Immunotherapies in Cancer. Int J Mol Sci 2023; 24:7359. [PMID: 37108522 PMCID: PMC10138513 DOI: 10.3390/ijms24087359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Ionizing radiation (IR) is used to treat 50% of cancers. While the cytotoxic effects related to DNA damage with IR have been known since the early 20th century, the role of the immune system in the treatment response is still yet to be fully determined. IR can induce immunogenic cell death (ICD), which activates innate and adaptive immunity against the cancer. It has also been widely reported that an intact immune system is essential to IR efficacy. However, this response is typically transient, and wound healing processes also become upregulated, dampening early immunological efforts to overcome the disease. This immune suppression involves many complex cellular and molecular mechanisms that ultimately result in the generation of radioresistance in many cases. Understanding the mechanisms behind these responses is challenging as the effects are extensive and often occur simultaneously within the tumor. Here, we describe the effects of IR on the immune landscape of tumors. ICD, along with myeloid and lymphoid responses to IR, are discussed, with the hope of shedding light on the complex immune stimulatory and immunosuppressive responses involved with this cornerstone cancer treatment. Leveraging these immunological effects can provide a platform for improving immunotherapy efficacy in the future.
Collapse
Affiliation(s)
- Gary Hannon
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Maggie L. Lesch
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Scott A. Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
12
|
Wang K, Wen D, Xu X, Zhao R, Jiang F, Yuan S, Zhang Y, Gao Y, Li Q. Extracellular matrix stiffness-The central cue for skin fibrosis. Front Mol Biosci 2023; 10:1132353. [PMID: 36968277 PMCID: PMC10031116 DOI: 10.3389/fmolb.2023.1132353] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Skin fibrosis is a physiopathological process featuring the excessive deposition of extracellular matrix (ECM), which is the main architecture that provides structural support and constitutes the microenvironment for various cellular behaviors. Recently, increasing interest has been drawn to the relationship between the mechanical properties of the ECM and the initiation and modulation of skin fibrosis, with the engagement of a complex network of signaling pathways, the activation of mechanosensitive proteins, and changes in immunoregulation and metabolism. Simultaneous with the progression of skin fibrosis, the stiffness of ECM increases, which in turn perturbs mechanical and humoral homeostasis to drive cell fate toward an outcome that maintains and enhances the fibrosis process, thus forming a pro-fibrotic "positive feedback loop". In this review, we highlighted the central role of the ECM and its dynamic changes at both the molecular and cellular levels in skin fibrosis. We paid special attention to signaling pathways regulated by mechanical cues in ECM remodeling. We also systematically summarized antifibrotic interventions targeting the ECM, hopefully enlightening new strategies for fibrotic diseases.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dongsheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuewen Xu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zhao
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Feipeng Jiang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Shengqin Yuan
- School of Public Administration, Sichuan University, Chengdu, Sichuan, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| |
Collapse
|
13
|
Kim JM, Kim H, Oh SH, Jang WI, Lee SB, Park M, Kim S, Park S, Shim S, Jang H. Combined Administration of Pravastatin and Metformin Attenuates Acute Radiation-Induced Intestinal Injury in Mouse and Minipig Models. Int J Mol Sci 2022; 23:ijms232314827. [PMID: 36499155 PMCID: PMC9739896 DOI: 10.3390/ijms232314827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
Radiation-induced gastrointestinal (GI) damage is one of the critical factors that serve as basis for the lethality of nuclear accidents or terrorism. Further, there are no Food and Drug Administration-approved agents available to mitigate radiation-induced intestinal injury. Although pravastatin (PS) has been shown to exhibit anti-inflammatory and epithelial reconstructive effects following radiation exposure using mouse and minipig models, the treatment failed to improve the survival rate of high-dose irradiated intestinal injury. Moreover, we previously found that metformin (MF), a common drug used for treating type 2 diabetes mellitus, has a mitigating effect on radiation-induced enteropathy by promoting stem cell properties. In this study, we investigated whether the combined administration of PS and MF could achieve therapeutic effects on acute radiation-induced intestinal injury in mouse and minipig models. We found that the combined treatment markedly increased the survival rate and attenuated histological damage in a radiation-induced intestinal injury mouse model, in addition to epithelial barrier recovery, anti-inflammatory effects, and improved epithelial proliferation with stem cell properties. Furthermore, in minipig models, combined treatment with PS and MF ameliorates gross pathological damage in abdominal organs and attenuated radiation-induced intestinal histological damage. Therefore, the combination of PS and MF effectively alleviated radiation-induced intestinal injury in the mouse and minipig models. We believe that the combined use of PS and MF is a promising therapeutic approach for treating radiation-induced intestinal injury.
Collapse
Affiliation(s)
- Jung Moon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Su Hyun Oh
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Won Il Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Mineon Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Soyeon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| | - Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| |
Collapse
|
14
|
Garcia AN, Casanova NG, Kempf CL, Bermudez T, Valera DG, Song JH, Sun X, Cai H, Moreno-Vinasco L, Gregory T, Oita RC, Hernon VR, Camp SM, Rogers C, Kyubwa EM, Menon N, Axtelle J, Rappaport J, Bime C, Sammani S, Cress AE, Garcia JGN. eNAMPT Is a Novel Damage-associated Molecular Pattern Protein That Contributes to the Severity of Radiation-induced Lung Fibrosis. Am J Respir Cell Mol Biol 2022; 66:497-509. [PMID: 35167418 PMCID: PMC9116358 DOI: 10.1165/rcmb.2021-0357oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/17/2021] [Indexed: 11/24/2022] Open
Abstract
The paucity of therapeutic strategies to reduce the severity of radiation-induced lung fibrosis (RILF), a life-threatening complication of intended or accidental ionizing radiation exposure, is a serious unmet need. We evaluated the contribution of eNAMPT (extracellular nicotinamide phosphoribosyltransferase), a damage-associated molecular pattern (DAMP) protein and TLR4 (Toll-like receptor 4) ligand, to the severity of whole-thorax lung irradiation (WTLI)-induced RILF. Wild-type (WT) and Nampt+/- heterozygous C57BL6 mice and nonhuman primates (NHPs, Macaca mulatta) were exposed to a single WTLI dose (9.8 or 10.7 Gy for NHPs, 20 Gy for mice). WT mice received IgG1 (control) or an eNAMPT-neutralizing polyclonal or monoclonal antibody (mAb) intraperitoneally 4 hours after WTLI and weekly thereafter. At 8-12 weeks after WTLI, NAMPT expression was assessed by immunohistochemistry, biochemistry, and plasma biomarker studies. RILF severity was determined by BAL protein/cells, hematoxylin and eosin, and trichrome blue staining and soluble collagen assays. RNA sequencing and bioinformatic analyses identified differentially expressed lung tissue genes/pathways. NAMPT lung tissue expression was increased in both WTLI-exposed WT mice and NHPs. Nampt+/- mice and eNAMPT polyclonal antibody/mAb-treated mice exhibited significantly attenuated WTLI-mediated lung fibrosis with reduced: 1) NAMPT and trichrome blue staining; 2) dysregulated lung tissue expression of smooth muscle actin, p-SMAD2/p-SMAD1/5/9, TGF-β, TSP1 (thrombospondin-1), NOX4, IL-1β, and NRF2; 3) plasma eNAMPT and IL-1β concentrations; and 4) soluble collagen. Multiple WTLI-induced dysregulated differentially expressed lung tissue genes/pathways with known tissue fibrosis involvement were each rectified in mice receiving eNAMPT mAbs.The eNAMPT/TLR4 inflammatory network is essentially involved in radiation pathobiology, with eNAMPT neutralization an effective therapeutic strategy to reduce RILF severity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hua Cai
- Department of Anesthesiology, University of California Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | | | | - Jay Rappaport
- Tulane National Primate Research Center, New Orleans, Louisiana
| | | | | | - Anne E. Cress
- Department of Cell and Molecular Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | | |
Collapse
|
15
|
Artlett CM. The Mechanism and Regulation of the NLRP3 Inflammasome during Fibrosis. Biomolecules 2022; 12:biom12050634. [PMID: 35625564 PMCID: PMC9138796 DOI: 10.3390/biom12050634] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Fibrosis is often the end result of chronic inflammation. It is characterized by the excessive deposition of extracellular matrix. This leads to structural alterations in the tissue, causing permanent damage and organ dysfunction. Depending on the organ it effects, fibrosis can be a serious threat to human life. The molecular mechanism of fibrosis is still not fully understood, but the NLRP3 (NOD-, LRR- and pyrin–domain–containing protein 3) inflammasome appears to play a significant role in the pathogenesis of fibrotic disease. The NLRP3 inflammasome has been the most extensively studied inflammatory pathway to date. It is a crucial component of the innate immune system, and its activation mediates the secretion of interleukin (IL)-1β and IL-18. NLRP3 activation has been strongly linked with fibrosis and drives the differentiation of fibroblasts into myofibroblasts by the chronic upregulation of IL-1β and IL-18 and subsequent autocrine signaling that maintains an activated inflammasome. Both IL-1β and IL-18 are profibrotic, however IL-1β can have antifibrotic capabilities. NLRP3 responds to a plethora of different signals that have a common but unidentified unifying trigger. Even after 20 years of extensive investigation, regulation of the NLRP3 inflammasome is still not completely understood. However, what is known about NLRP3 is that its regulation and activation is complex and not only driven by various activators but controlled by numerous post-translational modifications. More recently, there has been an intensive attempt to discover NLRP3 inhibitors to treat chronic diseases. This review addresses the role of the NLRP3 inflammasome in fibrotic disorders across many different tissues. It discusses the relationships of various NLRP3 activators to fibrosis and covers different therapeutics that have been developed, or are currently in development, that directly target NLRP3 or its downstream products as treatments for fibrotic disorders.
Collapse
Affiliation(s)
- Carol M Artlett
- Department of Microbiology & Immunology, College of Medicine, Drexel University, Philadelphia, PA 19129, USA
| |
Collapse
|
16
|
Fakhri S, Piri S, Moradi SZ, Khan H. Phytochemicals Targeting Oxidative Stress, Interconnected Neuroinflammatory, and Neuroapoptotic Pathways Following Radiation. Curr Neuropharmacol 2022; 20:836-856. [PMID: 34370636 PMCID: PMC9881105 DOI: 10.2174/1570159x19666210809103346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/19/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
The radiation for therapeutic purposes has shown positive effects in different contexts; however, it can increase the risk of many age-related and neurodegenerative diseases such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and Parkinson's disease (PD). These different outcomes highlight a dose-response phenomenon called hormesis. Prevailing studies indicate that high doses of radiation could play several destructive roles in triggering oxidative stress, neuroapoptosis, and neuroinflammation in neurodegeneration. However, there is a lack of effective treatments in combating radiation-induced neurodegeneration, and the present drugs suffer from some drawbacks, including side effects and drug resistance. Among natural entities, polyphenols are suggested as multi-target agents affecting the dysregulated pathogenic mechanisms in neurodegenerative disease. This review discusses the destructive effects of radiation on the induction of neurodegenerative diseases by dysregulating oxidative stress, apoptosis, and inflammation. We also describe the promising effects of polyphenols and other candidate phytochemicals in preventing and treating radiation-induced neurodegenerative disorders, aiming to find novel/potential therapeutic compounds against such disorders.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;,Address correspondence to these author at the Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; E-mail: Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan; E-mail:
| | - Sana Piri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;,These authors have contributed equally to this work.
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;,These authors have contributed equally to this work.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan,Address correspondence to these author at the Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; E-mail: Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan; E-mail:
| |
Collapse
|
17
|
Zeng X, Xie L, Ge Y, Zhou Y, Wang H, Chen Y, Zhu X, Liu H, Liao Q, Kong Y, Pan L, Li J, Xue L, Li S, Zhou X, Shi C, Sheng X. Satellite Cells are Activated in a Rat Model of Radiation-Induced Muscle Fibrosis. Radiat Res 2022; 197:638-649. [PMID: 35294551 DOI: 10.1667/rade-21-00183.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
Radiation-induced muscle fibrosis is a long-term side effect of radiotherapy that significantly affects the quality of life and even reduces the survival of cancer patients. We have demonstrated that radiation induces satellite cell (SC) activation at the molecular level; however, cellular evidence in a rat model of radiation-induced muscle fibrosis was lacking. In this study, we evaluated SC activation in vivo and investigated whether radiation affects the proliferation and differentiation potential of SCs in vitro. For in vivo studies, Sprague-Dawley rats were randomly divided into six groups (n = 6 per group): non-irradiated controls, 90 Gy/1 week-, 90 Gy/2 weeks-, 90 Gy/4 weeks-, 90 Gy/12 weeks- and 90 Gy/24 weeks-postirradiation groups. Rats received a single dose of radiation in the left groin area and rectus femoris tissues were collected in the indicated weeks. Fibrosis, apoptosis, and autophagy were evaluated by Masson's trichrome staining, TUNEL staining, and electron microscopy, respectively. SC activation and central nuclear muscle fibers were evaluated by immunofluorescence staining and hematoxylin and eosin staining. IL-1β concentrations in serum and irradiated muscle tissue samples were determined by ELISA. For in vitro studies, SCs were isolated from rats with radiation-induced muscle fibrosis and their proliferation and differentiation were evaluated by immunofluorescence staining. In vivo, fibrosis increased over time postirradiation. Apoptosis and autophagy levels, IL-1β concentrations in serum and irradiated skin tissues, and the numbers of SCs and central nuclear muscle fibers were increased in the irradiated groups when compared with the control group. In vitro, cultured SCs from irradiated muscle were positive for the proliferation marker Pax7, and differentiated SCs were positive for the myogenic differentiation marker MyHC. This study provided cellular evidence of SC activation and proliferation in rats with radiation-induced muscle fibrosis.
Collapse
Affiliation(s)
- Xiaoling Zeng
- Graduate Collaborative Training of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Department of Head and Neck Surgery, Central laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Luyuan Xie
- Changsha Medical University, Changsha, Hunan Province, China
| | - Yuxin Ge
- Graduate Collaborative Training of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Department of Head and Neck Surgery, Central laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Yue Zhou
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Hui Wang
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Yongyi Chen
- Nursing Department, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Xiaomei Zhu
- Nursing Department, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Huayun Liu
- Nursing Department, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Qianjin Liao
- Graduate Collaborative Training of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Department of Head and Neck Surgery, Central laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Yu Kong
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Lijun Pan
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Junjun Li
- Pathology Department, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Lei Xue
- Pathology Department, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Sha Li
- Graduate Collaborative Training of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Department of Head and Neck Surgery, Central laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Xiao Zhou
- Graduate Collaborative Training of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Department of Head and Neck Surgery, Central laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| | - Xiaowu Sheng
- Graduate Collaborative Training of Hunan Cancer Hospital, Hengyang Medical School, University of South China, Department of Head and Neck Surgery, Central laboratory, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, Hunan Province, China
| |
Collapse
|
18
|
Monjazeb AM, Schalper KA, Villarroel-Espindola F, Nguyen A, Shiao SL, Young K. Effects of Radiation on the Tumor Microenvironment. Semin Radiat Oncol 2021; 30:145-157. [PMID: 32381294 DOI: 10.1016/j.semradonc.2019.12.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A malignant tumor consists of malignant cells as well as a wide array of normal host tissues including stroma, vasculature, and immune infiltrate. The interaction between cancer and these host tissues is critical as these host tissues play a variety of roles in supporting or resisting disease progression. Radiotherapy (RT) has direct effects on malignant cells, but, also, critically important effects on these other components of the tumor microenvironment (TME). Given the growing role of immune checkpoint inhibitors and other immunotherapy strategies, understanding how RT affects the TME, particularly the immune compartment, is essential to advance RT in this new era of cancer therapy. The interactions between RT and the TME are complex, affecting the innate and adaptive arms of the immune system. RT can induce both proinflammatory effects and immune suppressive effects that can either promote or impede antitumor immunity. It is likely that the initial proinflammatory effects of RT eventually lead to rebound immune-suppression as chronic inflammation sets in. The exact kinetics and nature of how RT changes the TME likely depends on timing, dose, fractionation, site irradiated, and tumor type. With increased understanding of the effects of RT on the TME, in the future it is likely that we will be able to personalize RT by varying the dose, site, and timing of intervention to generate the desired response to partner with immunotherapy strategies.
Collapse
Affiliation(s)
- Arta M Monjazeb
- UC Davis Comprehensive Cancer Center, Department of Radiation Oncology, Sacramento, CA.
| | - Kurt A Schalper
- Yale University School of Medicine, Department of Pathology, New Haven, CT
| | | | - Anthony Nguyen
- Cedars-Sinai Medical Center, Department of Radiation Oncology, Los Angeles, CA
| | - Stephen L Shiao
- Cedars-Sinai Medical Center, Department of Radiation Oncology, Los Angeles, CA
| | - Kristina Young
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR; Radiation Oncology Division, The Oregon Clinic, Portland, OR
| |
Collapse
|
19
|
Linking Serine/Glycine Metabolism to Radiotherapy Resistance. Cancers (Basel) 2021; 13:cancers13061191. [PMID: 33801846 PMCID: PMC8002185 DOI: 10.3390/cancers13061191] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Hyperactivation of the de novo serine/glycine biosynthesis across different cancer types and its critical contribution in tumor initiation, progression, and therapy resistance indicate the relevance of serine/glycine metabolism-targeted therapies as therapeutic intervention in cancer. In this review, we specifically focus on the contribution of the de novo serine/glycine biosynthesis pathway to radioresistance. We provide a future perspective on how de novo serine/glycine biosynthesis inhibition and serine-free diets may improve the outcome of radiotherapy. Future research in this field is needed to better understand serine/glycine metabolic reprogramming of cancer cells in response to radiation and the influence of this pathway in the tumor microenvironment, which may provide the rationale for the optimal combination therapies. Abstract The activation of de novo serine/glycine biosynthesis in a subset of tumors has been described as a major contributor to tumor pathogenesis, poor outcome, and treatment resistance. Amplifications and mutations of de novo serine/glycine biosynthesis enzymes can trigger pathway activation; however, a large group of cancers displays serine/glycine pathway overexpression induced by oncogenic drivers and unknown regulatory mechanisms. A better understanding of the regulatory network of de novo serine/glycine biosynthesis activation in cancer might be essential to unveil opportunities to target tumor heterogeneity and therapy resistance. In the current review, we describe how the activation of de novo serine/glycine biosynthesis in cancer is linked to treatment resistance and its implications in the clinic. To our knowledge, only a few studies have identified this pathway as metabolic reprogramming of cancer cells in response to radiation therapy. We propose an important contribution of de novo serine/glycine biosynthesis pathway activation to radioresistance by being involved in cancer cell viability and proliferation, maintenance of cancer stem cells (CSCs), and redox homeostasis under hypoxia and nutrient-deprived conditions. Current approaches for inhibition of the de novo serine/glycine biosynthesis pathway provide new opportunities for therapeutic intervention, which in combination with radiotherapy might be a promising strategy for tumor control and ultimately eradication. Further research is needed to gain molecular and mechanistic insight into the activation of this pathway in response to radiation therapy and to design sophisticated stratification methods to select patients that might benefit from serine/glycine metabolism-targeted therapies in combination with radiotherapy.
Collapse
|
20
|
Witherel CE, Sao K, Brisson BK, Han B, Volk SW, Petrie RJ, Han L, Spiller KL. Regulation of extracellular matrix assembly and structure by hybrid M1/M2 macrophages. Biomaterials 2021; 269:120667. [PMID: 33450585 PMCID: PMC7870567 DOI: 10.1016/j.biomaterials.2021.120667] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/22/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022]
Abstract
Aberrant extracellular matrix (ECM) assembly surrounding implanted biomaterials is the hallmark of the foreign body response, in which implants become encapsulated in thick fibrous tissue that prevents their proper function. While macrophages are known regulators of fibroblast behavior, how their phenotype influences ECM assembly and the progression of the foreign body response is poorly understood. In this study, we used in vitro models with physiologically relevant macrophage phenotypes, as well as controlled release of macrophage-modulating cytokines from gelatin hydrogels implanted subcutaneously in vivo to investigate the role of macrophages in ECM assembly. Primary human macrophages were polarized to four distinct phenotypes, which have each been associated with fibrosis, including pro-inflammatory M1, pro-healing M2, and a hybrid M1/M2, generated by exposing macrophages to M1-and M2-promoting stimuli simultaneously. Additionally, macrophages were first polarized to M1 and then to M2 (M1→M2) to generate a phenotype typically observed during normal wound healing. Human dermal fibroblasts that were cultured in macrophage-conditioned media upregulated numerous genes involved in regulation of ECM assembly, especially in M2-conditioned media. Hybrid M1/M2 macrophage-conditioned media caused fibroblasts to produce a matrix with thicker and less aligned fibers, while M2 macrophage-conditioned media caused the formation of a more aligned matrix with thinner fibers. Gelatin methacrylate hydrogels containing interleukin-4 (IL4) and IL13-loaded poly(lactic-co-glycolic acid) (PLGA) microparticles were designed to promote the M2 phenotype in a murine subcutaneous in vivo model. NanoString multiplex gene expression analysis of hydrogel explants showed that hydrogels without cytokines caused mostly M1 phenotype markers to be highly expressed at an early time point (3 days), but the release of IL4+IL13 promoted upregulation of M2 markers and genes associated with regulation of ECM assembly, such as Col5a1 and Col6a1. Biochemical analysis and second harmonic generation microscopy showed that the release of IL4+IL13 increased total sulfated glycosaminoglycan content and decreased fibril alignment, which is typically associated with less fibrotic tissue. Together, these results show that hybrid M1/M2 macrophages regulate ECM assembly, and that shifting the balance towards M2 may promote architectural and compositional changes in ECM with enhanced potential for downstream remodeling.
Collapse
Affiliation(s)
- Claire E Witherel
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Kimheak Sao
- Department of Biology, College of Arts and Sciences, Drexel University, Philadelphia, PA, USA
| | - Becky K Brisson
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Biao Han
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Susan W Volk
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Ryan J Petrie
- Department of Biology, College of Arts and Sciences, Drexel University, Philadelphia, PA, USA
| | - Lin Han
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Comparison of time and dose dependent gene expression and affected pathways in primary human fibroblasts after exposure to ionizing radiation. Mol Med 2020; 26:85. [PMID: 32907548 PMCID: PMC7488023 DOI: 10.1186/s10020-020-00203-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/23/2020] [Indexed: 02/08/2023] Open
Abstract
Background Exposure to ionizing radiation induces complex stress responses in cells, which can lead to adverse health effects such as cancer. Although a variety of studies investigated gene expression and affected pathways in human fibroblasts after exposure to ionizing radiation, the understanding of underlying mechanisms and biological effects is still incomplete due to different experimental settings and small sample sizes. Therefore, this study aims to identify the time point with the highest number of differentially expressed genes and corresponding pathways in primary human fibroblasts after irradiation at two preselected time points. Methods Fibroblasts from skin biopsies of 15 cell donors were exposed to a high (2Gy) and a low (0.05Gy) dose of X-rays. RNA was extracted and sequenced 2 h and 4 h after exposure. Differentially expressed genes with an adjusted p-value < 0.05 were flagged and used for pathway analyses including prediction of upstream and downstream effects. Principal component analyses were used to examine the effect of two different sequencing runs on quality metrics and variation in expression and alignment and for explorative analysis of the radiation dose and time point of analysis. Results More genes were differentially expressed 4 h after exposure to low and high doses of radiation than after 2 h. In experiments with high dose irradiation and RNA sequencing after 4 h, inactivation of the FAT10 cancer signaling pathway and activation of gluconeogenesis I, glycolysis I, and prostanoid biosynthesis was observed taking p-value (< 0.05) and (in) activating z-score (≥2.00 or ≤ − 2.00) into account. Two hours after high dose irradiation, inactivation of small cell lung cancer signaling was observed. For low dose irradiation experiments, we did not detect any significant (p < 0.05 and z-score ≥ 2.00 or ≤ − 2.00) activated or inactivated pathways for both time points. Conclusions Compared to 2 h after irradiation, a higher number of differentially expressed genes were found 4 h after exposure to low and high dose ionizing radiation. Differences in gene expression were related to signal transduction pathways of the DNA damage response after 2 h and to metabolic pathways, that might implicate cellular senescence, after 4 h. The time point 4 h will be used to conduct further irradiation experiments in a larger sample.
Collapse
|
22
|
Hasan HF, Elgazzar EM, Mostafa DM. Diminazene aceturate extenuate the renal deleterious consequences of angiotensin-II induced by γ-irradiation through boosting ACE2 signaling cascade. Life Sci 2020; 253:117749. [PMID: 32380079 DOI: 10.1016/j.lfs.2020.117749] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/24/2020] [Accepted: 05/01/2020] [Indexed: 10/24/2022]
Abstract
AIM This work aims to explore the role of diminazene aceturate (DIZE) in the enhancement of angiotensin-converting enzyme-2 (ACE2) to prevent the inflammatory and fibrotic response induced by γ-irradiation through activating the protective axis ACE2/angiotensin (1-7)/Mas receptor (ACE2/Ang(1-7)/Mas). METHODS Male rats were injected i.p. with 15 mg/kg DIZE daily for 7 days pre and post-irradiation, where 7.5 Gy of γ-radiation as a single dose was used. KEY FINDINGS Gamma radiation induced a significant elevation of renal biochemical parameters: urea, creatinine and blood urea nitrogen (BUN) in serum with a significant disturbance in oxidative stress markers: elevation in malondialdehyde (MDA) associated with a depletion of reduced glutathione (GSH) and superoxide dismutase (SOD). Beside elevation in the level of angiotensin II (AngII) that lead to remarkably increases in the levels of the renal inflammatory mediators: tumor necrosis factor-α (TNF-α), nuclear factor kappa B (NF-κB) and interleukin-1β (IL-1β) as well as renal fibrogenic markers: transforming growth factor-β1 (TGF-β1), connective tissue growth factor (CTGF), and hydroxyproline content in the renal tissues. DIZE caused marked expansion in the expression of ACE2 consequently decreased the expression of AngII and increased the expression of Ang(1-7) which through its Mas receptor ameliorates the biochemical and histopathological damage induced by radiation. SIGNIFICANCE DIZE-induced stimulation of ACE2 subdues the renal deleterious consequences induced by γ-radiation via activation of ACE2/Ang(1-7)/Mas axis in rats.
Collapse
Affiliation(s)
- Hesham Farouk Hasan
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Egypt.
| | - Eman M Elgazzar
- Health Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Dalia M Mostafa
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Egypt
| |
Collapse
|
23
|
A novel bispecific antibody alleviates bleomycin-induced systemic sclerosis injury. Int Immunopharmacol 2020; 85:106644. [PMID: 32474387 DOI: 10.1016/j.intimp.2020.106644] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/22/2020] [Accepted: 05/23/2020] [Indexed: 11/23/2022]
Abstract
Systemic sclerosis (SSc) is induced by variety of factors and eventually causes multiple organ damage. In recent years, biological agents targeting cytokines and cell surface molecules have gradually come to market. Here, the anti-inflammatory and antifibrotic effects of a novel bispecific antibody (FL-BsAb1/17) targeting interleukin-17A (IL-17A) and interleukin-1β (IL-1β) were detected. Bleomycin (BLM) was subcutaneously injected for 21 consecutive days to establish the SSc mouse model, and mice were subsequently treated with etanercept or different doses (1, 5, 10 mg/kg) of FL-BsAb1/17. The results showed that FL-BsAb1/17 treatment (10 mg/kg, 5 mg/kg) significantly attenuated BLM-induced SSc-like inflammation by inhibiting the expression of inflammatory factors (IL-17A, IL-1β, IL-8, IL-22, IL-23, IL-6) and fibrosis, with specific outcomes of dermis thickening and lung fibrosis, by inhibiting the expression of fibrotic factors (TGF-β, α-sma, Col-1, Col-3) in the serum, skin and lungs. In addition, FL-BsAb1/17 (10 mg/kg, 5 mg/kg) downregulated protein levels of TGF-β and phosphorylated Smad2/3 in the skin and lungs and reduced collagen 1 protein levels. This indicated that FL-BsAb1/17 can inhibit the development of fibrosis by inhibiting the TGF-β/Smad2/3 signaling pathway. FL-BsAb1/17 (10 mg/kg, 5 mg/kg) could also effectively reduce the content of MDA, increase the activity of SOD and CAT, and improve the total antioxidant capacity (T-AOC). In conclusion, FL-BsAb1/17 alleviated BLM-induced SSc by downregulating inflammatory cascades, relieving oxidative stress and inhibiting TGF-β/Smad2/3 signaling. These data suggest that FL-BsAb1/17 has potential as a novel therapeutic candidate for SSc.
Collapse
|
24
|
Farid A, Kamel D, Abdelwahab Montaser S, Mohamed Ahmed M, El Amir M, El Amir A. Assessment of antioxidant, immune enhancement, and antimutagenic efficacy of fennel seed extracts in irradiated human blood cultures. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2020. [DOI: 10.1080/16878507.2020.1728963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Alyaa Farid
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Dina Kamel
- Radiation Protection Department, Nuclear Power Plant Authority (NPPA), Cairo, Egypt
| | - Sherien Abdelwahab Montaser
- Radiation Biology Dept., National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mahmoud Mohamed Ahmed
- Radiation Biology Dept., National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mona El Amir
- Internal Medicine Department, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Giza, Egypt
| | - Azza El Amir
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
25
|
Farid A, Kamel D, Abdelwahab Montaser S, Mohamed Ahmed M, El Amir M, El Amir A. Synergetic role of senna and fennel extracts as antioxidant, anti-inflammatory and anti-mutagenic agents in irradiated human blood lymphocyte cultures. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2020. [DOI: 10.1080/16878507.2020.1723948] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Alyaa Farid
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Dina Kamel
- Radiation Protection Department, Nuclear Power Plant Authority (NPPA), Nasr City, Cairo, Egypt
| | - Sherien Abdelwahab Montaser
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Nasr City, Cairo, Egypt
| | - Mahmoud Mohamed Ahmed
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Nasr City, Cairo, Egypt
| | - Mona El Amir
- Internal Medicine Department, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Giza, Egypt
| | - Azza El Amir
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
26
|
McKelvey KJ, Hudson AL, Prasanna Kumar R, Eade T, Clarke SJ, Wheeler HR, Diakos CI, Howell VM. Sub-acute Toxicity in Non-cancerous Tissue and Immune-Related Adverse Events of a Novel Combination Therapy for Cancer. Front Oncol 2020; 9:1504. [PMID: 32010614 PMCID: PMC6971197 DOI: 10.3389/fonc.2019.01504] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
Brain, lung, and colon tissue experience deleterious immune-related adverse events when immune-oncological agents or radiation are administered. However, there is a paucity of information regarding whether the addition of radiation to immuno-oncological regimens exacerbates the tissue inflammatory response. We used a murine model to evaluate sub-acute tissue damage and the systemic immune response in C57Bl/6 mice when administered systemic anti-programmed cell death protein 1 (αPD-1) immunotherapy alone or in combination with stereotactic fractionated 10 gray/5 X-ray radiation to normal brain, lung or colon tissue. The model indicated that combinatorial αPD-1 immunotherapy and radiation may alter normal colon cell proliferation and cerebral blood vasculature, and induce systemic thrombocytopenia, lymphopenia, immune suppression, and altered immune repertoire (including interleukin-1β). Therein our data supports close monitoring of hematological and immune-related adverse events in patients receiving combination therapy.
Collapse
Affiliation(s)
- Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, The University of Sydney Northern Clinical School and Northern Sydney Local Health District, St Leonards, NSW, Australia.,Sydney Vital Translational Cancer Research Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,The Brain Cancer Group, St Leonards, NSW, Australia
| | - Amanda L Hudson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, The University of Sydney Northern Clinical School and Northern Sydney Local Health District, St Leonards, NSW, Australia.,Sydney Vital Translational Cancer Research Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,The Brain Cancer Group, St Leonards, NSW, Australia
| | - Ramyashree Prasanna Kumar
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, The University of Sydney Northern Clinical School and Northern Sydney Local Health District, St Leonards, NSW, Australia.,Sydney Vital Translational Cancer Research Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Thomas Eade
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Stephen J Clarke
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, The University of Sydney Northern Clinical School and Northern Sydney Local Health District, St Leonards, NSW, Australia.,Sydney Vital Translational Cancer Research Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Helen R Wheeler
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, The University of Sydney Northern Clinical School and Northern Sydney Local Health District, St Leonards, NSW, Australia.,Sydney Vital Translational Cancer Research Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,The Brain Cancer Group, St Leonards, NSW, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Connie I Diakos
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, The University of Sydney Northern Clinical School and Northern Sydney Local Health District, St Leonards, NSW, Australia.,Sydney Vital Translational Cancer Research Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Viive M Howell
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, The University of Sydney Northern Clinical School and Northern Sydney Local Health District, St Leonards, NSW, Australia.,Sydney Vital Translational Cancer Research Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,The Brain Cancer Group, St Leonards, NSW, Australia
| |
Collapse
|
27
|
Bai L, Zhou BS, Zhao YX. Dynamic changes in T-cell subsets and C-reactive protein after radiation therapy in lung cancer patients and correlation with symptomatic radiation pneumonitis treated with steroid therapy. Cancer Manag Res 2019; 11:7925-7931. [PMID: 31686908 PMCID: PMC6709788 DOI: 10.2147/cmar.s209286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/25/2019] [Indexed: 12/14/2022] Open
Abstract
Objectives To investigate relationships among serum T-cell subsets, CRP, levels and radiation pneumonitis (RP) in lung cancer patients receiving radiotherapy. Methods A case-control study with frequency matching was carried out. The case group comprised 36 lung cancer patients who had developed grade ≥2 RP after thoracic radiotherapy. The control group was 36 patients with lung cancer without RP. Patients in the case group received steroid therapy for 1 month after diagnosis of RP and were followed up for 3 months. T-cell subsets, CRP, and pulmonary function were detected at three time points (onset of RP and 1 and 3 months after diagnosis). Data for the control group were collected 3 months after radiotherapy. Treatment effectiveness was evaluated at 1 and 3 months after diagnosis of RP. Results Of the 36 patients in the case group, three with grade5 RP died from respiratory failure. The other 33 cases had all improved with steroid therapy at 3 months after RP diagnosis. In these 33, CD3+T-cell quantity, CD4+T-cell quantity, and of CD4+:CD8+ ratio in T-cell subsets decreased significantly and CRP increased (P<0.05) at the onset of RP compared with the control group. After steroid therapy, CD4+T-cell quantity increased significantly compared to before treatment. The same change was seen in CD4+:CD8+ ratio, whereas CRP levels decreased obviously, with treatment effectiveness improved. In addition, with the damage level of RP increased, CD4+ T -cell quantity decreased obviously and CRP levels increased accordingly at the onset of RP (P<0.05). Conclusion T-cell subsets and CRP may become effective immunological biomarkers for predicting damage from RP and evaluating treatment effectivesness of steroid therapy.
Collapse
Affiliation(s)
- Lu Bai
- Department of Radiation Oncology, First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Bao-Sen Zhou
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang 110122, People's Republic of China.,Liaoning Provincial Department of Education, Key Laboratory of Cancer Etiology and Prevention, First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Yu-Xia Zhao
- Department of Radiation Oncology, Fourth Affiliated Hospital of China Medical University, Shenyang 110032, People's Republic of China
| |
Collapse
|
28
|
Jang H, Lee J, Park S, Kim JS, Shim S, Lee SB, Han SH, Myung H, Kim H, Jang WS, Lee SJ, Myung JK. Baicalein Mitigates Radiation-Induced Enteritis by Improving Endothelial Dysfunction. Front Pharmacol 2019; 10:892. [PMID: 31474856 PMCID: PMC6707809 DOI: 10.3389/fphar.2019.00892] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022] Open
Abstract
Background and Aims: Radiation-induced intestinal injury occurred in application of radiotherapy for abdominal and pelvic cancers or in nuclear accidents. Radiation-induced enteritis may be considered an ideal model of gastrointestinal inflammation. The endothelium is a crucial component of inflammation, and the endothelial dysfunction following radiation exposure induces the intestinal proinflammatory response and progression of radiation enteritis. Baicalein (5,6,7-trihydroxyflavonoid) is a flavonoid from Scutellaria baicalensis used in oriental herbal medicine. Baicalein has been found to have multiple beneficial properties including antioxidant, anti-inflammatory, anti-allergic, and anti-cancer activities. Here, we investigated the therapeutic effects of baicalein on endothelial dysfunction in radiation-induced intestinal inflammation. Materials and Methods: We performed histological analysis, bacterial translocation, and intestinal permeability assays and also assessed infiltration of leukocytes and inflammatory cytokine expression using a mouse model of radiation-induced enteritis. In addition, to investigate the effect of baicalein in endothelial dysfunction, we analyzed endothelial-derived adherent molecules in human umbilical vein endothelial cells (HUVECs) and irradiated intestinal tissue. Results: Histological damage such as shortening of villi length and impaired intestinal crypt function was observed in the radiation-induced enteritis mouse model. Intestinal damage was attenuated in baicalein-treated groups with improvement of intestinal barrier function. Baicalein inhibited the expression of radiation-induced adherent molecules in HUVECs and intestine of irradiated mouse and decreased leukocyte infiltration in the radiation-induced enteritis. Conclusions: Baicalein could accelerate crypt regeneration via recovery of endothelial damage. Therefore, baicalein has a therapeutic effect on radiation-induced intestinal inflammation by attenuating endothelial damage.
Collapse
Affiliation(s)
- Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Janet Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.,Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Joong Sun Kim
- Herbal Medicine Resources Center, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sung-Honn Han
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Hyunwook Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Won-Suk Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sun-Joo Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.,Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| |
Collapse
|
29
|
Hindman B, Ma Q. Carbon nanotubes and crystalline silica stimulate robust ROS production, inflammasome activation, and IL-1β secretion in macrophages to induce myofibroblast transformation. Arch Toxicol 2019; 93:887-907. [PMID: 30847537 DOI: 10.1007/s00204-019-02411-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/21/2019] [Indexed: 12/12/2022]
Abstract
Pulmonary exposure to inhaled particulates elicits complex inflammatory and fibrotic responses that may progress to chronic fibrosis. The fibrogenic potentials of respirable particulates are influenced by their physicochemical properties and their interactions with major pathways to drive fibrotic development in the lung. Macrophages were exposed to six carbon nanotubes (CNTs) of varying dimensions, crystalline silica, or carbon black (CB), with lipopolysaccharide (LPS) and transforming growth factor (TGF)-β1 as positive controls. Macrophage-conditioned media was collected and applied to cultures of human pulmonary fibroblast line WI38-VA13 to induce myofibroblast transformation. Multi-walled and single-walled CNTs (MWCNTs and SWCNTs, respectively) and silica, but not CB, stimulated robust myofibroblast transformation through macrophage-conditioned media. On an equal weight basis, MWCNTs induced higher induction than SWCNTs. High induction was observed for MWCNTs with a long and slender or a short and rigid shape, and silica, at levels comparable to those by LPS and TGF-β1. Fibrogenic particulates induced high levels of IL-1β mRNA expression and protein secretion that are required for macrophage-guided myofibroblast transformation. Induction of IL-1β is dependent on the activation of the NLRP3 (NOD-like receptor family, pyrin domain containing 3) inflammasome and ROS (reactive oxygen species) production in macrophages, as inhibition of NLRP3 by MCC950 and reduction of ROS production by N-acetylcysteine blocked NLRP3 activation, IL-1β induction, and fibroblast activation and differentiation. Therefore, fibrogenic CNTs and silica, but not CB, elicit robust macrophage-guided myofibroblast transformation, which depends on the induction of IL-1β through the NLRP3 inflammasome pathway and the increased production of ROS in macrophages.
Collapse
Affiliation(s)
- Bridget Hindman
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, 26505, USA
| | - Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, 26505, USA.
| |
Collapse
|
30
|
Perraud AL, Rao DM, Kosmacek EA, Dagunts A, Oberley-Deegan RE, Gally F. The ion channel, TRPM2, contributes to the pathogenesis of radiodermatitis. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2019; 58:89-98. [PMID: 30483886 PMCID: PMC6394656 DOI: 10.1007/s00411-018-0769-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 11/20/2018] [Indexed: 06/09/2023]
Abstract
Radiodermatitis is a painful side effect for cancer patients undergoing radiotherapy. Irradiation of the skin causes inflammation and breakdown of the epidermis and can lead to significant morbidity and mortality in severe cases, as seen in exposure from accidents or weapons such as "dirty bombs" and ultimately leads to tissue fibrosis. However, the pathogenesis of radiodermatitis is not fully understood. Using a mouse model of radiodermatitis, we showed that the Transient Receptor Potential Melastatin 2 (TRPM2) ion channel plays a significant role in the development of dermatitis following exposure to ionizing radiation. Irradiated TRPM2-deficient mice developed less inflammation, fewer severe skin lesions and decreased fibrosis when compared to wild type mice. The TRPM2-deficient mice also showed a faster recovery period as seen by their increased weight gain post irradiation. Finally, TRPM2-deficient mice exhibited lower systemic inflammation with a reduction in inflammatory cytokines present in the serum. These findings suggest that TRPM2 may be a potential therapeutic target for reducing the severity of radiodermatitis.
Collapse
Affiliation(s)
- Anne-Laure Perraud
- Department of Biomedical Research, National Jewish Health, 1400 Jackson St., Room K827, Denver, CO, 80206, USA
- Department of Immunology and Microbiology, University of Colorado Denver, Denver, USA
| | - Deviyani M Rao
- Department of Biomedical Research, National Jewish Health, 1400 Jackson St., Room K827, Denver, CO, 80206, USA
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, USA
| | - Aleksandra Dagunts
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, USA
| | - Fabienne Gally
- Department of Biomedical Research, National Jewish Health, 1400 Jackson St., Room K827, Denver, CO, 80206, USA.
| |
Collapse
|
31
|
Treatment with maresin 1, a docosahexaenoic acid-derived pro-resolution lipid, protects skin from inflammation and oxidative stress caused by UVB irradiation. Sci Rep 2019; 9:3062. [PMID: 30816324 PMCID: PMC6395735 DOI: 10.1038/s41598-019-39584-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/22/2019] [Indexed: 12/19/2022] Open
Abstract
Acute exposure to UVB irradiation causes skin inflammation and oxidative stress, and long-term exposure to UVB irradiation may lead to carcinogenesis. Our organism has endogenous mechanisms to actively limit inflammation. Maresin 1 (MaR1; 7R,14S-dihydroxy-docosa-4Z,8E,10E,12Z,16Z,19Z-hexaenoic acid) is a pro-resolution lipid mediator derived from the docosahexaenoic acid, which presents anti-inflammatory and pro-resolution effects. However, it remains to be determined if treatment with MaR1 can inhibit inflammatory and oxidative alterations in the skin triggered by UVB. The treatment with MaR1 (0.1-10 ng/mice at -10 min relative to the UVB irradiation protocol) reduced UVB-induced skin edema, neutrophil recruitment (MPO; myeloperoxidase activity, and migration of LysM-eGFP+ cells), cytokine production, matrix metalloproteinase-9 activity, keratinocyte apoptosis, epidermal thickening, mast cells counts and degradation of skin collagen in hairless mice. UVB irradiation caused a decrease of GSH (reduced glutathione) levels, activity of the enzyme catalase, ferric reducing ability (FRAP), and ABTS radical scavenging capacity as well as induced lipid hydroperoxide, superoxide anion production, and gp91phox mRNA expression. These parameters that indicate oxidative stress were inhibited by MaR1 treatment. Therefore, these data suggest MaR1 as a promising pharmacological tool in controlling the deleterious effects related to UVB irradiation.
Collapse
|
32
|
Ridner SH, Dietrich MS, Sonis ST, Murphy B. Biomarkers Associated with Lymphedema and Fibrosis in Patients with Cancer of the Head and Neck. Lymphat Res Biol 2018; 16:516-524. [PMID: 30484735 PMCID: PMC6306661 DOI: 10.1089/lrb.2017.0074] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This study examined interrelationships of selected interleukins (ILs), tumor growth factors, matrix metalloproteinases (MMPs), and C-reactive protein, interferon-gamma (IFN-γ), and tumor necrosis factor α (TNF-α) with lymphedema/fibrosis in patients with head and neck cancer (HNC). METHODS AND RESULTS Patients newly diagnosed with ≥Stage II HNC (N = 100) were assessed for external/internal lymphedema and/or fibrosis before treatment, end-of-treatment, and at regularly established intervals through 72 weeks posttreatment and blood was drawn. Data from 83 patients were analyzed. Group-based trajectory modeling generated patient groups with similar longitudinal biomarker and lymphedema-fibrosis trajectories. Area-under-the-curve (AUC) values were also generated for each biomarker and severity of lymphedema-fibrosis. Associations among and between biomarkers and lymphedema-fibrosis trajectories and AUCs were tested (log-likelihood chi-square, correlations). The strongest evidence for the association of biomarkers with the overall and trajectory patterns and severity of lymphedema-fibrosis was observed for IL-6, IL-1β, TNF-α, TGF-β1, and MMP-9 (all p < 0.05). Convergence of joint trajectory patterns and AUC were observed with IL-6 with all lymphedema-fibrosis trajectories and internal lymphedema AUC. IL-1β trajectories converged with external lymphedema trajectories and all lymphedema-fibrosis AUCs. TNF-α and TGF-β1 converged most strongly with fibrosis in terms of trajectory patterns. However TNF-α demonstrated stronger association with lymphedema-fibrosis AUC (fibrosis: rs = 0.49). MMP-9 demonstrated convergence with lymphedema-fibrosis AUCs (lymphedema: 0.43-0.42; fibrosis: 0.35). CONCLUSION Systemic levels of selected mediators of proinflammatory processes track with acute and chronic clinical phenotypes of lymphedema/fibrosis in HNC patients suggesting their potential role in the pathogenesis of these conditions.
Collapse
Affiliation(s)
| | - Mary S. Dietrich
- Vanderbilt University School of Nursing, Nashville, Tennessee
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Stephen T. Sonis
- Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts
| | | |
Collapse
|
33
|
Najafi M, Motevaseli E, Shirazi A, Geraily G, Rezaeyan A, Norouzi F, Rezapoor S, Abdollahi H. Mechanisms of inflammatory responses to radiation and normal tissues toxicity: clinical implications. Int J Radiat Biol 2018; 94:335-356. [PMID: 29504497 DOI: 10.1080/09553002.2018.1440092] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 01/23/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE Cancer treatment is one of the most challenging diseases in the present era. Among a few modalities for cancer therapy, radiotherapy plays a pivotal role in more than half of all treatments alone or combined with other cancer treatment modalities. Management of normal tissue toxicity induced by radiation is one of the most important limiting factors for an appropriate radiation treatment course. The evaluation of mechanisms of normal tissue toxicity has shown that immune responses especially inflammatory responses play a key role in both early and late side effects of exposure to ionizing radiation (IR). DNA damage and cell death, as well as damage to some organelles such as mitochondria initiate several signaling pathways that result in the response of immune cells. Massive cell damage which is a common phenomenon following exposure to a high dose of IR cause secretion of a lot of inflammatory mediators including cytokines and chemokines. These mediators initiate different changes in normal tissues that may continue for a long time after irradiation. In this study, we reviewed the mechanisms of inflammatory responses to IR that are involved in normal tissue toxicity and considered as the most important limiting factors in radiotherapy. Also, we introduced some agents that have been proposed for management of these responses. CONCLUSIONS The early inflammation during the radiation treatment is often a limiting factor in radiotherapy. In addition to the limiting factors, chronic inflammatory responses may increase the risk of second primary cancers through continuous free radical production, attenuation of tumor suppressor genes, and activation of oncogenes. Moreover, these effects may influence non-irradiated tissues through a mechanism named bystander effect.
Collapse
Affiliation(s)
- Masoud Najafi
- a Radiology and Nuclear Medicine Department, School of Paramedical Sciences , Kermanshah University of Medical Science , Kermanshah , Iran
| | - Elahe Motevaseli
- b Department of Molecular Medicine, School of Advanced Technologies in Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Alireza Shirazi
- c Department of Medical Physics and Biomedical Engineering, Faculty of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Ghazale Geraily
- c Department of Medical Physics and Biomedical Engineering, Faculty of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Abolhasan Rezaeyan
- d Department of Medical Physics, School of Medicine , Iran University of Medical Sciences , Tehran , Iran
| | - Farzad Norouzi
- e Science and Research Branch , Azad University , Tehran , Iran
| | - Saeed Rezapoor
- f Department of Radiology, Faculty of Paramedical Sciences , Tehran University of Medical Sciences , Tehran , Iran
| | - Hamid Abdollahi
- d Department of Medical Physics, School of Medicine , Iran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
34
|
Villani F, Busia A, Villani M, Vismara C, Viviani S, Bonfante V. Serum Cytokine in Response to Chemo-Radiotherapy for Hodgkin's Disease. TUMORI JOURNAL 2018; 94:803-8. [DOI: 10.1177/030089160809400605] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and Background Mediastinal radiotherapy and multiple-drug chemotherapy, including bleomycin employed in the treatment of Hodgkin's disease, can produce lung toxicity leading to fibrosis. There is increasing evidence of the involvement in the fibrosing process of different cytokines and growth factors such as TNF-alfa, IL-1 beta, TGF-beta and PDGF. Material and Methods In a pilot study, we evaluated lung function in 20 patients suffering from Hodgkin's disease, mainly in stage II A, submitted to multiple-drug chemotherapy including bleomycin (ABVD) and mediastinal radiotherapy and correlated its modifications with serum concentration of the cytokines determined by immunoenzymatic assay. Spirometry and transfer lung function for carbon monoxide (DLCO) were performed before, at the end of chemotherapy, at the end of radiotherapy and after a follow-up of 6 and 12 months. Results DLCO decreased at the end of the combined treatment and then remained constantly decreased. TNF-alfa, TGF-beta and PDGF-alfa concentrations did not change, whereas IL-1 beta significantly increased after the completion of the combined treatment and after a follow-up of 6–months and then declined to normal values after 12 months. The serum concentration of the cytokine was significantly higher in patients who had a DLCO <75% of predicted after 1 year than in patients with a DLCO >75%. Conclusions The results indicate a potential role of IL-1 beta in the pathogenesis of chemoradiotherapy-induced lung toxicity, which needs to be confirmed in a larger patient population.
Collapse
Affiliation(s)
- Fabrizio Villani
- UO di Pneumologia e Fisiopatologia Respiratoria, Istituto Nazionale Tumori, Milan, Italy
| | - Alessandra Busia
- UO di Pneumologia e Fisiopatologia Respiratoria, Istituto Nazionale Tumori, Milan, Italy
| | - Massimiliano Villani
- UO di Pneumologia e Fisiopatologia Respiratoria, Istituto Nazionale Tumori, Milan, Italy
| | - Chiara Vismara
- UO Analisi Clinica e Microbiologia, Istituto Nazionale Tumori, Milan, Italy
| | - Simonetta Viviani
- UO Oncologia Medica, Fondazione IRCCS, Istituto Nazionale Tumori, Milan, Italy
| | - Valeria Bonfante
- UO Oncologia Medica, Fondazione IRCCS, Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
35
|
Groves AM, Johnston CJ, Williams JP, Finkelstein JN. Role of Infiltrating Monocytes in the Development of Radiation-Induced Pulmonary Fibrosis. Radiat Res 2018; 189:300-311. [PMID: 29332538 DOI: 10.1667/rr14874.1] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Lung exposure to radiation induces an injury response that includes the release of cytokines and chemotactic mediators; these signals recruit immune cells to execute inflammatory and wound-healing processes. However, radiation alters the pulmonary microenvironment, dysregulating the immune responses and preventing a return to homeostasis. Importantly, dysregulation is observed as a chronic inflammation, which can progress into pneumonitis and promote pulmonary fibrosis; inflammatory monocytes, which are bone marrow derived and express CCR2, have been shown to migrate into the lung after radiation exposure. Although the extent to which recruited inflammatory monocytes contribute to radiation-induced pulmonary fibrosis has not been fully investigated, we hypothesize that its pathogenesis is reliant on this population. The CC chemokine ligand, CCL2, is a chemotactic mediator responsible for trafficking of CCR2+ inflammatory cells into the lung. Therefore, the contribution of this mediator to fibrosis development was analyzed. Interleukin (IL)-1β, a potent pro-inflammatory cytokine expressed during the radiation response, and its receptor, IL-1R1, were also evaluated. To this end, CCR2-/-, IL-1β-/- and IL-1R1-/- chimeric mice were generated and exposed to 12.5 Gy thoracic radiation, and their response was compared to wild-type (C57BL/6) syngeneic controls. Fibrotic foci were observed in the periphery of the lungs of C57 syngeneic mice and CCR2-/- recipient mice that received C57 bone marrow (C57 > CCR2-/-) by 16 and 12 weeks after irradiation, respectively. In contrast, in the mice that had received bone marrow lacking CCR2 (CCR2-/- > C57 and CCR2-/- syngeneic mice), no pulmonary fibrosis was observed at 22 weeks postirradiation. This observation correlated with decreased numbers of infiltrating and interstitial macrophages compared to controls, as well as reduced proportions of pro-inflammatory Ly6C+ macrophages observed at 12-18 weeks postirradiation, suggesting that CCR2+ macrophages contribute to radiation-induced pulmonary fibrosis. Interestingly, reduced proportions of CD206+ lung macrophages were also present at these time points in CCR2-/- chimeric mice, regardless of donor bone marrow type, suggesting that the phenotype of resident subsets may be influenced by CCR2. Furthermore, chimeras, in which either IL-1β was ablated from infiltrating cells or IL-1R1 from lung tissues, were also protected from fibrosis development, correlating with attenuated CCL2 production; these data suggest that IL-1β may influence chemotactic signaling after irradiation. Overall, our data suggest that CCR2+ infiltrating monocyte-derived macrophages may play a critical role in the development of radiation-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Angela M Groves
- Department of a Pediatrics M&D Neonatology, University of Rochester Medical Center, Rochester, New York
| | - Carl J Johnston
- Department of a Pediatrics M&D Neonatology, University of Rochester Medical Center, Rochester, New York.,b Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Jacqueline P Williams
- b Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Jacob N Finkelstein
- Department of a Pediatrics M&D Neonatology, University of Rochester Medical Center, Rochester, New York.,b Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
36
|
Jang H, Myung H, Lee J, Myung JK, Jang WS, Lee SJ, Bae CH, Kim H, Park S, Shim S. Impaired Skin Barrier Due to Sebaceous Gland Atrophy in the Latent Stage of Radiation-Induced Skin Injury: Application of Non-Invasive Diagnostic Methods. Int J Mol Sci 2018; 19:ijms19010185. [PMID: 29316698 PMCID: PMC5796134 DOI: 10.3390/ijms19010185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/01/2018] [Accepted: 01/03/2018] [Indexed: 12/18/2022] Open
Abstract
Radiation-induced skin injury can take the form of serious cutaneous damage and have specific characteristics. Asymptomatic periods are classified as the latent stage. The skin barrier plays a critical role in the modulation of skin permeability and hydration and protects the body against a harsh external environment. However, an analysis on skin barrier dysfunction against radiation exposure in the latent stage has not been conducted. Thus, we investigated whether the skin barrier is impaired by irradiation in the latent stage and aimed to identify the molecules involved in skin barrier dysfunction. We analyzed skin barrier function and its components in SKH1 mice that received 20 and 40 Gy local irradiation. Increased transepidermal water loss and skin pH were observed in the latent stage of the irradiated skin. Skin barrier components, such as structural proteins and lipid synthesis enzymes in keratinocyte, increased in the irradiated group. Interestingly, we noted sebaceous gland atrophy and increased serine protease and inflammatory cytokines in the irradiated skin during the latent period. This finding indicates that the main factor of skin barrier dysfunction in the latent stage of radiation-induced skin injury is sebaceous gland deficiency, which could be an intervention target for skin barrier impairment.
Collapse
Affiliation(s)
- Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Hyunwook Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Janet Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Won-Suk Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Sun-Joo Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Chang-Hwan Bae
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| |
Collapse
|
37
|
Pereira FC, Parisi JR, Maglioni CB, Machado GB, Barragán-Iglesias P, Silva JRT, Silva ML. Antinociceptive effects of low-level laser therapy at 3 and 8 j/cm2in a rat model of postoperative pain: possible role of endogenous Opioids. Lasers Surg Med 2017; 49:844-851. [DOI: 10.1002/lsm.22696] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Fabio C. Pereira
- Department of Physiotherapy; College of Nursing of the Federal University of Alfenas-UNIFAL; Alfenas Brazil
| | - Julia R. Parisi
- Department of Physical Therapy; Federal University of São Carlos-UFSCar; São Carlos Brazil
| | - Caio B. Maglioni
- Department of Physiotherapy; College of Nursing of the Federal University of Alfenas-UNIFAL; Alfenas Brazil
| | - Gabriel B. Machado
- Department of Physiotherapy; College of Nursing of the Federal University of Alfenas-UNIFAL; Alfenas Brazil
| | | | - Josie R. T. Silva
- Department of Physiotherapy; College of Nursing of the Federal University of Alfenas-UNIFAL; Alfenas Brazil
| | - Marcelo L. Silva
- Department of Physiotherapy; College of Nursing of the Federal University of Alfenas-UNIFAL; Alfenas Brazil
| |
Collapse
|
38
|
Kainthola A, Haritwal T, Tiwari M, Gupta N, Parvez S, Tiwari M, Prakash H, Agrawala PK. Immunological Aspect of Radiation-Induced Pneumonitis, Current Treatment Strategies, and Future Prospects. Front Immunol 2017; 8:506. [PMID: 28512460 PMCID: PMC5411429 DOI: 10.3389/fimmu.2017.00506] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/12/2017] [Indexed: 12/21/2022] Open
Abstract
Delivery of high doses of radiation to thoracic region, particularly with non-small cell lung cancer patients, becomes difficult due to subsequent complications arising in the lungs of the patient. Radiation-induced pneumonitis is an early event evident in most radiation exposed patients observed within 2-4 months of treatment and leading to fibrosis later. Several cytokines and inflammatory molecules interplay in the vicinity of the tissue developing radiation injury leading to pneumonitis and fibrosis. While certain cytokines may be exploited as biomarkers, they also appear to be a potent target of intervention at transcriptional level. Initiation and progression of pneumonitis and fibrosis thus are dynamic processes arising after few months to year after irradiation of the lung tissue. Currently, available treatment strategies are challenged by the major dose limiting complications that curtails success of the treatment as well as well being of the patient's future life. Several approaches have been in practice while many other are still being explored to overcome such complications. The current review gives a brief account of the immunological aspects, existing management practices, and suggests possible futuristic approaches.
Collapse
Affiliation(s)
- Anup Kainthola
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Teena Haritwal
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Mrinialini Tiwari
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Noopur Gupta
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard University, New Delhi, India
| | - Manisha Tiwari
- Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Hrideysh Prakash
- School of Life Sciences, Science complex, University of Hyderabad, Hyderabad, India
| | - Paban K. Agrawala
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| |
Collapse
|
39
|
Liao W, Hei TK, Cheng SK. Radiation-Induced Dermatitis is Mediated by IL17-Expressing γδ T Cells. Radiat Res 2017; 187:454-464. [PMID: 28406748 PMCID: PMC5524538 DOI: 10.1667/rr007cc.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Radiation dermatitis is a serious cutaneous injury caused by radiation therapy or upon accidental nuclear exposure. However, the pathogenic immune mechanisms underlying this injury are still poorly understood. We seek to discover how the dysregulated immune response after irradiation orchestrates skin inflammation. The skin on the left flank of C57BL/6J wild-type and C57BL/6J Tcrd-/- mice, which are deficit in γδ T cells, was exposed to a single X-ray dose of 25 Gy, and the right-flank skin was used as a sham-irradiated control. At 4 weeks postirradiation, the wild-type skin exhibited signs of depilation, erythema and desquamation. Histological analysis showed hyperproliferation of keratinocytes and acanthosis. Dramatic elevation of IL17-expressing T cells was identified from the irradiated skin, which was mainly contributed by γδ T cells and innate lymphoid cells, rather than Th17 cells. Furthermore, protein levels of critical cytokines for IL17-expressing γδ T cell activation, IL1β and IL23 were found markedly upregulated. Lastly, radiation-induced dermatitis was significantly attenuated in γδ T cell knockout mice. In vitro, normal human epidermal keratinocytes (NHEKs) could be initiator cells of inflammation by providing a great number of pro-inflammatory mediators upon radiation, and as well as effector cells of epidermal hyperplasia in response to exogenous IL17 and/or IL22 treatment. Our findings implicate a novel role of IL17-expressing γδ T cells in mediating radiation-induced skin inflammation. This study reveals the innate immune response pathway as a potential therapeutic target for radiation skin injury.
Collapse
Affiliation(s)
- Wupeng Liao
- Department of Radiation Oncology, College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Tom K. Hei
- Center for Radiological Research, College of Physicians and Surgeons, Columbia University, New York, New York 10032
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032
| | - Simon K. Cheng
- Department of Radiation Oncology, College of Physicians and Surgeons, Columbia University, New York, New York 10032
| |
Collapse
|
40
|
Acheva A, Schettino G, Prise KM. Pro-inflammatory Signaling in a 3D Organotypic Skin Model after Low LET Irradiation-NF-κB, COX-2 Activation, and Impact on Cell Differentiation. Front Immunol 2017; 8:82. [PMID: 28239377 PMCID: PMC5300980 DOI: 10.3389/fimmu.2017.00082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/18/2017] [Indexed: 11/25/2022] Open
Abstract
Nearly 85% of radiotherapy patients develop acute radiation dermatitis, which is an inflammatory reaction of the skin at the treatment field and in the surrounding area. The aims of this study were to unravel the mechanisms of radiation-induced inflammatory responses after localized irradiation in a human 3D organotypic skin culture model. This could provide possible inflammatory targets for reduction of skin side effects. 3D organotypic skin cultures were set up and locally irradiated with 225 kVp X-rays, using a combination of full exposure and partial shielding (50%) of the cultures. The secretion of pro-inflammatory cytokines, the phenotype, and the differentiation markers expression of the cultures were assessed up to 10 days postirradiation. The pro-inflammatory transcription factor nuclear factor kappa B (NF-κB) and cyclooxygenase-2 (COX-2) pathways have been studied. The results showed fast activation of NF-κB, most likely triggered by DNA damage in the irradiated cells, followed by upregulation of p38 MAPK and COX-2 in the irradiated and surrounding, non-irradiated, areas of the 3D cultures. The application of the COX-2 inhibitor sc-236 was effective at reducing the COX-2 mRNA levels 4 h postirradiation. The same inhibitor also suppressed the PGE2 secretion significantly 72 h after the treatment. The expression of a pro-inflammatory phenotype and abnormal differentiation markers of the cultures were also reduced. However, the use of an NF-κB inhibitor (Bay 11-7085) did not have the predicted positive effect on the cultures phenotype postirradiation. Radiation-induced pro-inflammatory responses have been observed in the 3D skin model. The activated signaling pathways involved NF-κB transcription factor and its downstream target COX-2. Further experiments aiming to suppress the inflammatory response via specific inhibitors showed that COX-2 is a suitable target for reduction of the normal skin inflammatory responses at radiotherapy, while NF-κB inhibition had detrimental effects on the 3D skin model development.
Collapse
Affiliation(s)
- Anna Acheva
- Queen's University Belfast, Centre for Cancer Research and Cell Biology, Belfast, UK; Section of Pathology, Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Giuseppe Schettino
- Queen's University Belfast, Centre for Cancer Research and Cell Biology, Belfast, UK; National Physical Laboratory, Teddington, UK
| | - Kevin M Prise
- Queen's University Belfast, Centre for Cancer Research and Cell Biology , Belfast , UK
| |
Collapse
|
41
|
Cirincione R, Di Maggio FM, Forte GI, Minafra L, Bravatà V, Castiglia L, Cavalieri V, Borasi G, Russo G, Lio D, Messa C, Gilardi MC, Cammarata FP. High-Intensity Focused Ultrasound- and Radiation Therapy-Induced Immuno-Modulation: Comparison and Potential Opportunities. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:398-411. [PMID: 27780661 DOI: 10.1016/j.ultrasmedbio.2016.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 09/20/2016] [Accepted: 09/21/2016] [Indexed: 05/12/2023]
Abstract
In recent years, high-intensity focused ultrasound (HIFU) has emerged as a new and promising non-invasive and non-ionizing ablative technique for the treatment of localized solid tumors. Extensive pre-clinical and clinical studies have evidenced that, in addition to direct destruction of the primary tumor, HIFU-thermoablation may elicit long-term systemic host anti-tumor immunity. In particular, an important consequence of HIFU treatment includes the release of tumor-associated antigens (TAAs), the secretion of immuno-suppressing factors by cancer cells and the induction of cytotoxic T lymphocyte (CTL) activity. Radiation therapy (RT) is the main treatment modality used for many types of tumors and about 50% of all cancer patients receive RT, often used in combination with surgery and chemotherapy. It is well known that RT can modulate anti-tumor immune responses, modifying micro-environment and stimulating inflammatory factors that can greatly affect cell invasion, bystander effects, radiation tissue complications (such as fibrosis), genomic instability and thus, intrinsic cellular radio-sensitivity. To date, various combined therapeutic strategies (such as immuno-therapy) have been performed in order to enhance RT success in treating locally advanced and recurrent tumors. Recent works suggested the combined use of HIFU and RT treatments to increase the tumor cell radio-sensitivity, in order to synergize the effects reaching the maximum results with minimal doses of ionizing radiation (IR). Here, we highlight the opposite immuno-modulation roles of RT and HIFU, providing scientific reasons to test, by experimental approaches, the use of HIFU immune-stimulatory capacity to improve tumor radio-sensitivity, to reduce the RT induced inflammatory response and to decrease the dose-correlated side effects in normal tissues.
Collapse
Affiliation(s)
| | - Federica Maria Di Maggio
- IBFM CNR, Cefalù, Palermo, Italy; Department of Pathobiology and Medical Biotechnologies, University of Palermo, Palermo, Italy
| | | | | | - Valentina Bravatà
- IBFM CNR, Cefalù, Palermo, Italy; Department of Pathobiology and Medical Biotechnologies, University of Palermo, Palermo, Italy
| | | | - Vincenzo Cavalieri
- Laboratory of Molecular Biology and Functional Genomics, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | | | | | - Domenico Lio
- Department of Pathobiology and Medical Biotechnologies, University of Palermo, Palermo, Italy
| | - Cristina Messa
- IBFM CNR, Cefalù, Palermo, Italy; Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy; Nuclear Medicine Center, San Gerardo Hospital, Monza, Italy
| | - Maria Carla Gilardi
- IBFM CNR, Cefalù, Palermo, Italy; Department of Health Sciences, Tecnomed Foundation, University of Milano-Bicocca, Milan, Italy; Nuclear Medicine, San Raffaele Scientific Institute, Milan, Italy
| | | |
Collapse
|
42
|
Plantier L, Renaud H, Respaud R, Marchand-Adam S, Crestani B. Transcriptome of Cultured Lung Fibroblasts in Idiopathic Pulmonary Fibrosis: Meta-Analysis of Publically Available Microarray Datasets Reveals Repression of Inflammation and Immunity Pathways. Int J Mol Sci 2016; 17:ijms17122091. [PMID: 27983601 PMCID: PMC5187891 DOI: 10.3390/ijms17122091] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/02/2016] [Accepted: 12/05/2016] [Indexed: 12/21/2022] Open
Abstract
Heritable profibrotic differentiation of lung fibroblasts is a key mechanism of idiopathic pulmonary fibrosis (IPF). Its mechanisms are yet to be fully understood. In this study, individual data from four independent microarray studies comparing the transcriptome of fibroblasts cultured in vitro from normal (total n = 20) and IPF (total n = 20) human lung were compiled for meta-analysis following normalization to z-scores. One hundred and thirteen transcripts were upregulated and 115 were downregulated in IPF fibroblasts using the Significance Analysis of Microrrays algorithm with a false discovery rate of 5%. Downregulated genes were highly enriched for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional classes related to inflammation and immunity such as Defense response to virus, Influenza A, tumor necrosis factor (TNF) mediated signaling pathway, interferon-inducible absent in melanoma2 (AIM2) inflammasome as well as Apoptosis. Although upregulated genes were not enriched for any functional class, select factors known to play key roles in lung fibrogenesis were overexpressed in IPF fibroblasts, most notably connective tissue growth factor (CTGF) and serum response factor (SRF), supporting their role as drivers of IPF. The full data table is available as a supplement.
Collapse
Affiliation(s)
- Laurent Plantier
- Centre d'Étude des Pathologies Respiratoires-CEPR, Institut National de la Santé et de la Recherche Médicale-INSERM, Unité Mixte de Recherche-UMR1100, Labex Mabimprove, 37000 Tours, France.
- Université François Rabelais, 37000 Tours, France.
- Centre Hospitalier Régional Universitaire-CHRU de Tours, Hôpital Bretonneau, Service de Pneumologie et Explorations Fonctionnelles Respiratoires, 37000 Tours, France.
| | - Hélène Renaud
- Institut National de la Santé et de la Recherche Médicale-INSERM, Unité Mixte de Recherche-UMR1152, Labex Inflamex, 75018 Paris, France.
| | - Renaud Respaud
- Centre d'Étude des Pathologies Respiratoires-CEPR, Institut National de la Santé et de la Recherche Médicale-INSERM, Unité Mixte de Recherche-UMR1100, Labex Mabimprove, 37000 Tours, France.
- Université François Rabelais, 37000 Tours, France.
- Centre Hospitalier Régional Universitaire-CHRU de Tours, Hôpital Trousseau, Service de Pharmacie, 37170 Chambray-les-Tours, France.
| | - Sylvain Marchand-Adam
- Centre d'Étude des Pathologies Respiratoires-CEPR, Institut National de la Santé et de la Recherche Médicale-INSERM, Unité Mixte de Recherche-UMR1100, Labex Mabimprove, 37000 Tours, France.
- Université François Rabelais, 37000 Tours, France.
- Centre Hospitalier Régional Universitaire-CHRU de Tours, Hôpital Bretonneau, Service de Pneumologie et Explorations Fonctionnelles Respiratoires, 37000 Tours, France.
| | - Bruno Crestani
- Institut National de la Santé et de la Recherche Médicale-INSERM, Unité Mixte de Recherche-UMR1152, Labex Inflamex, 75018 Paris, France.
- Université Paris Diderot, PRES Sorbonne Paris Cité, 75018 Paris, France.
- AP-HP, Hôpital Bichat, Service de Pneumologie A, DHU FIRE, 75018 Paris, France.
| |
Collapse
|
43
|
Riccobono D, Agay D, François S, Scherthan H, Drouet M, Forcheron F. Contribution of INTRAMUSCULAR Autologous Adipose Tissue-Derived Stem Cell Injections to Treat Cutaneous Radiation Syndrome: Preliminary Results. HEALTH PHYSICS 2016; 111:117-126. [PMID: 27356055 DOI: 10.1097/hp.0000000000000515] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cutaneous radiation syndrome caused by high dose located irradiation is characterized by delayed symptoms, incomplete wound healing, and poor revascularization. Subcutaneous adipose tissue derived stromal/stem cells have been shown to improve skin repair in a minipig model of cutaneous radiation syndrome despite a subcutaneous defect being a consequence of radio-induced muscular fibrosis. Based on the pro-myogenic potential of stromal/stem cells, a new protocol combining subcutaneous and intramuscular injections was evaluated in a preliminary study. Six female minipigs were locally irradiated at the dose of 50 Gy using a Co source (0.6 Gy min) and randomly divided into two groups. Three animals received the vehicle (phosphate-buffer-saline solution) and three animals received three injections of 75 × 10 adipose tissue derived stromal/stem cells each time (day 25, 46, and 66 post-irradiation). Pigs were euthanized on day 76 post-irradiation before development of clinical skin symptoms. All minipigs exhibited a homogeneous skin evolution. Macroscopic observation of irradiated muscles showed prominent fibrosis and necrosis areas in controls as opposed to adipose tissue-derived stromal/stem cells injected animals. Moreover, muscle biopsy analysis highlighted a recruitment of myofibroblasts (Immune Reactive Score: p < 0.01), an interleukin 10 secretion and a muscle regeneration pathway activation after intramuscular injections of adipose tissue-derived stromal/stem cells (western-blot: respectively, 200-fold change difference and twofold higher in treated animals). Globally, these preliminary data suggest that intramuscular injections of adipose tissue-derived stromal/stem cells improve muscle regeneration in the cutaneous-radiation syndrome. Further work is ongoing to evaluate this therapeutic strategy on a larger animal number with a longer clinical follow-up.
Collapse
Affiliation(s)
- Diane Riccobono
- *Institut de Recherche Biomédicale des Armées (IRBA), Département des Effets Biologiques des Rayonnements, BP 73, Brétigny sur Orge Cedex, France; †Clinatec, 17 rue des Martyrs 38054, Grenoble cedex, France; ‡Institut für Radiobiologie der Bundeswehr, Neuherbergstraße 11, 80937 München, Germany
| | | | | | | | | | | |
Collapse
|
44
|
Chung EJ, Sowers A, Thetford A, McKay-Corkum G, Chung SI, Mitchell JB, Citrin DE. Mammalian Target of Rapamycin Inhibition With Rapamycin Mitigates Radiation-Induced Pulmonary Fibrosis in a Murine Model. Int J Radiat Oncol Biol Phys 2016; 96:857-866. [PMID: 27663762 DOI: 10.1016/j.ijrobp.2016.07.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/18/2016] [Accepted: 07/21/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Radiation-induced pulmonary fibrosis (RIPF) is a late toxicity of therapeutic radiation. Signaling of the mammalian target of rapamycin drives several processes implicated in RIPF, including inflammatory cytokine production, fibroblast proliferation, and epithelial senescence. We sought to determine if mammalian target of rapamycin inhibition with rapamycin would mitigate RIPF. METHODS AND MATERIALS C57BL/6NCr mice received a diet formulated with rapamycin (14 mg/kg food) or a control diet 2 days before and continuing for 16 weeks after exposure to 5 daily fractions of 6 Gy of thoracic irradiation. Fibrosis was assessed with Masson trichrome staining and hydroxyproline assay. Cytokine expression was evaluated by quantitative real-time polymerase chain reaction. Senescence was assessed by staining for β-galactosidase activity. RESULTS Administration of rapamycin extended the median survival of irradiated mice compared with the control diet from 116 days to 156 days (P=.006, log-rank test). Treatment with rapamycin reduced hydroxyproline content compared with the control diet (irradiation plus vehicle, 45.9 ± 11.8 μg per lung; irradiation plus rapamycin, 21.4 ± 6.0 μg per lung; P=.001) and reduced visible fibrotic foci. Rapamycin treatment attenuated interleukin 1β and transforming growth factor β induction in irradiated lungs compared with the control diet. Type II pneumocyte senescence after irradiation was reduced with rapamycin treatment at 16 weeks (3-fold reduction at 16 weeks, P<.001). CONCLUSIONS Rapamycin protected against RIPF in a murine model. Rapamycin treatment reduced inflammatory cytokine expression, extracellular matrix production, and senescence in type II pneumocytes.
Collapse
Affiliation(s)
- Eun Joo Chung
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Anastasia Sowers
- Radiation Biology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Angela Thetford
- Radiation Biology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Grace McKay-Corkum
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Su I Chung
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Deborah E Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
45
|
Wang W, Luo J, Sheng W, Xue J, Li M, Ji J, Liu P, Zhang X, Cao J, Zhang S. Proteomic Profiling of Radiation-Induced Skin Fibrosis in Rats: Targeting the Ubiquitin-Proteasome System. Int J Radiat Oncol Biol Phys 2016; 95:751-60. [PMID: 27045812 DOI: 10.1016/j.ijrobp.2016.01.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 01/05/2016] [Accepted: 01/12/2016] [Indexed: 01/31/2023]
Abstract
PURPOSE To investigate the molecular changes underlying the pathogenesis of radiation-induced skin fibrosis. METHODS AND MATERIALS Rat skin was irradiated to 30 or 45 Gy with an electron beam. Protein expression in fibrotic rat skin and adjacent normal tissues was quantified by label-free protein quantitation. Human skin cells HaCaT and WS-1 were treated by x-ray irradiation, and the proteasome activity was determined with a fluorescent probe. The effect of proteasome inhibitors on Transforming growth factor Beta (TGF-B) signaling was measured by Western blot and immunofluorescence. The efficacy of bortezomib in wound healing of rat skin was assessed by the skin injury scale. RESULTS We found that irradiation induced epidermal and dermal hyperplasia in rat and human skin. One hundred ninety-six preferentially expressed and 80 unique proteins in the irradiated fibrotic skin were identified. Through bioinformatic analysis, the ubiquitin-proteasome pathway showed a significant fold change and was investigated in greater detail. In vitro experiments demonstrated that irradiation resulted in a decline in the activity of the proteasome in human skin cells. The proteasome inhibitor bortezomib suppressed profibrotic TGF-β downstream signaling but not TGF-β secretion stimulated by irradiation in HaCaT and WS-1 cells. Moreover, bortezomib ameliorated radiation-induced skin injury and attenuated epidermal hyperplasia. CONCLUSION Our findings illustrate the molecular changes during radiation-induced skin fibrosis and suggest that targeting the ubiquitin-proteasome system would be an effective countermeasure.
Collapse
Affiliation(s)
- Wenjie Wang
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China; Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| | - Judong Luo
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, China
| | - Wenjiong Sheng
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jiao Xue
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Ming Li
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jiang Ji
- Department of Dermatology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Pengfei Liu
- Department of Gastroenterology, the Affiliated Jiangyin Hospital of Southeast University, Jiangyin, China
| | - Xueguang Zhang
- Institute of Medical Biotechnology and Jiangsu Stem Cell Key Laboratory, Medical College of Soochow University, Suzhou, China
| | - Jianping Cao
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Shuyu Zhang
- School of Radiation Medicine and Protection and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China; Cyrus Tang Hematology Center, Soochow University, Suzhou, China.
| |
Collapse
|
46
|
Simoniello P, Wiedemann J, Zink J, Thoennes E, Stange M, Layer PG, Kovacs M, Podda M, Durante M, Fournier C. Exposure to Carbon Ions Triggers Proinflammatory Signals and Changes in Homeostasis and Epidermal Tissue Organization to a Similar Extent as Photons. Front Oncol 2016; 5:294. [PMID: 26779439 PMCID: PMC4705223 DOI: 10.3389/fonc.2015.00294] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/10/2015] [Indexed: 12/27/2022] Open
Abstract
The increasing application of charged particles in radiotherapy requires a deeper understanding of early and late side effects occurring in skin, which is exposed in all radiation treatments. We measured cellular and molecular changes related to the early inflammatory response of human skin irradiated with carbon ions, in particular cell death induction and changes in differentiation and proliferation of epidermal cells during the first days after exposure. Model systems for human skin from healthy donors of different complexity, i.e., keratinocytes, coculture of skin cells, 3D skin equivalents, and skin explants, were used to investigate the alterations induced by carbon ions (spread-out Bragg peak, dose-averaged LET 100 keV/μm) in comparison to X-ray and UV-B exposure. After exposure to ionizing radiation, in none of the model systems, apoptosis/necrosis was observed. Carbon ions triggered inflammatory signaling and accelerated differentiation of keratinocytes to a similar extent as X-rays at the same doses. High doses of carbon ions were more effective than X-rays in reducing proliferation and inducing abnormal differentiation. In contrast, changes identified following low-dose exposure (≤0.5 Gy) were induced more effectively after X-ray exposure, i.e., enhanced proliferation and change in the polarity of basal cells.
Collapse
Affiliation(s)
- Palma Simoniello
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung , Darmstadt , Germany
| | - Julia Wiedemann
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany; Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Joana Zink
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung , Darmstadt , Germany
| | - Eva Thoennes
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung , Darmstadt , Germany
| | - Maike Stange
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung , Darmstadt , Germany
| | - Paul G Layer
- Department of Biology, Technische Universität Darmstadt , Darmstadt , Germany
| | | | - Maurizio Podda
- Department of Dermatology, Darmstadt Hospital , Darmstadt , Germany
| | - Marco Durante
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany; Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Claudia Fournier
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany; Hochschule Darmstadt, Darmstadt, Germany
| |
Collapse
|
47
|
Dixon D, Coates J, del Carpio Pons A, Horabin J, Walker A, Abdul N, Kalson NS, Brewster NT, Weir DJ, Deehan DJ, Mann DA, Borthwick LA. A potential mode of action for Anakinra in patients with arthrofibrosis following total knee arthroplasty. Sci Rep 2015; 5:16466. [PMID: 26553966 PMCID: PMC4639732 DOI: 10.1038/srep16466] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 10/05/2015] [Indexed: 12/21/2022] Open
Abstract
Arthrofibrosis is a fibroproliferative disease characterised by excessive deposition of extracellular matrix components intra-articularly leading to pain and restricted range of movement. Although frequently observed following total knee arthroplasty (TKA) no therapeutic options exist. A pilot study demonstrated that intra-articular injection of Anakinra, an IL-1R antagonist, improved range of movement and pain in patients with arthrofibrosis however the mechanism of action is unknown. We hypothesise that IL-1α/β will drive an inflammatory phenotype in fibroblasts isolated from the knee, therefore identifying a potential mechanism of action for Anakinra in arthrofibrosis following TKA. Fibroblasts isolated from synovial membranes and infra-patellar fat pad of patients undergoing TKA express high levels of IL-1R1. Stimulation with IL-1α/β induced a pro-inflammatory phenotype characterised by increased secretion of GMCSF, IL-6 and IL-8. No significant difference in the inflammatory response was observed between fibroblasts isolated from synovial membrane or infra-patellar fat pad. IL-1α/β treatments induced a pro-inflammatory phenotype in fibroblasts from both synovial membrane and infra-patellar fat pad and therefore Anakinra can likely have an inhibitory effect on fibroblasts present in both tissues in vivo. It is also likely that fibroblast responses in the tissues are controlled by IL-1α/β availability and not their ability to respond to it.
Collapse
Affiliation(s)
- David Dixon
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Jonathon Coates
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Alicia del Carpio Pons
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Joanna Horabin
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Andrew Walker
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Nicole Abdul
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, High Heaton, Newcastle upon Tyne NE7 7DN, UK
| | - Nicholas S Kalson
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, High Heaton, Newcastle upon Tyne NE7 7DN, UK
| | - Nigel T Brewster
- Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, High Heaton, Newcastle upon Tyne NE7 7DN, UK
| | - David J Weir
- Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, High Heaton, Newcastle upon Tyne NE7 7DN, UK
| | - David J Deehan
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Musculoskeletal Unit, Freeman Hospital, Newcastle Hospitals, NHS Trust, High Heaton, Newcastle upon Tyne NE7 7DN, UK
| | - Derek A Mann
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Lee A Borthwick
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
48
|
Prophylactic amifostine prevents a pathologic vascular response in a murine model of expander-based breast reconstruction. J Plast Reconstr Aesthet Surg 2015; 69:234-40. [PMID: 26631290 DOI: 10.1016/j.bjps.2015.10.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/13/2015] [Indexed: 11/22/2022]
Abstract
BACKGROUND Although expander-based breast reconstruction is the most commonly used method of reconstruction worldwide, it continues to be plagued with complication rates as high as 60% when radiotherapy is implemented. We hypothesized that quantitative measures of radiotherapy-induced vascular injury can be mitigated by utilizing amifostine in a murine model of expander-based breast reconstruction. METHODS 30 rats were divided into three groups: expander placement (Control), expander placement followed by radiotherapy (XRT), and expander placement followed by radiotherapy with amifostine (AMF/XRT). All groups underwent placement of a sub-latissimus tissue expander. After a 45 day recovery period, all groups underwent vascular perfusion and micro-CT analysis. RESULTS Micro-CT analysis was used to calculate vessel volume fraction (VVF), vessel number (VN), and vessel separation (VSp). A significant increase in VN was seen in the XRT group as compared to the Control (p = 0.021) and the AMF/XRT (p = 0.027). There was no difference between Control and AMF/XRT (p = 0.862). VVF was significantly higher in XRT than either Control (p = 0.043) and AMF/XRT (p = 0.040), however no difference was seen between Control and AMF/XRT (p = 0.980). VSp of XRT was smaller when compared to both Control and AMF/XRT specimens (p = 0.05 and p = 0.048, respectively), and no difference was seen between Control and AMF/XRT (p = 0.339). CONCLUSIONS Amifostine administered prior to radiotherapy preserved vascular metrics similar to those of non-radiated specimens. Elevated vascularity demonstrated within the XRT group was not seen in either the Control or AMF/XRT groups. These results indicate that amifostine protects soft tissue in our model from a radiotherapy-induced pathologic vascular response.
Collapse
|
49
|
Islam A, Bolduc DL, Zhai M, Kiang JG, Swift JM. Captopril Increases Survival after Whole-Body Ionizing Irradiation but Decreases Survival when Combined with Skin-Burn Trauma in Mice. Radiat Res 2015; 184:273-9. [PMID: 26305295 DOI: 10.1667/rr14113.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Past and recent radiation events have involved a high incidence of radiation combined injury where victims often succumb to serious infections as a consequence of bacterial translocation and subsequent sepsis. The risk of infection is exacerbated in radiation combined skin-burn injury (RCI), which increase vulnerability. Furthermore, no suitable countermeasures for radiation combined skin-burn injury have been established. In this study, we evaluated captopril as a potential countermeasure to radiation combined skin-burn injury. Captopril is an FDA-approved angiotensin-converting enzyme inhibitor that was previously reported to stimulate hematopoietic recovery after exposure to ionizing radiation. Female B6D2F1/J mice were whole-body bilateral (60)Co gamma-photon irradiated (dose rate of 0.4 Gy/min) with 9.5 Gy (LD70/30 for RCI), followed by nonlethal dorsal skin-burn injury under anesthesia (approximately 15% total-body surface-area burn). Mice were provided with acidified drinking water with or without dissolved captopril (0.55 g/l) for 30 days immediately after injury and were administered topical gentamicin (0.1% cream; day 1-10) and oral levofloxacin (90-100 mg/kg; day 3-16). Surviving mice were euthanized on day 30 after analyses of water consumption, body weight and survival. Our data demonstrate that, while treatment with captopril did mitigate mortality induced by radiation injury (RI) alone (55% captopril vs. 80% vehicle; n = 20, P < 0.05), it also resulted in decreased survival after radiation combined skin-burn injury (22% captopril vs. 41% vehicle; n = 22, P < 0.05). Moreover, captopril administration via drinking water produced an uneven dosage pattern among the different injury groups ranging from 74 ± 5.4 to 115 ± 2.2 mg/kg/day. Captopril treatment also did not counteract the negative alterations in hematology, splenocytes or bone marrow cellularity after either radiation injury or radiation combined skin-burn injury. These data suggest that captopril may exert its actions differently between the two injury models (RI vs. RCI) and that captopril dosing, when combined with topical and systemic antibiotic treatments, may not be a suitable countermeasure for RCI.
Collapse
Affiliation(s)
- Aminul Islam
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - David L Bolduc
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - Min Zhai
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - Juliann G Kiang
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland.,b Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,c Department of Radiation Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Joshua M Swift
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland.,c Department of Radiation Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,d Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
50
|
Paun A, Kunwar A, Haston CK. Acute adaptive immune response correlates with late radiation-induced pulmonary fibrosis in mice. Radiat Oncol 2015; 10:45. [PMID: 25889053 PMCID: PMC4342202 DOI: 10.1186/s13014-015-0359-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/16/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The lung response to radiation exposure can involve an immediate or early reaction to the radiation challenge, including cell death and an initial immune reaction, and can be followed by a tissue injury response, of pneumonitis or fibrosis, to this acute reaction. Herein, we aimed to determine whether markers of the initial immune response, measured within days of radiation exposure, are correlated with the lung tissue injury responses occurring weeks later. METHODS Inbred strains of mice known to be susceptible (KK/HIJ, C57BL/6J, 129S1/SvImJ) or resistant (C3H/HeJ, A/J, AKR/J) to radiation-induced pulmonary fibrosis and to vary in time to onset of respiratory distress post thoracic irradiation (from 10-23 weeks) were studied. Mice were untreated (controls) or received 18 Gy whole thorax irradiation and were euthanized at 6 h, 1d or 7 d after radiation treatment. Pulmonary CD4+ lymphocytes, bronchoalveolar cell profile & cytokine level, and serum cytokine levels were assayed. RESULTS Thoracic irradiation and inbred strain background significantly affected the numbers of CD4+ cells in the lungs and the bronchoalveolar lavage cell differential of exposed mice. At the 7 day timepoint greater numbers of pulmonary Th1 and Th17 lymphocytes and reduced lavage interleukin17 and interferonγ levels were significant predictors of late stage fibrosis. Lavage levels of interleukin-10, measured at the 7 day timepoint, were inversely correlated with fibrosis score (R=-0.80, p=0.05), while serum levels of interleukin-17 in control mice significantly correlated with post irradiation survival time (R=0.81, p=0.04). Lavage macrophage, lymphocyte or neutrophil counts were not significantly correlated with either of fibrosis score or time to respiratory distress in the six mouse strains. CONCLUSION Specific cytokine and lymphocyte levels, but not strain dependent lavage cell profiles, were predictive of later radiation-induced lung injury in this panel of inbred strains.
Collapse
Affiliation(s)
- Alexandra Paun
- Department of Human Genetics, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.
| | - Amit Kunwar
- Department of Human Genetics, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.
| | - Christina K Haston
- Department of Human Genetics, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada. .,Department of Medicine, Meakins-Christie Laboratories, McGill University, 3626 St. Urbain, H2X 2P2, Montreal, QC, Canada.
| |
Collapse
|