1
|
Zeng Y, Li C, Yang F, Zhang L, Xu W, Wang L, Wu A, Zou W, Wu J, Huang F. Sheng Xue Ning as a Novel Agent that Promotes SCF-Driven Hematopoietic Stem/Progenitor Cell Proliferation to Promote Erythropoiesis. Biomolecules 2024; 14:1147. [PMID: 39334913 PMCID: PMC11429878 DOI: 10.3390/biom14091147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Stimulating erythropoiesis is essential in the treatment of various types of anemia. Sheng Xue Ning (SXN) is commonly used in China as an iron supplement to treat iron deficiency anemia, renal anemia, and anemia in pregnancy. This research reports a novel effect of SXN in enhancing the proliferation of hematopoietic stem/progenitor cell (HSPC) to promote erythropoiesis in the bone marrow, which is distinct from conventional iron supplements that primarily aid in the maturation of red blood cells. Employing a model of hematopoietic dysfunction induced by X-ray exposure, we evaluated the efficacy of SXN in restoring hematopoietic function. SXN significantly promoted the recovery of peripheral erythroid cells and enhanced the proliferation and differentiation of Lin-/c-KIT+/Sca-1+ HSPC in mice exposed to X-ray irradiation. Our results showed that SXN elevated the expression of stem cell factor (SCF) and activated the SCF/c-KIT/PI3K/AKT signaling pathway, facilitating the proliferation and differentiation of HSPC. In vitro, SXN markedly enhanced the proliferation of bone marrow nucleated cell (BMNC) and the colony-forming capacity of BFU-E, CFU-E, and CFU-GM, while also elevating the expression of proteins involved in the SCF/c-KIT/PI3K/AKT pathway in BMNC. Additionally, SXN enhanced the proliferation and differentiation of mesenchymal stem cell (MSC) and increased SCF secretion. In conclusion, SXN demonstrates the capacity to enhance erythropoiesis by upregulating SCF expression, thereby promoting HSPC proliferation and differentiation via the SCF/c-KIT/PI3K/AKT pathway. SXN may offer a new strategy for improving the activity of HSPC and promoting erythropoiesis in the treatment of hematopoiesis disorders.
Collapse
Affiliation(s)
- Yueying Zeng
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Chunlu Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fei Yang
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Ling Zhang
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wanqi Xu
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Long Wang
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Anguo Wu
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Wenjun Zou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Feihong Huang
- Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
2
|
Moloudi K, Azariasl S, Abrahamse H, George BP, Yasuda H. Expected role of photodynamic therapy to relieve skin damage in nuclear or radiological emergency: Review. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 110:104517. [PMID: 39032581 DOI: 10.1016/j.etap.2024.104517] [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: 03/18/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Nuclear and radiological accidents can occur due to poor management, in transportation, radiation therapy and nuclear wards in hospitals, leading to extreme radiation exposure and serious consequences for human health. Additionally, in many of previous radiological accidents, skin damage was observed in patients and survivors due to the high radiation exposure. However, as part of a medical countermeasures in a nuclear/radiological emergency, it is critical to plan for the treatment of radiation-induced skin damage. Hence, the new, non-invasive technology of photodynamic therapy (PDT) is projected to be more effectively used for treating skin damage caused by high-dose radiation. PDT plays an important role in treating, repairing skin damage and promoting wound healing as evidenced by research. This review, highlighted and recommended potential impacts of PDT to repair and decrease radiation-induced skin tissue damage. Moreover, we have suggested some photosensitizer (PS) agent as radio-mitigator drugs to decrease radiobiological effects.
Collapse
Affiliation(s)
- Kave Moloudi
- Laser Research Centre, Faculty of Health Science, Doornfontein Campus, University of Johannesburg, Johannesburg 2028, South Africa
| | - Samayeh Azariasl
- Department of Radiation Biophysics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Kasumi, Minami-ku 734-8553, Japan
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Science, Doornfontein Campus, University of Johannesburg, Johannesburg 2028, South Africa
| | - Blassan P George
- Laser Research Centre, Faculty of Health Science, Doornfontein Campus, University of Johannesburg, Johannesburg 2028, South Africa.
| | - Hiroshi Yasuda
- Department of Radiation Biophysics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Kasumi, Minami-ku 734-8553, Japan
| |
Collapse
|
3
|
Singh B, Patwardhan RS, Pal D, Maurya DK, Singh BG, Checker R, Sharma D, Sandur SK. Repurposing of FDA approved kinase inhibitor bosutinib for mitigation of radiation induced damage via inhibition of JNK pathway. Toxicol Appl Pharmacol 2024; 482:116792. [PMID: 38142783 DOI: 10.1016/j.taap.2023.116792] [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: 10/20/2023] [Revised: 12/09/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Radiotherapy is a common modality for cancer treatment. However, it is often associated with normal tissue toxicity in 20-80% of the patients. Radioprotectors can improve the outcome of radiotherapy by selectively protecting normal cells against radiation toxicity. In the present study, compound libraries containing 54 kinase inhibitors and 80 FDA-approved drugs were screened for radioprotection of lymphocytes using high throughput cell analysis. A second-generation FDA-approved kinase inhibitor, bosutinib, was identified as a potential radioprotector for normal cells. The radioprotective efficacy of bosutinib was evinced from a reduction in radiation induced DNA damage, caspase-3 activation, DNA fragmentation and apoptosis. Oral administration of bosutinib protected mice against whole body irradiation (WBI) induced morbidity and mortality. Bosutinib also reduced radiation induced bone-marrow aplasia and hematopoietic damage in mice exposed to 4 Gy and 6 Gy dose of WBI. Mechanistic studies revealed that the radioprotective action of bosutinib involved interaction with cellular thiols and modulation of JNK pathway. The addition of glutathione and N-acetyl cysteine significantly reduced the radioprotective efficacy of bosutinib. Moreover, bosutinib did not protect cancer cells against radiation induced toxicity. On the contrary, bosutinib per se exhibited anticancer activity against human cancer cell lines. The results highlight possible use of bosutinib as a repurposable radioprotective agent for mitigation of radiation toxicity in cancer patients undergoing radiotherapy.
Collapse
Affiliation(s)
- Babita Singh
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Debojyoti Pal
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Dharmendra K Maurya
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Beena G Singh
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India; Radiation and Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Rahul Checker
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
4
|
Khodamoradi E, Rahmani N, Rashidi K, Najafi M, Shahsavari S, Mohammadi M. Exploring the Potential of Metformin in Mitigating Radiation-induced Gastrointestinal and Hematopoietic System Injury in Rats After Whole-body X-ray Radiation: An Experimental Study. Curr Radiopharm 2024; 17:200-208. [PMID: 38231059 DOI: 10.2174/0118744710261673231115062547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/09/2023] [Accepted: 10/02/2023] [Indexed: 01/18/2024]
Abstract
BACKGROUND The modern world faces a growing concern about the possibility of accidental radiation events. The Hematopoietic system is particularly vulnerable to radiationinduced apoptosis, which can lead to death. Metformin, a drug used to treat diabetes, has been shown to protect normal cells and tissues from the toxic effects of radiation. This study aimed to evaluate the effectiveness of metformin in mitigating radiation injury to the gastrointestinal and hematological systems of rats. MATERIALS AND METHODS The study involved 73 male rats. After total body irradiation with 7.5 Gy of X-rays, rats were treated with metformin. Seven days later, the rats were sacrificed and blood samples were taken for evaluation. RESULTS The study found that metformin was not effective in mitigating radiation injury. The histopathological assessment showed no significant changes in goblet cell injury, villi shortening, inflammation, or mucous layer thickness. In terms of biochemical evaluation, metformin did not significantly affect oxidative stress markers, but irradiation increased the mean MDA level in the radiation group. The complete blood count revealed a significant decrease in WBC and platelet, counts in the radiation group compared to the control group, but no significant difference was found between the radiation and radiation + metformin groups. CONCLUSION In conclusion, metformin may not be a good option for reducing radiation toxicity after accidental exposure. Despite treatment, there was no improvement in platelet, white blood cell, and lymphocyte counts, nor was there any decrease in oxidative stress. Further research is needed to explore other potential treatments for radiation injury.
Collapse
Affiliation(s)
- Ehsan Khodamoradi
- Department of Radiology and Nuclear Medicine, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nafiseh Rahmani
- Student Research Committee, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Khodabakhsh Rashidi
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Najafi
- Department of Radiology and Nuclear Medicine, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soodeh Shahsavari
- Department of Health Information Technology, Faculty of Allied Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Mohammadi
- Student Research Committee, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
5
|
Dong Y, Zhang Y, Wang X, Li W, Zhang J, Lu L, Dong H, Fan S, Meng A, Li D. The protective effects of Xuebijing injection on intestinal injuries of mice exposed to irradiation. Animal Model Exp Med 2022; 5:565-574. [PMID: 36376997 PMCID: PMC9773304 DOI: 10.1002/ame2.12285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Gastrointestinal (GI) injury is one of the most common side effects of radiotherapy. However, there is no ideal therapy method except for symptomatic treatment in the clinic. Xuebijing (XBJ) is a traditional Chinese medicine, used to treat sepsis by injection. In this study, the protective effects of XBJ on radiation-induced intestinal injury (RIII) and its mechanism were explored. METHODS The effect of XBJ on survival of irradiated C57BL/6 mice was monitored. Histological changes including the number of crypts and the length of villi were evaluated by H&E. The expression of Lgr5+ intestinal stem cells (ISCs), Ki67+ cells, villin and lysozymes were examined by immunohistochemistry. The expression of cytokines in the intestinal crypt was detected by RT-PCR. DNA damage and apoptosis rates in the small intestine were also evaluated by immunofluorescence. RESULTS In the present study, XBJ improved the survival rate of the mice after 8.0 and 9.0 Gy total body irradiation (TBI). XBJ attenuated structural damage of the small intestine, maintained regenerative ability and promoted proliferation and differentiation of crypt cells, decreased apoptosis rate and reduced DNA damage in the intestine. Elevation of IL-6 and TNF-α was limited, but IL-1, TNF-𝛽 and IL-10 levels were increased in XBJ-treated group after irradiation. The expression of Bax and p53 were decreased after XBJ treatment. CONCLUSIONS Taken together, XBJ provides a protective effect on RIII by inhibiting inflammation and blocking p53-related apoptosis pathway.
Collapse
Affiliation(s)
- Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - YuanYang Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Xinyue Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Wenxuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Junling Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Lu Lu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Hui Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Aimin Meng
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing Engineering Research Center for Laboratory Animal Models of Human Critical Diseases, National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC)BeijingChina
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| |
Collapse
|
6
|
Jit BP, Pattnaik S, Arya R, Dash R, Sahoo SS, Pradhan B, Bhuyan PP, Behera PK, Jena M, Sharma A, Agrawala PK, Behera RK. Phytochemicals: A potential next generation agent for radioprotection. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154188. [PMID: 36029645 DOI: 10.1016/j.phymed.2022.154188] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/13/2022] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Radiation hazards are accountable for extensive damage in the biological system and acts as a public health burden. Owing to the rapid increasing in radiation technology, both Ionizing radiation (IR) from natural and man made source poses detrimental outcome to public health. IR releases free radicals which induces oxidative stress and deleterious biological damage by modulating radiation induced signalling intermediates. The efficacy of existing therapeutic approach and treatment strategy are limited owing to their toxicity and associated side effects. Indian system of traditional medicine is enriched with prospective phytochemicals with potential radioprotection ability. PURPOSE The present review elucidated and summarized the potential role of plant derived novel chemical compound with prospective radioprotective potential. METHOD So far as the traditional system of Indian medicine is concerned, plant kingdom is enriched with potential bioactive molecules with diverse pharmacological activities. We reviewed several compounds mostly secondary metabolites from plant origin using various search engines. RESULTS Both compounds from land plants and marine source exhibited antioxidant antiinflammatory, free radical scavenging ability. These compounds have tremendous potential in fine-tuning of several signalling intermediates, which are actively participated in the progression and development of a pathological condition associated with radiation stress. CONCLUSION Development and explore of an operational radioprotective agent from originated from plant source that can be used as a novel molecular tool to eliminate the widespread damage caused by space exploration, ionizing radiation, nuclear war and radiotherapy has been significantly appreciated. Through extensive literature search we highlighted several compounds from both land plant and marine origin can be implemented for a better therapeutic potential against radiation induced injury. Furthermore, extensive clinical trials must be carried out in near future for better therapeutic modality and clinical efficacy.
Collapse
Affiliation(s)
- Bimal Prasad Jit
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India; School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India
| | - Subhaswaraj Pattnaik
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India; Centre of Excellence in Natural Products and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Rakesh Arya
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India; School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India
| | - Rutumbara Dash
- Departement of Gastroenterology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | | | - Biswajita Pradhan
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur, Odisha 760007, India; Department of Biotechnology, Sangmyung University, Seoul 03016, South Korea
| | - Prajna Paramita Bhuyan
- Department of Botany, Maharaja Sriram Chandra Bhanja Deo University, Baripada, Odisha 757003, India
| | - Pradyota Kumar Behera
- Department of Chemistry, Berhampur University, Bhanja Bihar, Berhampur, Odisha 760007, India
| | - Mrutyunjay Jena
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur, Odisha 760007, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Paban Kumar Agrawala
- Institute of Nuclear Medicine and Allied Science, Defence Research and Development Organization, New Delhi 110054, India
| | | |
Collapse
|
7
|
Guo JJ, Liu N, Ma Z, Gong ZJ, Liang YL, Cheng Q, Zhong XG, Yao ZJ. Dose-Response Effects of Low-Dose Ionizing Radiation on Blood Parameters in Industrial Irradiation Workers. Dose Response 2022; 20:15593258221105695. [PMID: 35693871 PMCID: PMC9174562 DOI: 10.1177/15593258221105695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 05/17/2022] [Indexed: 11/24/2022] Open
Abstract
While previous studies have focused on the health effects of occupational
exposure of radiations on medical radiation workers, few have analyzed the
dose-response relationship between low radiation doses and changes in blood
parameters. Even fewer studies have been conducted on industrial worker
populations. Using a prospective cohort study design, this study collected
health examination reports and personal dose monitoring data from 705 industrial
irradiation workers who underwent regular physical examinations at Dongguan
Sixth People’s Hospital. The dose-response effects of low-dose ionizing
radiation on blood parameters were assessed using a generalized linear model and
restricted cubic spline model. Red blood cell counts decreased then increased,
before decreasing again with increasing ionizing radiation. This was in contrast
to the curve of the total platelet count after irradiation. Additionally, a
radiation dose of 2.904 mSv was the turning point for the nonlinear curve of
hemoglobin count changes. In conclusion, long-term, low-dose ionizing radiation
affects blood cell levels in industrial irradiation workers. There is a
nonlinear dose-response relationship between red blood cell, platelet, and
hemoglobin counts and the cumulative radiation dose. These findings should alert
radiation workers to seek preventive medical treatment before the occurrence of
any serious hematopoietic disease.
Collapse
Affiliation(s)
- Jia-jia Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ning Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zheng Ma
- The Sixth People’s Hospital of Dongguan, Dongguan, China
| | - Zi-jun Gong
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yue-lang Liang
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qi Cheng
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | | | - Zhen-jiang Yao
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
8
|
Mann Z, Sengar M, Verma YK, Rajalingam R, Raghav PK. Hematopoietic Stem Cell Factors: Their Functional Role in Self-Renewal and Clinical Aspects. Front Cell Dev Biol 2022; 10:664261. [PMID: 35399522 PMCID: PMC8987924 DOI: 10.3389/fcell.2022.664261] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/14/2022] [Indexed: 01/29/2023] Open
Abstract
Hematopoietic stem cells (HSCs) possess two important properties such as self-renewal and differentiation. These properties of HSCs are maintained through hematopoiesis. This process gives rise to two subpopulations, long-term and short-term HSCs, which have become a popular convention for treating various hematological disorders. The clinical application of HSCs is bone marrow transplant in patients with aplastic anemia, congenital neutropenia, sickle cell anemia, thalassemia, or replacement of damaged bone marrow in case of chemotherapy. The self-renewal attribute of HSCs ensures long-term hematopoiesis post-transplantation. However, HSCs need to be infused in large numbers to reach their target site and meet the demands since they lose their self-renewal capacity after a few passages. Therefore, a more in-depth understanding of ex vivo HSCs expansion needs to be developed to delineate ways to enhance the self-renewability of isolated HSCs. The multifaceted self-renewal process is regulated by factors, including transcription factors, miRNAs, and the bone marrow niche. A developed classical hierarchical model that outlines the hematopoiesis in a lineage-specific manner through in vivo fate mapping, barcoding, and determination of self-renewal regulatory factors are still to be explored in more detail. Thus, an in-depth study of the self-renewal property of HSCs is essentially required to be utilized for ex vivo expansion. This review primarily focuses on the Hematopoietic stem cell self-renewal pathway and evaluates the regulatory molecular factors involved in considering a targeted clinical approach in numerous malignancies and outlining gaps in the current knowledge.
Collapse
Affiliation(s)
- Zoya Mann
- Independent Researcher, New Delhi, India
| | - Manisha Sengar
- Department of Zoology, Deshbandhu College, University of Delhi, Delhi, India
| | - Yogesh Kumar Verma
- Stem Cell and Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), Delhi, India
| | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Pawan Kumar Raghav
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
9
|
He M, Xu H, Liu G, Yang M, Zhang W, Li Y, Zhang H, Wang C, Zhang Y, Liu X, Xu S, Ding Y, Li Y, Gao Y, Zhang Q. Levistilide A Promotes Expansion of Human Umbilical Cord Blood Hematopoietic Stem Cells by Enhancing Antioxidant Activity. Front Pharmacol 2022; 13:806837. [PMID: 35250558 PMCID: PMC8895481 DOI: 10.3389/fphar.2022.806837] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
Several approaches to expand human hematopoietic stem cells (hHSCs) clinically along with retainable capability of multipotential differentiation have been reported, but only a few have advanced to evaluation in clinical trials, which limits the application of HSC-based therapy. Here we show a phthalide derivative, Levistilide A (LA), can serve as a promising molecule to expand functional human umbilical cord blood (UCB) HSCs ex vivo. An in-house screen identified LA out of nine natural products as an outstanding candidate for hHSCs expansion. Additionally, our data indicated that LA treatment not only increased the numbers of phenotype-defined HSCs, but also enhanced their colony formation ability. Xenotransplantation assays showed that LA treatment could maintain unaffected engraftment of hHSCs with multilineage differentiation capacity. Further experiments revealed that LA enhanced the antioxidant activity of hHSCs by reducing intracellular and mitochondrial reactive oxygen species (ROS) levels. The identification of LA provides a new strategy in solving the clinical issue of limited numbers of UCB HSCs.
Collapse
Affiliation(s)
- Mei He
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hui Xu
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Guangju Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Ming Yang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Wenshan Zhang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yafang Li
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hexiao Zhang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Chaoqun Wang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yiran Zhang
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaolei Liu
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shiqi Xu
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yahui Ding
- College of Chemistry, Nankai University, Tianjin, China
- *Correspondence: Quan Zhang, ; Yingdai Gao, ; Yinghui Li, ; Yahui Ding,
| | - Yinghui Li
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- *Correspondence: Quan Zhang, ; Yingdai Gao, ; Yinghui Li, ; Yahui Ding,
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- *Correspondence: Quan Zhang, ; Yingdai Gao, ; Yinghui Li, ; Yahui Ding,
| | - Quan Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- *Correspondence: Quan Zhang, ; Yingdai Gao, ; Yinghui Li, ; Yahui Ding,
| |
Collapse
|
10
|
Chen G, Che L, Cai X, Zhu P, Ran J. Bioinformatic Analysis Identifies Biomarkers and Treatment Targets in Primary Sjögren's Syndrome Patients with Fatigue. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7697558. [PMID: 35075430 PMCID: PMC8783724 DOI: 10.1155/2022/7697558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022]
Abstract
We aim to identify the common genes, biological pathways, and treatment targets for primary Sjögren's syndrome patients with varying degrees of fatigue features. We select datasets about transcriptomic analyses of primary Sjögren's syndrome (pSS) patients with different degrees of fatigue features and normal controls in peripheral blood. We identify common differentially expressed genes (DEGs) to find shared pathways and treatment targets for pSS patients with fatigue and design a protein-protein interaction (PPI) network by some practical bioinformatic tools. And hub genes are detected based on the PPI network. We perform biological pathway analysis of common genes by Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway. Lastly, potential treatment targets for pSS patients with fatigue are found by the Enrichr platform. We discovered that 27 DEGs are identified in pSS patients with fatigue features and the severe fatigued pSS-specific gene is RTP4. DEGs are mainly localized in the mitochondria, endosomes, endoplasmic reticulum, and cytoplasm and are involved in the biological process by which interferon acts on cells and cells defend themselves against viruses. Molecular functions mainly involve the process of RNA synthesis. The DEGs of pSS are involved in the signaling pathways of viruses such as hepatitis C, influenza A, measles, and EBV. Acetohexamide PC3 UP, suloctidil HL60 UP, prenylamine HL60 UP, and chlorophyllin CTD 00000324 are the four most polygenic drug molecules. PSS patients with fatigue features have specific gene regulation, and chlorophyllin may alleviate fatigue symptoms in pSS patients.
Collapse
Affiliation(s)
- Guangshu Chen
- Department of Endocrinology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| | - Li Che
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xingdong Cai
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ping Zhu
- Department of Endocrinology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| | - Jianmin Ran
- Department of Endocrinology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| |
Collapse
|
11
|
Hanuma Kumar GEN, Kumar SS, Balaji M, Maurya DK, Kesavulu M. Pterocarpus santalinus L. extract mitigates gamma radiation-inflicted derangements in BALB/c mice by Nrf2 upregulation. Biomed Pharmacother 2021; 141:111801. [PMID: 34146850 DOI: 10.1016/j.biopha.2021.111801] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/24/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022] Open
Abstract
Plant-based natural extracts contain several nutrients and bioactive compounds, such as phenolics and flavonoids, that possess various health-promoting activities. This study investigated the effects of polyphenols from Pterocarpus santalinus hydroalcoholic extract (PSHE) against gamma radiation-induced derangements via the upregulation of Nrf2. Ultra High Performance Liquid Chromatography Coupled to High Resolution Mass Spectrometry (UHPLC-HRMS/MS) analysis was performed to identify the possible radioprotectors. In vivo and in vitro studies, namely Real-Time-PCR (RT-PCR) analysis, Reactive Oxygen Species (ROS) scavenging activity, lipid peroxidation and GSH levels, DNA damage and cell death studies, anti-inflammatory (Sandwich ELISA), immunomodulatory studies (antibody staining), and model free radical scavenging assays, were performed. Vanillic acid, protocatechuic acid, para-hydroxybenzoic acid, chlorogenic acid, TNF-α inhibitor (Eudesmin), isoflavone (Daidzein 7-o-glucoside), astragalin (Kaempferol 3-o-glycoside), and other polyphenols were identified in PSHE using UHPLC-HRMS/MS analysis. Prophylactic administration of PSHE (-1 h) rendered more than 33% survival in mice exposed to 8 Gy whole-body-irradiation with increased mice survival and recovery of bone marrow and spleen cellularity. Real-time RT-PCR analysis showed that PSHE treatment (50 µg/mL) upregulated Nrf2, HO-1, and GPX-1 in mice splenocytes. At 50 µg/mL, PSHE reduced ROSscavenging activity, mitochondrial and spleen membrane lipid peroxidation levels, DNA damage, and cell death, and increased GSH levels. At 10 µg/mL, PSHE treatment diminished the content of IL-6 and TNF-α. At 50 µg/mL, PSHE suppressed lymphocyte proliferation. These findings indicate that polyphenols of PSHE possess marked antioxidant, anti-inflammatory, and immunomodulatory capacities, which play important roles in the prevention of radiation damage.
Collapse
Affiliation(s)
- Ghali E N Hanuma Kumar
- Department of Biochemistry, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India
| | - Sandopu Sravan Kumar
- Department of Biochemistry, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India
| | - Meriga Balaji
- Department of Biochemistry, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India.
| | - Dharmendra Kumar Maurya
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India.
| | - Muppuru Kesavulu
- Sree Vidyanikethan Engineering College, Tirupati 517102, Andhra Pradesh, India
| |
Collapse
|
12
|
Ding L, Tan Y, Xu L, Jin Y, Liu Y, Tu H, Zhang D, Wu B, Chen Y, Shen H, Liu L, Huang T, Zhou F. Shengxuening Extracted from Silkworm Excrement Mitigates Myelosuppression via SCF-Mediated JAK2/STAT3 Signaling. Chem Biodivers 2021; 18:e2100139. [PMID: 33973702 DOI: 10.1002/cbdv.202100139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/28/2021] [Indexed: 11/08/2022]
Abstract
Shengxuening (SXN) is a Chinese patent medicine with main ingredients (including chlorophyll derivatives and sodium iron chlorophyllin) extracted from silkworm excrement. SXN exhibited efficacy in clinical trials of renal anemia and iron deficiency anemia; however, the specific mechanisms remain unclear. This study found that SXN increased the number of peripheral blood cells and improved the bone marrow morphology in myelosuppressed mouse model, reversed the reduction in body weight and spleen indices, and increased the serum levels of erythropoietin and granulocyte-macrophage colony-stimulating factor. Quantitative real-time PCR array and Western blot analysis showed the enhanced expression of stem cell factor (SCF), JAK2, and STAT3 in the liver. These results suggested that SXN promoted the recovery of hemopoietic function in myelosuppressed models by increasing the secretion of hematopoietic factors and activating the JAK2/STAT3 pathway. Therefore, this medicine may be applied as therapeutic pharmaceutical drug to mitigate myelosuppression.
Collapse
Affiliation(s)
- Lu Ding
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Yuxin Tan
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Lulin Xu
- Wuhan United Pharmaceutical Co., Ltd., Wuhan, 430071, P. R. China
| | - Yanxia Jin
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Yin Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Honglei Tu
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Dongdong Zhang
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Balu Wu
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Yiran Chen
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Hui Shen
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Li Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Tingting Huang
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhang District, Wuhan, 430071, P. R. China
| |
Collapse
|
13
|
Clark NF, Taylor-Robinson AW. COVID-19 Therapy: Could a Copper Derivative of Chlorophyll a Be Used to Treat Lymphopenia Associated With Severe Symptoms of SARS-CoV-2 Infection? Front Med (Lausanne) 2021; 8:620175. [PMID: 33777973 PMCID: PMC7994343 DOI: 10.3389/fmed.2021.620175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Affiliation(s)
- Nicole F. Clark
- Institute for Applied Ecology, University of Canberra, Bruce, ACT, Australia
- Centre for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Andrew W. Taylor-Robinson
- Infectious Diseases Research Group, School of Health, Medical and Applied Sciences, Central Queensland University, Brisbane, QLD, Australia
- College of Health and Human Sciences, Charles Darwin University, Casuarina, NT, Australia
| |
Collapse
|
14
|
Dashwood RH. Cancer interception by interceptor molecules: mechanistic, preclinical and human translational studies with chlorophylls. Genes Environ 2021; 43:8. [PMID: 33676582 PMCID: PMC7937315 DOI: 10.1186/s41021-021-00180-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/18/2021] [Indexed: 01/14/2023] Open
Abstract
Before 'cancer interception' was first advocated, 'interceptor molecules' had been conceived as a sub-category of preventive agents that interfered with the earliest initiation steps in carcinogenesis. Three decades ago, a seminal review cataloged over fifty synthetic agents and natural products that were known or putative interceptor molecules. Chlorophylls and their derivatives garnered much interest based on the potent antimutagenic activity in the Salmonella assay, and the subsequent mechanistic work that provided proof-of-concept for direct molecular complexes with planar aromatic carcinogens. As the 'interceptor molecule' hypothesis evolved, mechanistic experiments and preclinical studies supported the view that chlorophylls can interact with environmental heterocyclic amines, aflatoxins, and polycyclic aromatic hydrocarbons to limit their uptake and bioavailability in vivo. Support also came from human translational studies involving ultralow dose detection in healthy volunteers, as well as intervention in at-risk subjects. Antimutagenic and antigenotoxic effects of natural and synthetic chlorophylls against small alkylating agents also highlighted the fact that non-interceptor mechanisms existed. This gave impetus to investigations broadly related to free radical scavenging, anti-inflammatory effects, immune modulation and photodynamic therapy. Therapeutic aspects of chlorophylls also were investigated, with evidence for cell cycle arrest and apoptosis in human cancer cells. As the science has evolved, new mechanistic leads continue to support the use and development of chlorophylls and their porphyrin derivatives for cancer interception, beyond the initial interest as interceptor molecules.
Collapse
Affiliation(s)
- Roderick H Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, 2121 West Holcombe Blvd, Houston, TX, 77030, USA.
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX, USA.
| |
Collapse
|
15
|
Joshi HA, Patwardhan RS, Sharma D, Sandur SK, Devarajan PV. Pre-clinical evaluation of an innovative oral nano-formulation of baicalein for modulation of radiation responses. Int J Pharm 2020; 595:120181. [PMID: 33359537 DOI: 10.1016/j.ijpharm.2020.120181] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/27/2020] [Accepted: 12/11/2020] [Indexed: 02/04/2023]
Abstract
There is an unmet medical need for non-toxic and effective radiation countermeasures for prevention of radiation toxicity during planned exposures. We have earlier shown that intraperitoneal administration of baicalein (BCL) offers significant survival benefit in animal model. Safety, tolerability, pharmacokinetics (PK) and pharmacodynamics of baicalein has been reported in pre-clinical model systems and also in healthy human volunteers. However, clinical translation of baicalein is hindered owing to poor bioavailability due to lipophilicity. In view of this, we fabricated and characterized in-situ solid lipid nanoparticles of baicalein (SLNB) with effective drug entrapment and release kinetics. SLNB offered significant protection to murine splenic lymphocytes against 4 Gy ionizing radiation (IR) induced apoptosis. Oral administration of SLNB exhibited ~70% protection to mice against whole body irradiation (WBI 7.5 Gy) induced mortality. Oral relative bioavailability of BCL was enhanced by over ~300% after entrapment in the SLNB as compared to BCL. Oral dosing of SLNB resulted in transient increase in neutrophil abundance in peripheral blood. Interestingly, we observed that treatment of human lung cancer cells (A549) with radioprotective dose of SLNB exhibited radio-sensitization as evinced by decrease in survival and clonogenic potential. Contrary to antioxidant nature of baicalein in normal cells, SLNB treatment induced significant increase in cellular ROS levels in A549 cells probably due to higher uptake and inhibition of TrxR. Thus, a pharmaceutically acceptable SLNB exhibited improved bioavailability, better radioprotection to normal cells and sensitized cancer cells to radiation induced killing as compared to BCL suggesting its possible utility as an adjuvant during cancer radiotherapy.
Collapse
Affiliation(s)
- Harsh A Joshi
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Matunga, Mumbai 400019, India
| | - Raghavendra S Patwardhan
- Radiation Biology and Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India.
| | - Deepak Sharma
- Radiation Biology and Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Santosh K Sandur
- Radiation Biology and Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Padma V Devarajan
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Matunga, Mumbai 400019, India.
| |
Collapse
|
16
|
Bandekar M, Maurya DK, Sharma D, Checker R, Gota V, Mishra N, Sandur SK. Xenogeneic transplantation of human WJ-MSCs rescues mice from acute radiation syndrome via Nrf-2-dependent regeneration of damaged tissues. Am J Transplant 2020; 20:2044-2057. [PMID: 32040239 DOI: 10.1111/ajt.15819] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 01/25/2023]
Abstract
There is an unmet medical need for radiation countermeasures that can be deployed for treatment of exposed individuals during ionizing radiation (IR) accidents or terrorism. Wharton's jelly mesenchymal stem cells (WJ-MSCs) from human umbilical cord have been shown to avoid allorecognition and induce a tissue-regenerating microenvironment, which makes them an attractive candidate for mitigating IR injury. We found that WJ-MSCs protected mice from a lethal dose of IR even when transplanted up to 24 hours after irradiation, and a combination of WJ-MSCs and antibiotic (tetracycline) could further expand the window of protection offered by WJ-MSCs. This combinatorial approach mitigated IR-induced damage to the hematopoietic and gastrointestinal system. WJ-MSCs increased the serum concentration of the cytoprotective cytokines granulocyte colony-stimulating factor (G-CSF) and IL-6 in mice. Knockdown of G-CSF and IL-6 in WJ-MSCs before injection to lethally irradiated mice or transplantation of WJ-MSCs to lethally irradiated Nrf-2 knockout mice significantly nullified the therapeutic protective efficacy. Hence, WJ-MSCs could be a potential cell-based therapy for individuals accidentally exposed to radiation.
Collapse
Affiliation(s)
- Mayuri Bandekar
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,University of Mumbai, Kalina, Mumbai, India
| | - Dharmendra K Maurya
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Deepak Sharma
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Rahul Checker
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Vikram Gota
- Clinical Pharmacology, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | | | - Santosh K Sandur
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| |
Collapse
|
17
|
Pérez-Gálvez A, Viera I, Roca M. Carotenoids and Chlorophylls as Antioxidants. Antioxidants (Basel) 2020; 9:E505. [PMID: 32526968 PMCID: PMC7346216 DOI: 10.3390/antiox9060505] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/29/2020] [Accepted: 06/06/2020] [Indexed: 01/06/2023] Open
Abstract
Chlorophylls and carotenoids are natural pigments that are present in our daily diet, especially with the increasing tendency towards more natural and healthy behaviors among consumers. As disturbed antioxidant homeostasis capacities seem to be implicated in the progress of different pathologies, the antioxidant properties of both groups of lipophilic compounds have been studied. The objective of this review was to analyze the state-of-the-art advances in this field. We conducted a systematic bibliographic search (Web of Science™ and Scopus®), followed by a comprehensive and critical description of the results, with special emphasis on highly cited and more recently published research. In addition to an evaluative description of the methodologies, this review discussed different approaches used to obtain a physiological perspective, from in vitro studies to in vivo assays using oxidative biomarkers. From a chemical viewpoint, many studies have demonstrated how a pigment's structure influences its antioxidant response and the underlying mechanisms. The major outcome is that this knowledge is essential for interpreting new data in a metabolic networks context in the search for more direct applications to health. A promising era is coming where the term "antioxidant" is understood in terms of its broadest significance.
Collapse
Affiliation(s)
| | | | - María Roca
- Food Phytochemistry Department, Instituto de la Grasa (CSIC), University Campus, Building 46, 41013 Sevilla, Spain; (A.P.-G.); (I.V.)
| |
Collapse
|
18
|
Merino JJ, Cabaña-Muñoz ME, Pelaz MJ. The Bluegreen Algae (AFA) Consumption over 48 Hours Increases the Total Number of Peripheral CD34+ Cells in Healthy Patients: Effect of Short-Term and Long-Term Nutritional Supplementation (Curcumin/AFA) on CD34+ Levels (Blood). J Pers Med 2020; 10:E49. [PMID: 32521810 PMCID: PMC7354690 DOI: 10.3390/jpm10020049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/31/2020] [Accepted: 06/02/2020] [Indexed: 12/03/2022] Open
Abstract
Several active principles from plants could trigger the release of stem cells from the bone marrow. Stem cell mobilizers have shown side effects in patients. Thus, the purpose of this paper is to find the natural products from plants (curcuminoids, glycosinolate of sulforaphane, AFA bluegreen algae), which could be potential stem mobilizes without adverse side effects. The antioxidant curcumin [1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-2,5-dione], glycosinolate of sulforaphane (broccoli) or AFA (Aphanizomenon flos) extract promote beneficial effects in patients. The number of circulating stem cells were monitored by HSC marker-CD34 by flow cytometry in peripheral blood from healthy subjects. CD34 is a hematological stem cells (HSC) marker. A double-blind study was conducted in 22 healthy subjects. We have evaluated whether short-term AFA-Aphanizomenon flos aquae-algae or curcuminoids consumption (powder or liquid formulation) over 48 consecutive hours could increase the total number of peripheral CD34+ blood cells (n = 22, n = 5 subjects/group). The total number of circulating CD34+ cells were quantified after short-term and long-term nutritional supplementation; their levels were compared with their own basal levels (n = 5/group, controls: before taking any supplement) or placebo-treated patients (n = 7); their average age was 54 years old. We also evaluated whether long-term nutritional supplementation with several nutraceuticals could enhance HSC mobilization by increasing the total number of peripheral CD-34+ cell after seven or 38 consecutive days of administration (n = 5, with seven placebo-treated patients). The long-term administration take place with these doses/day [curcuminoids: 2000 mg/day, equivalent to 120 mg of curcuminoids/day), glycosinolate of sulforaphane (66 mg/day), plus AFA Algae bluegreen extract (400 mg/day)]. On the last day (10 A.M.) of treatment, blood samples were collected six hours after taking these supplements; the average age was 54 years old. Notably, the blue green AFA algae extract consumption over 48 h enhances HSC mobilization by increasing the total number of peripheral CD34+ cells. The long-term administration with curcuminoids, glycosinolate of sulforaphane, and AFA bluegreen algae extract also increased the total number of CD34-HSC cells after seven or 38 days of consecutive of administration in healthy subjects.
Collapse
Affiliation(s)
- José Joaquín Merino
- Dpto. Farmacologia, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | | | | |
Collapse
|
19
|
Liu Z, Xia S, Wang X, Lan Q, Li P, Xu W, Wang Q, Lu L, Jiang S. Sodium Copper Chlorophyllin Is Highly Effective against Enterovirus (EV) A71 Infection by Blocking Its Entry into the Host Cell. ACS Infect Dis 2020; 6:882-890. [PMID: 32233455 DOI: 10.1021/acsinfecdis.0c00096] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human enteroviruses (HEVs) pose an ongoing threat to global public health. Particularly, enterovirus-A71 (EV-A71), the main pathogen causing hand-foot-and-mouth disease (HFMD), has caused ongoing outbreaks globally in recent years associated with severe neurological manifestations and several deaths. Currently, no effective antivirals are available for the prevention or treatment of EV-A71 infection. In this study, we found that sodium copper chlorophyllin (CHL), a health food additive and an over-the-counter anticancer medicine or treatment to reduce the odor of urine or feces, exhibited potent inhibitory activity against infection by divergent EV-A71 and coxsackievirus-A16 (CV-A16) isolates at a low micromolar concentration with excellent safety. The antiviral activity of each was confirmed by colorimetric viral infection and qRT-PCR assays. A series of mechanistic studies showed that CHL did not target the host cell but blocked the entry of EV-A71 and CV-A16 into the host cell at the postattachment stage. In the mouse model, CHL could significantly reduce the viral titer in the lungs and muscles. Since CHL has been used in clinics for many years with excellent safety, it has the potential to be further developed into a prophylactic or therapeutic to prevent or treat HFMD caused by EV-A71 or CV-A16 infection.
Collapse
Affiliation(s)
- Zezhong Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shuai Xia
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xinling Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qiaoshuai Lan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Peiyu Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wei Xu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qian Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York 10065, United States
| |
Collapse
|
20
|
Reactive Oxygen Species and Nrf2: Functional and Transcriptional Regulators of Hematopoiesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5153268. [PMID: 31827678 PMCID: PMC6885799 DOI: 10.1155/2019/5153268] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/09/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023]
Abstract
Hematopoietic stem cells (HSCs) are characterized by self-renewal and multilineage differentiation potentials. Although they play a central role in hematopoietic homeostasis and bone marrow (BM) transplantation, they are affected by multiple environmental factors in the BM. Here, we review the effects of reactive oxygen species (ROS) and Nrf2 on HSC function and BM transplantation. HSCs reside in the hypoxic microenvironment of BM, and ROS play an important role in HSPC regulation. Recently, an extraphysiologic oxygen shock/stress phenomenon was identified in human cord blood HSCs collected under ambient air conditions. Moreover, Nrf2 has been recently recognized as a master transcriptional factor that regulates multiple antioxidant enzymes. Since several years, the role of Nrf2 in hematopoiesis has been extensively studied, which has functional similarities of cellular oxygen sensor hypoxia-inducible factor-1 as transcriptional factors. Increasing evidence has revealed that abnormally elevated ROS production due to factors such as genetic defects, aging, and ionizing radiation unexceptionally resulted in lethal impairment of HSC function and hematopoiesis. Both experimental and clinical studies have identified elevated ROS levels as a major culprit of ineffective BM transplantation. Lastly, we discuss the possibility of using small molecule antioxidants, such as N-acetyl cysteine, resveratrol, and curcumin, to augment HSC function and improve the therapeutic efficacy of BM transplantation. Further research on the function of ROS levels and improving the efficacy of BM transplantation may have a great potential for broad clinical applications of HSCs.
Collapse
|
21
|
Checker R, Pal D, Patwardhan RS, Basu B, Sharma D, Sandur SK. Modulation of Caspase-3 activity using a redox active vitamin K3 analogue, plumbagin, as a novel strategy for radioprotection. Free Radic Biol Med 2019; 143:560-572. [PMID: 31493505 DOI: 10.1016/j.freeradbiomed.2019.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/30/2019] [Accepted: 09/01/2019] [Indexed: 12/15/2022]
Abstract
Radiation induced damage to normal cells is a major shortcoming of conventional radiotherapy, which necessitates the development of novel radio-protective drugs. An ideal radio-modulator would protect normal cells while having cytotoxic effects on cancer cells. Plumbagin is a potent anti-tumour agent and has been shown to sensitize tumour cells to radiation-induced damage. In the present study, we have evaluated the radio-protective potential of plumbagin and found that it protected normal lymphocytes against radiation-induced apoptosis, but did not protect cancer cells against radiation. Plumbagin offered radioprotection even when it was added to cells after irradiation. The ability of only thiol based antioxidants to abrogate the radio-protective effects of plumbagin suggested a pivotal role of thiol groups in the radio-protective activity of plumbagin. Further, protein interaction network (PIN) analysis was used to predict the molecular targets of plumbagin. Based on the inputs from plumbagin's PIN and in light of its well-documented ability to modulate thiol groups, we proposed that plumbagin may act via modulation of caspase enzyme which harbours a critical catalytic cysteine. Indeed, plumbagin suppressed radiation-induced increase in homogenous caspase and caspase-3 activity in lymphocytes. Plumbagin also inhibited the activity of recombinant caspase-3 and mass spectrometric analysis revealed that plumbagin covalently interacts with caspase-3. Further, the in vivo radioprotective efficacy of plumbagin (single dose of 2mg/kg body weight) was demonstrated by its ability to rescue mice against radiation (7.5 Gy; Whole Body Irradiation) induced mortality. These results indicate that plumbagin prevents radiation induced apoptosis specifically in normal cells by inhibition of caspase-3 activity.
Collapse
Affiliation(s)
- Rahul Checker
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India
| | - Debojyoti Pal
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085, India
| | - Bhakti Basu
- Molecular Biology Division, Bio-science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India.
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India.
| |
Collapse
|
22
|
Efficacy of SXN in the Treatment of Iron Deficiency Anemia: A Phase IV Clinical Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:8796234. [PMID: 30941201 PMCID: PMC6421013 DOI: 10.1155/2019/8796234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/22/2018] [Accepted: 02/07/2019] [Indexed: 11/17/2022]
Abstract
Shengxuening (SXN) tablet is extracted from the excrement of the silkworm and has effects on hematopoiesis. The main components of SXN are chlorophyll derivatives and sodium iron chlorophyllin (SIC). The present study aims to investigate the efficiency and safety of SXN on iron deficiency anemia. This phase IV, multicenter, open-label, randomized clinical trial was conducted in 31 hospitals in China from June 2001 to April 2002. Adults and children were randomly divided into low-dose (L-SXN), medium-dose (M-SXN), and high-dose (H-SXN) groups, respectively. The course of treatment was 1 month. Peripheral hemogram levels and iron status were examined before and after treatment. Adults in all three dose groups demonstrated a significant increase in hemoglobin (HGB) concentration. Children who received SXN treatment in medium and high doses also demonstrated increased HGB concentration. Reticulocyte counts increased at the end of treatment in the M-SXN and H-SXN adult groups and in the M-SXN child group. For both children and adults, SXN in the three dose groups was found to significantly elevate red blood cell level, mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular hemoglobin concentration. The total effective rate in the SXN-treated group reached 84.8%. The incidence of adverse events was 4.07%. The most common side effects were nausea (2.83%), diarrhea (0.74%), and rash (0.25%). SXN was proved to be efficient and safe for adults and children with iron deficiency anemia.
Collapse
|
23
|
The protective effects of 1,2-propanediol against radiation-induced hematopoietic injury in mice. Biomed Pharmacother 2019; 114:108806. [PMID: 30928804 DOI: 10.1016/j.biopha.2019.108806] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 01/17/2023] Open
Abstract
Agents that provide protection against irradiation-induced hematopoietic injury are urgently needed for radiotherapy. We examined the effects of the small molecule, 1,2-propanediol (PPD), on total body irradiation (TBI)-induced hematopoietic injury in C57BL/6 mice. PPD administration 1 h before TBI significantly increased hematopoietic parameters such as white blood cell, platelet, red blood cell, and lymphocyte counts in vivo and enhanced the survival of mice exposed to TBI (7.0 and 7.5 Gy). PPD administration 1 h before TBI improved bone marrow (BM) and spleen recovery after TBI, with increases in both BM cellularity and spleen index. The number of colony-forming-units in bone marrow mononuclear cells (BMNCs) in vitro also increased significantly. PPD pretreatment increased the numbers of hematopoietic stem cells and hematopoietic progenitor cells in BM. Importantly, PPD also maintained endogenous antioxidant status by decreasing levels of malondialdehyde and increasing the expression of reduced glutathione, superoxide dismutase and catalase in the serum of irradiated mice. PPD alleviated the levels of apoptosis in HSCs induced by TBI, thus increasing the proportion of dividing BMNCs. These results suggest that PPD protects against TBI-induced hematopoietic injury through the increased activities of antioxidant enzymes and the inhibition of apoptosis in HSCs. PPD increased the serum levels of granulocyte-colony stimulating factor and interleukin-6 irrespective of TBI. In conclusion, these data suggest that PPD acts as a radioprotector against radiation-induced hematopoietic injury.
Collapse
|
24
|
Gerić M, Gajski G, Mihaljević B, Miljanić S, Domijan AM, Garaj-Vrhovac V. Radioprotective properties of food colorant sodium copper chlorophyllin on human peripheral blood cells in vitro. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 845:403027. [PMID: 31561900 DOI: 10.1016/j.mrgentox.2019.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/31/2018] [Accepted: 02/25/2019] [Indexed: 12/12/2022]
Abstract
Sodium copper chlorophyllin (CHL) is a food colorant that exhibits many beneficial properties, including potential for use in radiotherapy. Nevertheless, genotoxicity studies investigating radioprotective properties against γ-radiation on human cells are rather scarce. The aim of this study was to assess the cytotoxicity, genotoxicity and induction of malondialdehyde formation on CHL pre-treated whole blood cells after an absorbed dose of 5 Gy γ-radiation. Irradiated whole blood cells pre-treated with 100, 500, and 1000 μg/mL CHL showed less DNA-strand breaks (10.92 ± 0.74%, 10.69 ± 0.68%, and 8.81 ± 0.69%, respectively) than untreated irradiated cells (12.58 ± 0.88%). At the same time, the level of malondialdehyde was lower in CHL pre-treated samples with 100, 500, and 1000 μg/mL CHL (14.11 ± 0.43, 16.35 ± 2.82, and 13.08 ± 1.03 μmol/L, respectively) compared to untreated irradiated samples (24.11 ± 0.25 μmol/L). Regarding cytotoxicity, no changes were observed in the samples tested. Another important finding is that CHL had no cyto/genotoxic properties toward human blood cells. Taken together, since CHL had no cyto/genotoxic effects and showed good radioprotective properties in human blood cells, further studies should be conducted in order to find its possible application in radiotherapy.
Collapse
Affiliation(s)
- Marko Gerić
- Institute for Medical Research and Occupational Health, Ksaverska cesta 2, Zagreb, Croatia.
| | - Goran Gajski
- Institute for Medical Research and Occupational Health, Ksaverska cesta 2, Zagreb, Croatia
| | | | - Saveta Miljanić
- Ruđer Bošković Institute, Bijenička cesta 54, Zagreb, Croatia
| | - Ana-Marija Domijan
- Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, Zagreb, Croatia
| | - Vera Garaj-Vrhovac
- Institute for Medical Research and Occupational Health, Ksaverska cesta 2, Zagreb, Croatia.
| |
Collapse
|
25
|
Cheng Y, Dong Y, Hou Q, Wu J, Zhang W, Tian H, Li D. The protective effects of XH-105 against radiation-induced intestinal injury. J Cell Mol Med 2019; 23:2238-2247. [PMID: 30663222 PMCID: PMC6378229 DOI: 10.1111/jcmm.14159] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/17/2018] [Accepted: 12/24/2018] [Indexed: 12/26/2022] Open
Abstract
Radiation-induced intestinal injury is one of the major side effects in patients receiving radiation therapy. There is no specific treatment for radiation enteritis in the clinic. We designed and synthesized a new compound named XH-105, which is expected to cleave into polyphenol and aminothiol in vivo to mitigate radiation injury. In the following study, we describe the beneficial effects of XH-105 against radiation-induced intestinal injury. C57BL/6J mice were treated by gavage with XH-105 1 hour before total body irradiation (TBI), and the survival rate was monitored. Histological changes were examined, and survival of Lgr5+ intestinal stem cells Ki67+ cells, villi+ enterocytes and lysozymes was determined by immunohistochemistry. DNA damage and cellular apoptosis in intestinal tissue were also evaluated. Compared to vehicle-treated mice after TBI, XH-105 treatment significantly enhanced the survival rate, attenuated structural damage of the small intestine, decreased the apoptotic rate, reduced DNA damage, maintained cell regeneration and promoted crypt proliferation and differentiation. XH-105 also reduced the expression of Bax and p53 in the small intestine. These data suggest that XH-105 is beneficial for the protection of radiation-induced intestinal injury by inhibiting the p53-dependent apoptosis signalling pathway.
Collapse
Affiliation(s)
- Ying Cheng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.,Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Qinlian Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Jing Wu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Wei Zhang
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| |
Collapse
|
26
|
Pinzur L, Akyuez L, Levdansky L, Blumenfeld M, Volinsky E, Aberman Z, Reinke P, Ofir R, Volk HD, Gorodetsky R. Rescue from lethal acute radiation syndrome (ARS) with severe weight loss by secretome of intramuscularly injected human placental stromal cells. J Cachexia Sarcopenia Muscle 2018; 9:1079-1092. [PMID: 30334381 PMCID: PMC6240751 DOI: 10.1002/jcsm.12342] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 07/09/2018] [Accepted: 07/17/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Most current cell-based regenerative therapies are based on the indirect induction of the affected tissues repair. Xenogeneic cell-based treatment with expanded human placenta stromal cells, predominantly from fetal origin (PLX-RAD cells), were shown to mitigate significantly acute radiation syndrome (ARS) following high dose irradiation in mice, with expedited regain of weight loss and haematopoietic function. The current mechanistic study explores the indirect effect of the secretome of PLX-RAD cells in the rescue of the irradiated mice. METHODS The mitigation of the ARS was investigated following two intramuscularly (IM) injected 2 × 106 PLX-RAD cells, 1 and 5 days following 7.7 Gy irradiation. The mice survival rate and their blood or bone marrow (BM) cell counts were followed up and correlated with multiplex immunoassay of a panel of related human proteins of PLX-RAD derived secretome, as well as endogenous secretion of related mouse proteins. PLX-RAD secretome was also tested in vitro for its effect on the induction of the migration of BM progenitors. RESULTS A 7.7 Gy whole body mice irradiation resulted in ~25% survival by 21 days. Treatment with two IM injections of 2 × 106 PLX-RAD cells on days 1 and 5 after irradiation mitigated highly significantly the subsequent lethal ARS, with survival rate increase to nearly 100% and fast regain of the initial weight loss (P < 0,0001). This was associated with a significant faster haematopoiesis recovery from day 9 onwards (P < 0.01). Nine out of the 65 human proteins tested were highly significantly elevated in the mouse circulation, peaking on days 6-9 after irradiation, relative to negligible levels in non-irradiated PLX-RAD injected mice (P < 0.01). The highly elevated proteins included human G-CSF, GRO, MCP-1, IL-6 and lL-8, reaching >500 pg/mL, while MCP-3, ENA, Eotaxin and fractalkine levels ranged between ~60-160pg/mL. The detected radiation-induced PLX-RAD secretome correlated well with the timing of the fast haematopoiesis regeneration. The radiation-induced PLX-RAD secretome seemed to reinforce the delayed high levels secretion of related mouse endogenous cytokines, including GCSF, KC, MCP-1 and IL-6. Additional supportive in vitro studies also confirmed the ability of cultured PLX-RAD secretome to induce accelerated migration of BM progenitors. CONCLUSIONS A well-regulated and orchestrated secretion of major pro-regenerative BM supporting secretome in high dose irradiated mice, treated with xenogeneic IM injected PLX-RAD cells, can explain the observed mitigation of ARS. This seemed to coincide with faster haematopoiesis regeneration, regain of severe weight loss and the increased survival rate. The ARS-related stress signals activating the IM injected PLX-RAD cells for the remote secretion of the relevant human proteins deserve further investigation.
Collapse
Affiliation(s)
- Lena Pinzur
- Pluristem LTD, Haifa, Israel.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT) and Institute of Medical Immunology and Department of Nephrology and Intensive Care, Charité-University Medicine Berlin, Berlin, Germany
| | - Levent Akyuez
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT) and Institute of Medical Immunology and Department of Nephrology and Intensive Care, Charité-University Medicine Berlin, Berlin, Germany
| | - Lilia Levdansky
- Laboratory of Biotechnology and Radiobiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Evgenia Volinsky
- Laboratory of Biotechnology and Radiobiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT) and Institute of Medical Immunology and Department of Nephrology and Intensive Care, Charité-University Medicine Berlin, Berlin, Germany
| | | | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT) and Institute of Medical Immunology and Department of Nephrology and Intensive Care, Charité-University Medicine Berlin, Berlin, Germany
| | - Raphael Gorodetsky
- Laboratory of Biotechnology and Radiobiology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
27
|
The Protective Effect of New Compound XH-103 on Radiation-Induced GI Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3920147. [PMID: 30116481 PMCID: PMC6079366 DOI: 10.1155/2018/3920147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/01/2018] [Accepted: 05/21/2018] [Indexed: 12/13/2022]
Abstract
Background Radiation-induced intestinal injury is one of the side effects in patients receiving radiotherapy. The aim of the present study was to investigate the protective effect of XH-103 on radiation-induced small intestinal injury and to explore its mechanism. Methods C57BL/6N mice were irradiated and treated with XH-103. Firstly, the survival rate of mice exposed to 9.0 Gy and 11.0 Gy total body irradiation (TBI) was examined. Subsequently, at 3.5 d after IR, the small intestinal morphological changes were examined by HE. The numbers of crypt cells, the villus height, the expression of Ki67 and Lgr5, and the apoptotic cells in the intestinal crypts were examined by immunohistochemistry. Furthermore, the expression of p53 and Bax was analyzed by WB. Results Compared to the irradiation group, XH-103 improved the mice survival rate, protected the intestinal morphology of mice, decreased the apoptotic rate of intestinal crypt cells, maintained cell regeneration, and promoted crypt proliferation and differentiation. XH-103 also reduced the expression of p53 and Bax in the small intestine compared to the IR group. Conclusion These data demonstrate that XH-103 can prevent radiation-induced intestinal injury, which is beneficial for the protection of radiation injuries.
Collapse
|
28
|
Long W, Zhang G, Dong Y, Li D. Dark tea extract mitigates hematopoietic radiation injury with antioxidative activity. JOURNAL OF RADIATION RESEARCH 2018; 59:387-394. [PMID: 29325132 PMCID: PMC6054171 DOI: 10.1093/jrr/rrx072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Indexed: 05/09/2023]
Abstract
The hematopoietic system is widely studied in radiation research. Tea has been proved to have antioxidative activity. In the present study, we describe the protective effects of dark tea extract (DTE) on radiation-induced hematopoietic injury. DTE administration significantly enhanced the survival rate of mice after 7.0 and 7.5 Gy total body irradiation (TBI). The results showed that DTE not only markedly increased the numbers and cloning potential of hematopoietic cells, but also decreased DNA damages after mice were exposed to 6.0 Gy total body irradiation (TBI). In addition, DTE also decreased the levels of reactive oxygen species (ROS) in hematopoietic cells by inhibiting NOX4 expression and increasing the dismutase, catalase and glutathione peroxidase in livers. These data demonstrate that DTE can prevent radiation-induced hematopoietic syndromes, which is beneficial for protection from radiation injuries.
Collapse
Affiliation(s)
- Wei Long
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, No. 238, Baidi Road, Nankai District, Tianjin, China
| | - Guanghui Zhang
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, No. 238, Baidi Road, Nankai District, Tianjin, China
| | - Yinping Dong
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, No. 238, Baidi Road, Nankai District, Tianjin, China
| | - Deguan Li
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, No. 238, Baidi Road, Nankai District, Tianjin, China
- Corresponding author. Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical College, No. 238, Baidi Road, Nankai District, Tianjin 300192, China. Tel: +86-022-85682340; Fax: +86-022-85683033;
| |
Collapse
|
29
|
Zhai X, Yang X, Zou P, Shao Y, Yuan S, Abd El-Aty AM, Wang J. Protective Effect of Chitosan Oligosaccharides Against Cyclophosphamide-Induced Immunosuppression and Irradiation Injury in Mice. J Food Sci 2018; 83:535-542. [PMID: 29350748 DOI: 10.1111/1750-3841.14048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/05/2017] [Accepted: 12/16/2017] [Indexed: 12/22/2022]
Abstract
Chitosan oligosaccharides (COS), hydrolyzed products of chitosan, was found to display various biological activities. Herein, we assessed the immunostimulatory activity of COS both in in vitro and in vivo studies. In vitro cytotoxicity studies to murine macrophage RAW264.7 revealed that COS is safe even at the maximum tested concentration of 1000 μg/mL. It also stimulates the production of nitric oxide (NO) and tumor necrosis factor (TNF-α) and enhances the phagocytosis in COS-stimulated RAW264.7. We have shown that the COS could significantly (P < 0.05) restore the reduced immune organs indices, phagocytic index, lymphocyte proliferation, natural killer cell activity, and antioxidant enzyme activities in a cyclophosphamide-induced immunosuppressed mice model. COS can also improve the survival rate in irradiation injury mice and significantly (P < 0.05) increased the spleen indices and up-regulates the CD4+/CD8+ ratio in splenocytes. In sum, the aforementioned results suggest that COS might has the potential to be used as an immunostimulatory agent in patients with immune dysfunctions or be a model for functional food development. PRACTICAL APPLICATION COS might has the potential to be used as an immunostimulatory agent in patients with immune dysfunctions or be a model for functional food development.
Collapse
Affiliation(s)
- Xingchen Zhai
- Dept. of Food Sciences and Engineering, School of Chemistry and Chemical Engineering, Harbin Inst. of Technology, 150090 Harbin, PR China.,Key Lab. of Agro-Product Quality and Safety, Inst. of Quality Standard and Testing Technology for Agro-Product, Chinese Acad. of Agricultural Sciences, 100081 Beijing, PR China.,the Dept. of Pharmacology and Toxicology, Beijing Inst. of Radiation Medicine, 100081 Beijing, PR China
| | - Xin Yang
- Dept. of Food Sciences and Engineering, School of Chemistry and Chemical Engineering, Harbin Inst. of Technology, 150090 Harbin, PR China
| | - Pan Zou
- Dept. of Food Sciences and Engineering, School of Chemistry and Chemical Engineering, Harbin Inst. of Technology, 150090 Harbin, PR China.,Key Lab. of Agro-Product Quality and Safety, Inst. of Quality Standard and Testing Technology for Agro-Product, Chinese Acad. of Agricultural Sciences, 100081 Beijing, PR China
| | - Yong Shao
- Key Lab. of Agro-Product Quality and Safety, Inst. of Quality Standard and Testing Technology for Agro-Product, Chinese Acad. of Agricultural Sciences, 100081 Beijing, PR China
| | - Shoujun Yuan
- the Dept. of Pharmacology and Toxicology, Beijing Inst. of Radiation Medicine, 100081 Beijing, PR China
| | - A M Abd El-Aty
- Dept. of Pharmacology, Faculty of Veterinary Medicine, Cairo Univ., 12211 Giza, Egypt
| | - Jing Wang
- Dept. of Food Sciences and Engineering, School of Chemistry and Chemical Engineering, Harbin Inst. of Technology, 150090 Harbin, PR China.,Key Lab. of Agro-Product Quality and Safety, Inst. of Quality Standard and Testing Technology for Agro-Product, Chinese Acad. of Agricultural Sciences, 100081 Beijing, PR China
| |
Collapse
|
30
|
Han X, Zhang J, Xue X, Zhao Y, Lu L, Cui M, Miao W, Fan S. Theaflavin ameliorates ionizing radiation-induced hematopoietic injury via the NRF2 pathway. Free Radic Biol Med 2017; 113:59-70. [PMID: 28939421 DOI: 10.1016/j.freeradbiomed.2017.09.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/08/2017] [Accepted: 09/17/2017] [Indexed: 12/24/2022]
Abstract
It has been well established that reactive oxygen species (ROS) play a critical role in ionizing radiation (IR)-induced hematopoietic injury. Theaflavin (TF), a polyphenolic compound from black tea, has been implicated in the regulation of endogenous cellular antioxidant systems. However, it remains unclear whether TF could ameliorate IR-induced hematopoietic injury, particularly the hematopoietic stem cell (HSC) injury. In this study, we explored the potential role of TF in IR-induced HSC injury and the underlying mechanism in a total body irradiation (TBI) mouse model. Our results showed that TF improved survival of irradiated wild-type mice and ameliorated TBI-induced hematopoietic injury by attenuating myelosuppression and myeloid skewing, increasing HSC frequency, and promoting reconstitution of irradiated HSCs. Furthermore, TF inhibited TBI-induced HSC senescence. These effects of TF were associated with a decline in ROS levels and DNA damage in irradiated HSCs. TF reduced oxidative stress mainly by up-regulating nuclear factor erythroid 2-related factor 2 (NRF2) and its downstream targets in irradiated Lineage-c-kit+ positive cells. However, TF failed to improve the survival, to increase HSC frequency and to reduce ROS levels of HSCs in irradiated Nrf2-/- mice. These findings suggest that TF ameliorates IR-induced HSC injury via the NRF2 pathway. Therefore, TF has the potential to be used as a radioprotective agent to ameliorate IR-induced hematopoietic injury.
Collapse
Affiliation(s)
- Xiaodan Han
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China.
| | - Junling Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China.
| | - Xiaolei Xue
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Lu Lu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Weimin Miao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300041,China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China.
| |
Collapse
|
31
|
Verma P, Kunwar A, Indira Priyadarsini K. Effect of Low-Dose Selenium Supplementation on the Genotoxicity, Tissue Injury and Survival of Mice Exposed to Acute Whole-Body Irradiation. Biol Trace Elem Res 2017; 179:130-139. [PMID: 28190183 DOI: 10.1007/s12011-017-0955-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/25/2017] [Indexed: 01/03/2023]
Abstract
The aim of the present study is to evaluate the radioprotective effect of low-dose selenium supplementation (multiple administrations) on radiation toxicities and mortality induced by lethal dose of whole-body irradiation (WBI). For this, BALB/c mice received sodium selenite (4 μg/kg body wt) intraperitoneally for five consecutive days and subjected to WBI at an absorbed dose of 8 Gy (60Co, 1 Gy/min). Administration of sodium selenite was continued even during the post irradiation days three times a week till the end of the experiment. The radioprotective effect was evaluated in terms of the improvement in 30 days post irradiation survival, protection from DNA damage, and biochemical and histological changes in radiosensitive organs. The results indicated that low-dose sodium selenite administration did not protect the mice from radiation-induced hematopoietic and gastrointestinal injuries and subsequent mortality. However, it significantly prevented the radiation-induced genotoxicity or DNA damage in peripheral leukocytes. Further sodium selenite administration modulated the messenger RNA (mRNA) expression of GPx1, GPx2, and GPx4 in the spleen and intestine differentially and led to a significant increase in GPx activity (∼1.5 to 2-folds) in these organs. In line with this observation, sodium selenite administration reduced the level of lipid peroxidation in the intestine. In conclusion, our study shows that low-dose sodium selenite supplementation can be an effective strategy to prevent WBI-induced genotoxicity but may not have an advantage against mortality sustained during nuclear emergencies.
Collapse
Affiliation(s)
- Prachi Verma
- Radiation and Photochemistry Division, Bhabha Atomic Research Centre, Mumbai, 400085, India
- Homi Bhabha National Institute, Mumbai, 400094, India
| | - Amit Kunwar
- Radiation and Photochemistry Division, Bhabha Atomic Research Centre, Mumbai, 400085, India.
- Homi Bhabha National Institute, Mumbai, 400094, India.
| | - K Indira Priyadarsini
- Homi Bhabha National Institute, Mumbai, 400094, India
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai, India
| |
Collapse
|
32
|
Calf Spleen Extractive Injection protects mice against cyclophosphamide-induced hematopoietic injury through G-CSF-mediated JAK2/STAT3 signaling. Sci Rep 2017; 7:8402. [PMID: 28827748 PMCID: PMC5566473 DOI: 10.1038/s41598-017-08970-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/20/2017] [Indexed: 12/22/2022] Open
Abstract
Calf Spleen Extractive Injection (CSEI), extracted from the spleen of healthy cows (within 24 hours of birth), is a small-peptide-enriched extraction and often used as an ancillary agent in cancer therapy. This study evaluated the hematopoietic function of CSEI and its underlying mechanisms, principally in CHRF, K562 cells, BMNCs and a mouse model of cyclophosphamide (CTX)-induced hematopoietic suppression. CSEI promoted the proliferation and differentiation of CHRF and K562 cells, activated hematopoietic- and proliferation-related factors RSK1p90, ELK1 and c-Myc, and facilitated the expression of differentiation- and maturation-related transcription factors GATA-1, GATA-2. In the mice with hematopoietic suppression, 3 weeks of CSEI administration enhanced the bodyweights and thymus indices, suppressed the spleen indices and strongly elevated the production of HSPCs, neutrophils and B cells in bone marrow, ameliorated bone marrow cellularity, and regulated the ratio of peripheral blood cells. Proteome profiling combined with ELISA revealed that CSEI regulated the levels of cytokines, especially G-CSF and its related factors, in the spleen and plasma. Additional data revealed that CSEI promoted phosphorylation of STAT3, which was stimulated by G-CSF in both mice spleen and cultured BMNCs. Taken together, CSEI has the potential to improve hematopoietic function via the G-CSF-mediated JAK2/STAT3 signaling pathway.
Collapse
|
33
|
Bagwe SM, Kale PP, Bhatt LK, Prabhavalkar KS. Herbal approach in the treatment of pancytopenia. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2017; 14:/j/jcim.ahead-of-print/jcim-2016-0053/jcim-2016-0053.xml. [PMID: 28195548 DOI: 10.1515/jcim-2016-0053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 10/18/2016] [Indexed: 11/15/2022]
Abstract
Pancytopenia is a health condition in which there is a reduction in the amount of leucocytes, erythrocytes and thrombocytes. If more than one of the blood cells is low then the condition is called as bicytopenia. The pancytopenic condition is observed in treatment of diseased conditions like thalassemia and hepatitis C. Iatrogenically pancytopenia is caused by some antibiotics and anti-HCV drugs. Medical conditions like aplastic anaemia, lymphoma, copper deficiency, and so forth can also cause pancytopenia. Pancytopenia can in turn decrease the immunity of the person and thereby can be fatal. Current therapies for pancytopenia include bone marrow stimulant drugs, blood transfusion and bone marrow transplant. The current therapies are very excruciating and have long-term side-effects. Therefore, treating these condition using herbal drugs is very important. Herbs like wheatgrass, papaya leaves and garlic are effective in treating single lineage cytopenias. The present review is focused on the potential effects of natural herbs for the treatment of pancytopenia.
Collapse
|
34
|
Yun KL, Wang ZY. Target/signalling pathways of natural plant-derived radioprotective agents from treatment to potential candidates: A reverse thought on anti-tumour drugs. Biomed Pharmacother 2017; 91:1122-1151. [DOI: 10.1016/j.biopha.2017.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/15/2017] [Accepted: 05/01/2017] [Indexed: 02/07/2023] Open
|
35
|
Newman NB, Moss RA, Maloney-Patel N, Donohue K, Brown TV, Nissenblatt MJ, Lu SE, Jabbour SK. Bone marrow tolerance during postoperative chemotherapy in colorectal carcinomas. J Gastrointest Oncol 2017; 8:547-555. [PMID: 28736641 DOI: 10.21037/jgo.2017.01.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND This study seeks to quantify and compare bone marrow tolerance during postoperative chemotherapy therapy between rectal cancer vs. colon cancer patients. During rectal cancer treatment, patients receive neoadjuvant chemoradiation (CRT) irradiation which can exacerbate the hematologic toxicity (HT) via incidental irradiation of the pelvic bone marrow (PBM) during myelosuppressive postoperative chemotherapy. In contrast, colon cancer patients receive the same postoperative myelosuppressive chemotherapy but do not routinely receive preoperative chemoradiation therapy. This comparison will help elucidate the lasting myelosuppressive effects of incidental pelvic bone marrow (PBM) irradiation on rectal cancer patients during neoadjuvant preoperative chemoradiation therapy. METHODS Rectal cancer patients treated with preoperative CRT followed by postoperative 5-Fluorouracil and oxaliplatin (OxF) chemotherapy (n=35) were compared to colon cancer patients who received only postoperative OxF chemotherapy (n=42). End points were ≥ grade 3 hematologic toxicity (HT3) or hematologic event (HE) defined as ≥ grade 2 HT and a dose reduction in OxF. Wilcoxon rank sum test tested continuous variables and Chi-squared test measured differences in categorical variables. HT3 and HE probability during postoperative chemotherapy was estimated with Kaplan-Meier curves and Cox regression analysis. RESULTS During OxF chemotherapy, 40.0% (n=14) of rectal cancer patients experienced HT3 compared to 26.1% (n=11) of colon cancer patients (P=0.4). HE was experienced by 48% (n=17) of rectal cancer patients compared to 36% (n=15) of colon cancer patients (P=0.36). Rectal cancer patients were likelier to experience HT3 on multivariable cox regression analysis, controlling for several clinical covariates, with a hazard ratio (HR) of 2.49, [(95% CI: 1.02-6.02), P=0.045] than colon cancer patients. While rectal cancer patients were more likely to experience HE than colon cancer patients on multivariable Cox regression analysis with a HR of 1.8 (95% CI: 0.95-3.75), this only trended in statistical significance, P=0.07. CONCLUSIONS Rectal cancer patients are more likely than colon cancer patients to experience hematologic toxicities impacting the tolerance of standard of care chemotherapeutics during adjuvant therapy. Focused PBM sparing during radiation therapy for rectal cancer patients may improve tolerance of myelosuppressive chemotherapeutic agents delivered in the postoperative setting.
Collapse
Affiliation(s)
- Neil B Newman
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Rebecca A Moss
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA.,Bristol-Myers Squibb, Lawrence, NJ, USA
| | - Nell Maloney-Patel
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Kristen Donohue
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Teresa V Brown
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | | | - Shou-En Lu
- Rutgers School of Public Health, Division of Biometrics, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
36
|
Song B, Zhang Y, Liu J, Feng X, Zhou T, Shao L. Is Neurotoxicity of Metallic Nanoparticles the Cascades of Oxidative Stress? NANOSCALE RESEARCH LETTERS 2016; 11:291. [PMID: 27295259 PMCID: PMC4905860 DOI: 10.1186/s11671-016-1508-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/30/2016] [Indexed: 05/31/2023]
Abstract
With the rapid development of nanotechnology, metallic (metal or metal oxide) nanoparticles (NPs) are widely used in many fields such as cosmetics, the food and building industries, and bio-medical instruments. Widespread applications of metallic NP-based products increase the health risk associated with human exposures. Studies revealed that the brain, a critical organ that consumes substantial amounts of oxygen, is a primary target of metallic NPs once they are absorbed into the body. Oxidative stress (OS), apoptosis, and the inflammatory response are believed to be the main mechanisms underlying the neurotoxicity of metallic NPs. Other studies have disclosed that antioxidant pretreatment or co-treatment can reverse the neurotoxicity of metallic NPs by decreasing the level of reactive oxygen species, up-regulating the activities of antioxidant enzymes, decreasing the proportion of apoptotic cells, and suppressing the inflammatory response. These findings suggest that the neurotoxicity of metallic NPs might involve a cascade of events following NP-induced OS. However, additional research is needed to determine whether NP-induced OS plays a central role in the neurotoxicity of metallic NPs, to develop a comprehensive understanding of the correlations among neurotoxic mechanisms and to improve the bio-safety of metallic NP-based products.
Collapse
Affiliation(s)
- Bin Song
- />Guizhou Provincial People’s Hospital, Guiyang, 550002 China
- />Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - YanLi Zhang
- />Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Jia Liu
- />Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - XiaoLi Feng
- />Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Ting Zhou
- />Guizhou Provincial People’s Hospital, Guiyang, 550002 China
| | - LongQuan Shao
- />Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| |
Collapse
|
37
|
Zhang J, Xue X, Han X, Yao C, Lu L, Li D, Hou Q, Miao W, Meng A, Fan S. Vam3 ameliorates total body irradiation-induced hematopoietic system injury partly by regulating the expression of Nrf2-targeted genes. Free Radic Biol Med 2016; 101:455-464. [PMID: 27989754 DOI: 10.1016/j.freeradbiomed.2016.10.501] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/13/2016] [Accepted: 10/26/2016] [Indexed: 12/26/2022]
Abstract
Vam3, a resveratrol dimer, has been implicated in the regulation of chronic obstructive pulmonary disease. However, the effect of Vam3 on total body irradiation (TBI)-induced hematopoietic progenitor cells (HPCs), and hematopoietic stem cells (HSCs) injury is unknown. In this study, we examined whether Vam3could ameliorate hematopoietic system injury induced by TBI. Our results indicated that Vam3 alleviated TBI-induced injury by improving the self-renewal and differentiation of HPCs, and HSCs. Vam3 decreased the intracellular ROS levels in irradiated mice HPCs/HSCs or c-kit positive cells and inhibited apoptosis and DNA damage in LSKs and HPCs after TBI. Vam3 up-regulated the expression of Nrf2 and related genes and proteins in irradiated c-kit positive cells in vitro. However, Vam3 did not increase the cell viability or the number of CFU-GM c-kit positive cells in irradiated Nrf2-/- mice but decreased the cellular ROS level. The above data showed that Vam3 ameliorates total body irradiation-induced hematopoietic system injury and that Nrf2 is essential in mediating Vam3's protective effect on the proliferation of c-kit positive cells after irradiation but not its ability to scavenge for free radicals.
Collapse
Affiliation(s)
- Junling Zhang
- Tianjin Key Lab of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Xiaolei Xue
- Tianjin Key Lab of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Xiaodan Han
- Tianjin Key Lab of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Chunsuo Yao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lu Lu
- Tianjin Key Lab of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Deguan Li
- Tianjin Key Lab of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Qi Hou
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Weimin Miao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300041, China
| | - Aimin Meng
- Institute of Laboratory Animal Science, Peking Union Medical College and Chinese Academy of Medical Science, Beijing 100021, China.
| | - Saijun Fan
- Tianjin Key Lab of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China.
| |
Collapse
|
38
|
Dihydroxyselenolane (DHS) supplementation improves survival following whole-body irradiation (WBI) by suppressing tissue-specific inflammatory responses. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 807:33-46. [DOI: 10.1016/j.mrgentox.2016.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/24/2016] [Accepted: 07/05/2016] [Indexed: 01/09/2023]
|
39
|
CHENG XIN, YU GUOJUN, HU JIANGPING, XU XUEFENG, LUO FANG, SHEN PING, ZHANG GUOSHENG, YANG NING. Clinical study of Shengxuening tablet combined with rHuEPO for the treatment of renal anemia of maintenance hemodialysis patients. Exp Ther Med 2016; 12:157-160. [PMID: 27347032 PMCID: PMC4906941 DOI: 10.3892/etm.2016.3307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/28/2016] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to investigate the clinical effects of Shengxuening tablet (silkworm excrement) combined with recombinant human erythropoietin (rHuEPO) for the treatment of renal anemia of maintenance hemodialysis (MHD) patients. Seventy-two MHD patients with renal anemia were included in the study and randomly divided into the control (n=34) and observation (n=38) groups. Patients in the control group were treated by hypodermic injection of 100-150 U/(kg·w) rHuEPO and patients in the observation group were treated by rHuEPO + 1.0 g t.i.d. p.o. Shengxuening tablet. The two groups were assisted by conventional treatments including iron, folic acid, vitamin B12 and L-carnitine. After 3 and 6 months, improvement of anemia was compared. After 3 months, the hemoglobin, hematocrit, serum ferritin and transferrin saturation levels of the observation group were significantly higher than those of the control group (p<0.05). In addition, C-reactive protein and superoxide dismutase levels of the observation group were significantly lower than those of the control group (p<0.05). After 6 months, indices of the observation group were ameliorated while the improvement of control group was not obvious, and indices of the observation group were significantly higher than those of the control group (p<0.05). Consumption of rHuEPO in the observation group was significantly less than that of the control group, and the total effective rate was significantly higher than that of the control group (p<0.05). In conclusion, Shengxuening tablet combined with rHuEPO was safe and effective for the treatment of renal anemia of MHD patients.
Collapse
Affiliation(s)
- XIN CHENG
- Division of Nephrology, Zhengzhou TCM Hospital, Zhengzhou, Henan 450000, P.R. China
| | - GUOJUN YU
- Division of Nephrology, The Affiliated Hospital of Henan Province Chinese Medicine Research Institute, Zhengzhou, Henan 450004, P.R. China
| | - JIANGPING HU
- Division of Nephrology, Zhengzhou TCM Hospital, Zhengzhou, Henan 450000, P.R. China
| | - XUEFENG XU
- Division of Nephrology, Zhengzhou TCM Hospital, Zhengzhou, Henan 450000, P.R. China
| | - FANG LUO
- Division of Endocrinology, Zhengzhou Central Hospital, Zhengzhou, Henan 450007, P.R. China
| | - PING SHEN
- Division of Nephrology, Zhengzhou TCM Hospital, Zhengzhou, Henan 450000, P.R. China
| | - GUOSHENG ZHANG
- Division of Nephrology, Zhengzhou TCM Hospital, Zhengzhou, Henan 450000, P.R. China
| | - NING YANG
- Department of Science and Education, Zhengzhou TCM Hospital, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
40
|
Li D, Tian Z, Tang W, Zhang J, Lu L, Sun Z, Zhou Z, Fan F. The Protective Effects of 5-Methoxytryptamine-α-lipoic Acid on Ionizing Radiation-Induced Hematopoietic Injury. Int J Mol Sci 2016; 17:ijms17060935. [PMID: 27314327 PMCID: PMC4926468 DOI: 10.3390/ijms17060935] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/27/2016] [Accepted: 06/03/2016] [Indexed: 02/06/2023] Open
Abstract
Antioxidants are prospective radioprotectors because of their ability to scavenge radiation-induced reactive oxygen species (ROS). The hematopoietic system is widely studied in radiation research because of its high radiosensitivity. In the present study, we describe the beneficial effects of 5-methoxytryptamine-α-lipoic acid (MLA), which was synthesized from melatonin and α-lipoic acid, against radiation-induced hematopoietic injury. MLA administration significantly enhanced the survival rate of mice after 7.2 Gy total body irradiation. The results showed that MLA not only markedly increased the numbers and clonogenic potential of hematopoietic cells but also decreased DNA damage, as determined by flow cytometric analysis of histone H2AX phosphorylation. In addition, MLA decreased the levels of ROS in hematopoietic cells by inhibiting NOX4 expression. These data demonstrate that MLA prevents radiation-induced hematopoietic syndrome by increasing the number and function of and by inhibiting DNA damage and ROS production in hematopoietic cells. These data suggest MLA is beneficial for the protection of radiation injuries.
Collapse
Affiliation(s)
- Deguan Li
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical Collage, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| | - Zhenyuan Tian
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical Collage, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| | - Weisheng Tang
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical Collage, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| | - Junling Zhang
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical Collage, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| | - Lu Lu
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical Collage, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| | - Zhaojin Sun
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical Collage, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| | - Zewei Zhou
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical Collage, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| | - Feiyue Fan
- Institute of Radiation Medicine, Chinese Academy of Medical Science and Peking Union Medical Collage, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| |
Collapse
|
41
|
Li H, Wang Z, Xu Y, Sun G. Pine polyphenols from Pinus koraiensis prevent injuries induced by gamma radiation in mice. PeerJ 2016; 4:e1870. [PMID: 27069807 PMCID: PMC4824883 DOI: 10.7717/peerj.1870] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 03/09/2016] [Indexed: 12/17/2022] Open
Abstract
Pine polyphenols (PPs) are bioactive dietary constituents that enhance health and help prevent diseases through antioxidants. Antioxidants reduce the level of oxidative damages caused by ionizing radiation (IR). The main purpose of this paper is to study the protective effect of PPs on peripheral blood, liver and spleen injuries in mice induced by IR. ICR (Institute of Cancer Research) male mice were administered orally with PPs (200 mg/kg b.wt.) once daily for 14 consecutive days prior to 7 Gy γ-radiations. PPs showed strong antioxidant activities. PPs significantly increased white blood cells, red blood cells and platelets counts. PPs also significantly reduced lipid peroxidation and increased the activities of superoxide dismutase, catalase and glutathione peroxidases, and the level of glutathione. PPs reduced the spleen morphologic injury. In addition, PPs inhibited mitochondria-dependent apoptosis pathways in splenocytes induced by IR. These results indicate that PPs are radioprotective promising reagents.
Collapse
Affiliation(s)
- Hui Li
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology , Harbin, Heilongjiang , China
| | - Zhenyu Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology , Harbin, Heilongjiang , China
| | - Yier Xu
- Department of Pharmacology, Pharmaceutical Academy of Harbin Pharmaceutical Group , Harbin, Heilongjiang , China
| | - Guicai Sun
- Department of Orthopaedics, The Fourth Hospital Affiliated to Nanchang University , Nanchang, Jiangxi , China
| |
Collapse
|
42
|
Newman NB, Sidhu MK, Baby R, Moss RA, Nissenblatt MJ, Chen T, Lu SE, Jabbour SK. Long-Term Bone Marrow Suppression During Postoperative Chemotherapy in Rectal Cancer Patients After Preoperative Chemoradiation Therapy. Int J Radiat Oncol Biol Phys 2016; 94:1052-60. [PMID: 27026312 DOI: 10.1016/j.ijrobp.2015.12.374] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 11/08/2015] [Accepted: 12/22/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE/OBJECTIVE(S) To quantify ensuing bone marrow (BM) suppression during postoperative chemotherapy resulting from preoperative chemoradiation (CRT) therapy for rectal cancer. METHODS AND MATERIALS We retrospectively evaluated 35 patients treated with preoperative CRT followed by postoperative 5-Fluorouracil and oxaliplatin (OxF) chemotherapy for locally advanced rectal cancer. The pelvic bone marrow (PBM) was divided into ilium (IBM), lower pelvis (LPBM), and lumbosacrum (LSBM). Dose volume histograms (DVH) measured the mean doses and percentage of BM volume receiving between 5-40 Gy (i.e.: PBM-V5, LPBM-V5). The Wilcoxon signed rank tests evaluated the differences in absolute hematologic nadirs during neoadjuvant vs. adjuvant treatment. Logistic regressions evaluated the association between dosimetric parameters and ≥ grade 3 hematologic toxicity (HT3) and hematologic event (HE) defined as ≥ grade 2 HT and a dose reduction in OxF. Receiver Operator Characteristic (ROC) curves were constructed to determine optimal threshold values leading to HT3. RESULTS During OxF chemotherapy, 40.0% (n=14) and 48% (n=17) of rectal cancer patients experienced HT3 and HE, respectively. On multivariable logistic regression, increasing pelvic mean dose (PMD) and lower pelvis mean dose (LPMD) along with increasing PBM-V (25-40), LPBM-V25, and LPBM-V40 were significantly associated with HT3 and/or HE during postoperative chemotherapy. Exceeding ≥36.6 Gy to the PMD and ≥32.6 Gy to the LPMD strongly correlated with causing HT3 during postoperative chemotherapy. CONCLUSIONS Neoadjuvant RT for rectal cancer has lasting effects on the pelvic BM, which are demonstrable during adjuvant OxF. Sparing of the BM during preoperative CRT can aid in reducing significant hematologic adverse events and aid in tolerance of postoperative chemotherapy.
Collapse
Affiliation(s)
- Neil B Newman
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Manpreet K Sidhu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Rekha Baby
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Rebecca A Moss
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Michael J Nissenblatt
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Ting Chen
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| | - Shou-En Lu
- Department of Biostatistics, School of Public Health, Rutgers University, Piscataway, New Jersey
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey.
| |
Collapse
|
43
|
Patwardhan RS, Sharma D, Thoh M, Checker R, Sandur SK. Baicalein exhibits anti-inflammatory effects via inhibition of NF-κB transactivation. Biochem Pharmacol 2016; 108:75-89. [PMID: 27019135 DOI: 10.1016/j.bcp.2016.03.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 03/16/2016] [Indexed: 12/22/2022]
Abstract
NF-κB is a crucial mediator of inflammatory and immune responses and a number of phytochemicals that can suppress this immune-regulatory transcription factor are known to have promising anti-inflammatory potential. However, we report that inducer of pro-inflammatory transcription factor NF-κB functions as an anti-inflammatory agent. Our findings reveal that a plant derived flavonoid baicalein could suppress mitogen induced T cell activation, proliferation and cytokine secretion. Treatment of CD4+ T cells with baicalein prior to transfer in to lymphopenic allogenic host significantly suppressed graft versus host disease. Interestingly, addition of baicalein to murine splenic lymphocytes induced DNA binding of NF-κB but did not suppress Concanavalin A induced NF-κB. Since baicalein did not inhibit NF-κB binding to DNA, we hypothesized that baicalein may be suppressing NF-κB trans-activation. Thioredoxin system is implicated in the regulation of NF-κB trans-activation potential and therefore inhibition of thioredoxin system may be responsible for suppression of NF-κB dependent genes. Baicalein not only inhibited TrxR activity in cell free system but also suppressed mitogen induced thioredoxin activity in the nuclear compartment of lymphocytes. Similar to baicalein, pharmacological inhibitors of thioredoxin system also could suppress mitogen induced T cell proliferation without inhibiting DNA binding of NF-κB. Further, activation of cellular thioredoxin system by the use of pharmacological activator or over-expression of thioredoxin could abrogate the anti-inflammatory action of baicalein. We propose a novel strategy using baicalein to limit NF-κB dependent inflammatory responses via inhibition of thioredoxin system.
Collapse
Affiliation(s)
- Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Maikho Thoh
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Rahul Checker
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India.
| |
Collapse
|