1
|
Redox Regulation of Autophagy in Cancer: Mechanism, Prevention and Therapy. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010098. [PMID: 36676047 PMCID: PMC9863886 DOI: 10.3390/life13010098] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
Reactive oxygen species (ROS), products of normal cellular metabolism, play an important role in signal transduction. Autophagy is an intracellular degradation process in response to various stress conditions, such as nutritional deprivation, organelle damage and accumulation of abnormal proteins. ROS and autophagy both exhibit double-edged sword roles in the occurrence and development of cancer. Studies have shown that oxidative stress, as the converging point of these stimuli, is involved in the mechanical regulation of autophagy process. The regulation of ROS on autophagy can be roughly divided into indirect and direct methods. The indirect regulation of autophagy by ROS includes post-transcriptional and transcriptional modulation. ROS-mediated post-transcriptional regulation of autophagy includes the post-translational modifications and protein interactions of AMPK, Beclin 1, PI3K and other molecules, while transcriptional regulation mainly focuses on p62/Keap1/Nrf2 pathway. Notably, ROS can directly oxidize key autophagy proteins, such as ATG4 and p62, leading to the inhibition of autophagy pathway. In this review, we will elaborate the molecular mechanisms of redox regulation of autophagy in cancer, and discuss ROS- and autophagy-based therapeutic strategies for cancer treatment.
Collapse
|
2
|
Liu Q, Ding X, Xu X, Lai H, Zeng Z, Shan T, Zhang T, Chen M, Huang Y, Huang Z, Dai X, Xia M, Cui S. Tumor-targeted hyaluronic acid-based oxidative stress nanoamplifier with ROS generation and GSH depletion for antitumor therapy. Int J Biol Macromol 2022; 207:771-783. [PMID: 35351548 DOI: 10.1016/j.ijbiomac.2022.03.139] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/15/2022] [Accepted: 03/22/2022] [Indexed: 01/01/2023]
Abstract
Tumor cells with innate oxidative stress are more susceptible to exogenous ROS-mediated oxidative damage than normal cells. However, the generated ROS could be scavenged by the overexpressed GSH in cancer cells, thus causing greatly restricted efficiency of ROS-mediated antitumor therapy. Herein, using cinnamaldehyde (CA) as a ROS generator while β-phenethyl isothiocyanate (PEITC) as a GSH scavenger, we designed a tumor-targeted oxidative stress nanoamplifier to elevate intracellular ROS level and synchronously suppress antioxidant systems, for thorough redox imbalance and effective tumor cells killing. First, an amphiphilic acid-sensitive cinnamaldehyde-modified hyaluronic acid conjugates (HA-CA) were synthesized, which could self-assemble into nano-assembly in aqueous media via strong hydrophobic interaction and π-π stacking. Then, aromatic PEITC was appropriately encapsulated into HA-CA nano-assembly to obtain HA-CA/PEITC nanoparticles. Through enhanced permeability retention (EPR) effect and specific CD44 receptor-mediated endocytosis, HA-CA/PEITC nanoparticles could accumulate in tumor tissues and successfully release CA and PEITC under acidic lysosomal environment. Both in vitro and in vivo results showed that the nanoparticles could efficiently boost oxidative stress of tumor cells via generating ROS and depleting GSH, and finally achieve superior antitumor efficacy. This nanoamplifier with good biosafety provides a potential strategy to augment ROS generation and suppress GSH for enhanced oxidation therapy.
Collapse
Affiliation(s)
- Qiuxing Liu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China
| | - Xin Ding
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China
| | - Xiaoyu Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Hualu Lai
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China
| | - Zishan Zeng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Ting Shan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Tao Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Meixu Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Zeqian Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Xiuling Dai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Meng Xia
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Shengmiao Cui
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China.
| |
Collapse
|
3
|
Orel VE, Krotevych M, Dasyukevich O, Rykhalskyi O, Syvak L, Tsvir H, Tsvir D, Garmanchuk L, Orel VВ, Sheina I, Rybka V, Shults NV, Suzuki YJ, Gychka SG. Effects induced by a 50 Hz electromagnetic field and doxorubicin on Walker-256 carcinosarcoma growth and hepatic redox state in rats. Electromagn Biol Med 2021; 40:475-487. [PMID: 34392747 DOI: 10.1080/15368378.2021.1958342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We compare the effects of an extremely low-frequency electromagnetic field (EMF) with the chemotherapeutic agent doxorubicin (DOX) on tumor growth and the hepatic redox state in Walker-256 carcinosarcoma-bearing rats. Animals were divided into five groups with one control (no tumor) and four tumor-bearing groups: no treatment, DOX, DOX combined with EMF and EMF. While DOX and DOX + EMF provided greater inhibition of tumor growth, treatment with EMF alone resulted in some level of antitumor effect (p < .05). Superoxide dismutase, catalase activity and glutathione content were significantly decreased in the liver of tumor-bearing animals as compared with the control group (p < .05). The decreases in antioxidant defenses accompanied histological findings of suspected liver damage. However, hepatic levels of thiobarbituric acid reactive substances, an indicator of lipid peroxidation, were three times lower in EMF and DOX + EMF groups than in no treatment and DOX (p < .05). EMF and DOX + EMF showed significantly lower activity of serum ALT than DOX alone (p < .05). These results indicate that EMF treatment can inhibit tumor growth, causing less pronounced oxidative stress damage to the liver. Therefore, EMF can be used as a therapeutic strategy to influence the hepatic redox state and combat cancer with reduced side-effects.
Collapse
Affiliation(s)
- Valerii E Orel
- Medical Physics and Bioengineering Research Laboratory, National Cancer Institute, Kyiv, Ukraine.,Biomedical Engineering Department, NTUU "Igor Sikorsky KPI", Kyiv, Ukraine
| | - Mykhailo Krotevych
- Research Department of the Pathological Anatomy, National Cancer Institute, Kyiv, Ukraine
| | - Olga Dasyukevich
- Medical Physics and Bioengineering Research Laboratory, National Cancer Institute, Kyiv, Ukraine
| | - Oleksandr Rykhalskyi
- Medical Physics and Bioengineering Research Laboratory, National Cancer Institute, Kyiv, Ukraine
| | - Liubov Syvak
- Research Department of Chemotherapy Solid Tumors, National Cancer Institute, Kyiv, Ukraine
| | | | - Dmytro Tsvir
- Medical Faculty, Bogomolets National Medical University, Kyiv, Ukraine
| | - Lyudmyla Garmanchuk
- Department of Biomedicine, NSC "Institute of Biology and Medicine" of the Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Valerii В Orel
- Biomedical Engineering Department, NTUU "Igor Sikorsky KPI", Kyiv, Ukraine.,Research Department of Radiodiagnostics, National Cancer Institute, Kyiv, Ukraine
| | - Iryna Sheina
- Department of Medical Physics and Biomedical Nanotechnologies, V. N. Karazin Kharkiv National University, Kharkiv, Ukraine
| | - Vladyslava Rybka
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Nataliia V Shults
- Department of Medical Physics and Biomedical Nanotechnologies, V. N. Karazin Kharkiv National University, Kharkiv, Ukraine
| | - Yuichiro J Suzuki
- Department of Medical Physics and Biomedical Nanotechnologies, V. N. Karazin Kharkiv National University, Kharkiv, Ukraine
| | - Sergiy G Gychka
- Department of Pathological Anatomy 2, Bogomolets National Medical University, Kyiv, Ukraine
| |
Collapse
|
4
|
Bovine Dialyzable Leukocyte Extract IMMUNEPOTENT-CRP Induces Selective ROS-Dependent Apoptosis in T-Acute Lymphoblastic Leukemia Cell Lines. JOURNAL OF ONCOLOGY 2020; 2020:1598503. [PMID: 32587616 PMCID: PMC7298273 DOI: 10.1155/2020/1598503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/15/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022]
Abstract
Immunotherapies strengthen the immune system to fight multiple diseases such as infections, immunodeficiencies, and autoimmune diseases, and recently, they are being used as an adjuvant in cancer treatment. IMMUNEPOTENT-CRP (I-CRP) is an immunotherapy made of bovine dialyzable leukocyte extract (bDLE) that has chemoprotective and immunomodulatory effects in different cellular populations of the immune system and antitumor activity in different cancer cell lines. Our recent results suggest that the antineoplastic effect of I-CRP is due to the characteristics of cancer cells. To confirm, we evaluated whether the selectivity is due to cell lineage or characteristics of cancer cells, testing cytotoxicity in T-acute lymphoblastic leukemia cells and their cell death mechanism. Here, we assessed the effect of I-CRP on cell viability and cell death. To determine the mechanism of cell death, we tested cell cycle, mitochondrial and nuclear alterations, and caspases and reactive oxygen species (ROS) and their role in cell death mechanism. Our results show that I-CRP does not affect cell viability in noncancer cells and induces selective cytotoxicity in a dose-dependent manner in leukemic cell lines. I-CRP also induces mitochondrial damage through proapoptotic and antiapoptotic protein modulation (Bax and Bcl-2) and ROS production, nuclear alterations including DNA damage (γ-H2Ax), overexpression of p53, cell cycle arrest, and DNA degradation. I-CRP induced ROS-dependent apoptosis in leukemic cells. Overall, here, we show that I-CRP cytotoxicity is selective to leukemic cells, inducing ROS-dependent apoptosis. This research opens the door to further exploration of their role in the immune system and the cell death mechanism that could potentially work in conjunction with other therapies including hematological malignances.
Collapse
|
5
|
Kong W, Li C, Qi Q, Shen J, Chang K. Cardamonin induces G2/M arrest and apoptosis via activation of the JNK-FOXO3a pathway in breast cancer cells. Cell Biol Int 2020; 44:177-188. [PMID: 31393045 DOI: 10.1002/cbin.11217] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 08/04/2019] [Indexed: 01/24/2023]
Abstract
Cardamonin (CD), a naturally occurring chalcone isolated from large black cardamom, was previously reported to suppress the proliferation of breast cancer cells. However, its precise molecular anti-tumor mechanisms have not been well elucidated. In this study, we found that CD markedly inhibited the proliferation of MDA-MB 231 and MCF-7 breast cancer cells through the induction of G2/M arrest and apoptosis. Reactive oxygen species (ROS) plays a pivotal role in the inhibition of CD-induced cell proliferation. Treatment with N-acetyl-cysteine (NAC), an ROS scavenger, blocked CD-induced G2/M arrest and apoptosis in this study. Quenching of ROS by overexpression of catalase also blocked CD-induced cell cycle arrest and apoptosis. We showed that CD enhanced the expression and nuclear translocation of Forkhead box O3 (FOXO3a) via upstream c-Jun N-terminal kinase, inducing the expression of FOXO3a and its target genes, including p21, p27, and Bim. This process led to the reduction of cyclin D1 and enhancement of activated caspase-3 expression. The addition of NAC markedly reversed these effects, knockdown of FOXO3a using small interfering RNA also decreased CD-induced G2/M arrest and apoptosis. In vivo, CD efficiently suppressed the growth of MDA-MB 231 breast cancer xenograft tumors. Taken together, our data provide a molecular mechanistic rationale for CD-induced cell cycle arrest and apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Weiwei Kong
- Department of Blood Transfusion, The Third Affiliated Hospital of Xinxiang Medical University, 453000, Xinxiang, China
| | - Chuang Li
- Department of Laboratory Medicine, The Third Affiliated Hospital of Xinxiang Medical University, 453000, Xinxiang, China
| | - Qiaofang Qi
- Xinxiang Medical University, 453000, Xinxiang, China
| | - Jiahui Shen
- Department of Blood Transfusion, The Third Affiliated Hospital of Xinxiang Medical University, 453000, Xinxiang, China
| | - Kaiwen Chang
- Xinxiang Medical University, 453000, Xinxiang, China
| |
Collapse
|
6
|
Lu H, Bai L, Zhou Y, Lu Y, Jiang Z, Shi J. Recent Study of Dual HDAC/PARP Inhibitor for the Treatment of Tumor. Curr Top Med Chem 2019; 19:1041-1050. [PMID: 31161991 DOI: 10.2174/1568026619666190603092407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/27/2019] [Accepted: 04/08/2019] [Indexed: 12/25/2022]
Abstract
The occurrence and development of tumors are closely related to epigenetic instability which modulates gene expression through DNA methylation, histone modification, chromatin remodeling, and RNA-related silencing. Histone deacetylase (HDAC) and poly (ADP-ribose) polymerase (PARP) are targets of epigenetic regulation. Over the years, a large number of studies have shown that HDAC inhibitors and PARP inhibitors have synergistic effects in the treatment of tumors, and there are reports of related dual HDAC/PARP inhibitors. This review will give a brief summary of the synergistic mechanisms of HDAC inhibitors and PARP inhibitors and introduce the design of the first dual HDAC/PARP inhibitor, which may guide the design of more dual HDAC/PARP inhibitors for the treatment of tumors.
Collapse
Affiliation(s)
- Haiying Lu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Bai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanping Zhou
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yongping Lu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhongliang Jiang
- Miller School of Medicine, University of Miami, Miami, Florida, United States
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
7
|
Bi W, Bi Y, Li P, Hou S, Yan X, Hensley C, Bammert CE, Zhang Y, Gibson KM, Ju J, Bi L. Indole Alkaloid Derivative B, a Novel Bifunctional Agent That Mitigates 5-Fluorouracil-Induced Cardiotoxicity. ACS OMEGA 2018; 3:15850-15864. [PMID: 30533582 PMCID: PMC6275955 DOI: 10.1021/acsomega.8b02139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/23/2018] [Indexed: 06/09/2023]
Abstract
Clinically approved therapeutics that mitigate chemotherapy-induced cardiotoxicity, a serious adverse effect of chemotherapy, are lacking. The aim of this study was to determine the putative protective capacity of a novel indole alkaloid derivative B (IADB) against 5-fluorouracil (5-FU)-induced cardiotoxicity. To assess the free-radical scavenging activities of IADB, the acetylcholine-induced relaxation assay in rat thoracic aorta was used. Further, IADB was tested in normal and cancer cell lines with assays gauging autophagy induction. We further examined whether IADB could attenuate cardiotoxicity in 5-FU-treated male ICR mice. We found that IADB could serve as a novel bifunctional agent (displaying both antioxidant and autophagy-modulating activities). Further, we demonstrated that IADB induced production of cytosolic autophagy-associated structures in both cancer and normal cell lines. We observed that IADB cytotoxicity was much lower in normal versus cancer cell lines, suggesting an enhanced potency toward cancer cells. The cardiotoxicity induced by 5-FU was significantly relieved in animals pretreated with IADB. Taken together, IADB treatment, in combination with chemotherapy, may lead to reduced cardiotoxicity, as well as the reduction of anticancer drug dosages that may further improve chemotherapeutic efficacy with decreased off-target effects. Our data suggest that the use of IADB may be therapeutically beneficial in minimizing cardiotoxicity associated with high-dose chemotherapy. On the basis of the redox status difference between normal and tumor cells, IADB selectively induces autophagic cell death, mediated by reactive oxygen species overproduction, in cancer cells. This novel mechanism could reveal novel therapeutic targets in chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Wei Bi
- Second
Hospital of HeBei Medical University, Shijiazhuang 050000, P. R. China
| | - Yue Bi
- Second
Hospital of HeBei Medical University, Shijiazhuang 050000, P. R. China
| | - Pengfei Li
- Second
Hospital of HeBei Medical University, Shijiazhuang 050000, P. R. China
| | - Shanshan Hou
- Department
of Chemistry and Biological Sciences, Michigan
Technological University, Houghton, Michigan 49931, United States
| | - Xin Yan
- Department
of Chemistry and Biological Sciences, Michigan
Technological University, Houghton, Michigan 49931, United States
| | - Connor Hensley
- Department
of Chemistry and Biological Sciences, Michigan
Technological University, Houghton, Michigan 49931, United States
| | - Catherine E. Bammert
- Department
of Chemistry and Biological Sciences, Michigan
Technological University, Houghton, Michigan 49931, United States
| | - Yanrong Zhang
- Second
Hospital of HeBei Medical University, Shijiazhuang 050000, P. R. China
| | - K. Michael Gibson
- Department
of Pharmacotherapy, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Jingfang Ju
- Translational
Research Laboratory, Department of Pathology, Stony Brook University, Stony
Brook, New York 11794, United States
| | - Lanrong Bi
- Department
of Chemistry and Biological Sciences, Michigan
Technological University, Houghton, Michigan 49931, United States
| |
Collapse
|
8
|
Lan H, Yuan H, Lin C. Sulforaphane induces p53‑deficient SW480 cell apoptosis via the ROS‑MAPK signaling pathway. Mol Med Rep 2017; 16:7796-7804. [PMID: 28944886 DOI: 10.3892/mmr.2017.7558] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 05/08/2017] [Indexed: 11/06/2022] Open
Abstract
Sulforaphane (SFN) has been revealed to inhibit the growth and induce apoptosis of cancer cells. However, the detailed anticancer effects of SFN on p53‑deficient colon cancer cells has yet to be clearly elucidated. The present study employed p53‑deficient SW480 cells to establish an SFN‑induced in vitro model of apoptosis. The critical events leading to apoptosis were then evaluated in SFN‑treated p53‑deficient SW480 cells, by performing an MTT assay, flow cytometry, western blotting and ELISA. The results demonstrated that SFN at concentrations of 5, 10, 15 and 20 µM induced mitochondria‑associated cell apoptosis, which was further confirmed by disruption of the mitochondrial membrane potential, an increase in the Bax/Bcl‑2 ratio, as well as activation of caspase‑3, ‑7 and ‑9. In addition, SFN‑induced apoptosis was associated with an increase in the generation of reactive oxygen species (ROS), and the activation of extracellular signal‑regulated kinases (Erk) and p38 mitogen‑activated protein kinases. However, SFN did not induce expression of the p53 family member, p73. SFN‑induced apoptosis was subsequently confirmed to be ROS‑dependent and associated with Erk/p38, as the specific inhibitors for ROS, phosphorylated (p)‑Erk and p‑p38, completely or partially attenuated the SFN‑induced reduction in SW480 cell viability. In addition, the results demonstrated that even at the lowest concentrations (5 µM), SFN increased the sensitivity of p53‑proficient HCT‑116 cells to cisplatin. In conclusion, the results suggest that SFN may induce apoptosis in p53‑deficient SW480 cells via p53/p73‑independent and ROS‑Erk/p38‑dependent signaling pathways.
Collapse
Affiliation(s)
- Hai Lan
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Hongyin Yuan
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Congyao Lin
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
9
|
Tietel Z, Masaphy S. True morels (Morchella)—nutritional and phytochemical composition, health benefits and flavor: A review. Crit Rev Food Sci Nutr 2017; 58:1888-1901. [DOI: 10.1080/10408398.2017.1285269] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Zipora Tietel
- Gilat Research Center, Agricultural Research Organization, M.P. Negev Israel
| | - Segula Masaphy
- Applied Microbiology and Mycology Department, MIGAL, Kiryat Shmona, Israel
- Tel Hai College, Upper Galilee, Israel
| |
Collapse
|