1
|
Tao XN, Liu HT, Xiang XW, Zhu CH, Qiu J, Zhao H, Liu KF. Regulating the Distribution and Accumulation of Charged Molecules by Progressive Electroporation for Improved Intracellular Delivery. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36063-36076. [PMID: 38958208 DOI: 10.1021/acsami.4c05340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The cell membrane separates the intracellular compartment from the extracellular environment, constraining exogenous molecules to enter the cell. Conventional electroporation typically employs high-voltage and short-duration pulses to facilitate the transmembrane transport of molecules impermeable to the membrane under natural conditions by creating temporary hydrophilic pores on the membrane. Electroporation not only enables the entry of exogenous molecules but also directs the intracellular distribution of the electric field. Recent advancements have markedly enhanced the efficiency of intracellular molecule delivery, achieved through the utilization of microstructures, microelectrodes, and surface modifications. However, little attention is paid to regulating the motion of molecules during and after passing through the membrane to improve delivery efficiency, resulting in an unsatisfactory delivery efficiency and high dose demand. Here, we proposed the strategy of regulating the motion of charged molecules during the delivery process by progressive electroporation (PEP), utilizing modulated electric fields. Efficient delivery of charged molecules with an expanded distribution and increased accumulation by PEP was demonstrated through numerical simulations and experimental results. The dose demand can be reduced by 10-40% depending on the size and charge of the molecules. We confirmed the safety of PEP for intracellular delivery in both short and long terms through cytotoxicity assays and transcriptome analysis. Overall, this work not only reveals the mechanism and effectiveness of PEP-enhanced intracellular delivery of charged molecules but also suggests the potential integration of field manipulation of molecular motion with surface modification techniques for biomedical applications such as cell engineering and sensitive cellular monitoring.
Collapse
Affiliation(s)
- Xiao-Nan Tao
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Hao-Tian Liu
- Academy for Engineering & Technology, Fudan University, Shanghai 200433, China
| | - Xiao-Wei Xiang
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Cai-Hui Zhu
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Jian Qiu
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Hui Zhao
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| | - Ke-Fu Liu
- School of Information Science and Technology, Fudan University, Shanghai 200433, China
| |
Collapse
|
2
|
Zhuang C, Sun R, Zhang Y, Zou Q, Zhou J, Dong N, Zhao X, Fu W, Geng X, Wang J, Li Q, Zhao RC. Treatment of Rheumatoid Arthritis Based on the Inherent Bioactivity of Black Phosphorus Nanosheets. Aging Dis 2024:AD.2024.0319. [PMID: 38913037 DOI: 10.14336/ad.2024.0319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that affects the living quality of patients, especially the elderly population. RA-related morbidity and mortality increase significantly with age, while current clinical drugs for RA are far from satisfactory and may have serious side effects. Therefore, the development of new drugs with higher biosafety and efficacy is demanding. Black phosphorus nanosheets (BPNSs) have been widely studied because of their excellent biocompatibility. Here, we focus on the inherent bioactivity of BPNSs, report the potential of BPNSs as a therapeutic drug for RA and elucidate the underlying therapeutic mechanism. We find that BPNSs inhibit autophagy at an early stage via the AMPK-mTOR pathway, switch the energy metabolic pathway to oxidative phosphorylation, increase intracellular ATP levels, suppress apoptosis, reduce inflammation and oxidative stress, and down-regulate senescence-associated secretory phenotype (SASP)-related genes in rheumatoid arthritis synovial fibroblasts (RA-SFs). Further, BPNSs induce the apoptosis of macrophages and promote their transition from the M1 to the M2 phenotype by regulating related cytokines. Significantly, the administration of BPNSs can alleviate key pathological features of RA in mice, revealing great therapeutic potential. This study provides a novel option for treating RA, with BPNSs emerging as a promising therapeutic candidate.
Collapse
Affiliation(s)
- Cheng Zhuang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Ruiqi Sun
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Yuchen Zhang
- School of Medicine, Shanghai University, Shanghai, China
| | - Qing Zou
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Jianxin Zhou
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Naijun Dong
- School of Life Sciences, Shanghai University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Xuyu Zhao
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Wenjun Fu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiaoke Geng
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qian Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
- Cell Energy Life Sciences Group Co. LTD, Qingdao, China, 266200
| | - Robert Chunhua Zhao
- School of Life Sciences, Shanghai University, Shanghai, China
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| |
Collapse
|
3
|
Thulasidas JS, Varadarajan GS, Camarillo I, Mittal L, Sundararajan R. Efficacy of electrical pulse mediated tomato lipophilic extract on human breast cancer cell. J Cancer Res Ther 2023; 19:S106-S115. [PMID: 37147990 DOI: 10.4103/jcrt.jcrt_1117_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Aim The purpose of this research is to study the effect of electrical pulse mediated tomato lipophilic extract (TLE) on human breast cancer MCF-7 and non-tumorigenic MCF-10A cells. Materials and Methods MCF-7 and MCF-10A cells were treated with 50 μg/mL TLE and eight 100 μs electric pulses of different electric field intensities (800, 1000, and 1200 V/cm), and the viability was studied using real time MT assay at 24 h of treatment. In addition, we studied cell viability of both the cells at 0 h using trypan blue assay and the ability to form colonies of both cells using colony forming unit (CFU) assay for all the treatments. We also imaged the cells at 24 h using microscope. Results With 50 μg/mL TLE, the cell viability of MCF-7 and MCF-10A was same (84%). When the same concentration of TLE is combined with eight electrical pulses of 1200 V/cm, the cell viability of MCF-7 and MCF-10A was 2% and 87%, respectively. These results indicate that the effect of electrical pulses mediated TLE was higher on cancerous MCF-7 cells when compared to non-cancerous MCF-10A cells. Conclusion The combination of electrical pulses with TLE is an effective strategy to selectively target cancer cells in the body.
Collapse
Affiliation(s)
- Jeya Shree Thulasidas
- Department of sEEE, College of Engineering, Division of High Voltage Engineering, Anna University, Chennai, Tamil Nadu, India
| | - Gowri Sree Varadarajan
- Department of sEEE, College of Engineering, Division of High Voltage Engineering, Anna University, Chennai, Tamil Nadu, India
| | - Ignacio Camarillo
- Department of Biological Sciences; Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA, India
| | - Lakshya Mittal
- School of Engineering Technology, Purdue University, West Lafayette, Indiana, USA
| | - Raji Sundararajan
- School of Engineering Technology, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
4
|
Marni R, Malla M, Chakraborty A, Malla R. Proteomic profiling and ROC analysis identify CD151 and ELAVL1 as potential therapy response markers for the antiviral drug in resistant TNBC. Life Sci 2023; 320:121534. [PMID: 36889667 DOI: 10.1016/j.lfs.2023.121534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023]
Abstract
Triple-negative breast cancer is high heterogeneous, aggressive, and metastatic with poor prognosis. Despite of advances in targeted therapies, TNBC has been reported to cause high morbidity and mortality. A rare subpopulation within the tumor microenvironment organized into a hierarchy of cancer stem cells is responsible for therapy resistance and tumor recurrence. Repurposing of antiviral drugs for cancer treatment is gaining momentum due to reduced cost, labour, and research time, but limited due to lack of prognostic, and predictive markers. The present study investigates proteomic profiling and ROC analysis to identify CD151 and ELAVL1 as potential therapy response markers for the antiviral drug 2-thio-6-azauridine (TAU) in resistant TNBC. The stemness of MDA-MB 231 and MDA-MD 468 adherent cells was enriched by culturing them under non-adherent and non-differentiation conditions. Then, CD151+ subpopulation was isolated and characterized for the enrichment of stemness. This study found that CD151 has overexpressed in stemness enriched subpopulations, and also showed CD44 high and CD24 low expression along with stem cell-related transcription factors octamer-binding transcription factor 4 (OCT4) and Sex determining Y-box 2 (SOX2). This study also found that TAU induced significant cytotoxicity and genotoxicity in the CD151+TNBC subpopulation and inhibited their proliferation by inducing DNA damage, cell cycle arrest at the G2M phase, and apoptosis. Further, a proteomic profiling study showed that the expression of CD151 along with ELAVL1, an RNA-binding protein, was significantly reduced with TAU treatment. KM plotter showed correlation of CD151 and ELAVL1 gene expression with a poor prognosis of TNBC. ROC analysis predicted and validated CD151 and ELAVL1 as best therapy response marker for TAU in TNBC. These findings provide new insight into repurposing antiviral drug TAU for treatment of metastatic and drug resistant TNBC.
Collapse
Affiliation(s)
- Rakshmitha Marni
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, A.P., India
| | - Manas Malla
- Department of Computer Science and Engineering, GITAM School of Technology, GITAM (Deemed to be University), Visakhapatnam 530045, A.P., India
| | | | - RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, A.P., India.
| |
Collapse
|
5
|
Advances of Electroporation-Related Therapies and the Synergy with Immunotherapy in Cancer Treatment. Vaccines (Basel) 2022; 10:vaccines10111942. [PMID: 36423037 PMCID: PMC9692484 DOI: 10.3390/vaccines10111942] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Electroporation is the process of instantaneously increasing the permeability of a cell membrane under a pulsed electric field. Depending on the parameters of the electric pulses and the target cell electrophysiological characteristics, electroporation can be either reversible or irreversible. Reversible electroporation facilitates the delivery of functional genetic materials or drugs to target cells, inducing cell death by apoptosis, mitotic catastrophe, or pseudoapoptosis; irreversible electroporation is an ablative technology which directly ablates a large amount of tissue without causing harmful thermal effects; electrotherapy using an electric field can induce cell apoptosis without any aggressive invasion. Reversible and irreversible electroporation can also activate systemic antitumor immune response and enhance the efficacy of immunotherapy. In this review, we discuss recent progress related to electroporation, and summarize its latest applications. Further, we discuss the synergistic effects of electroporation-related therapies and immunotherapy. We also propose perspectives for further investigating electroporation and immunotherapy in cancer treatment.
Collapse
|
6
|
Kulbacka J, Rembiałkowska N, Szewczyk A, Rossowska J, Drąg-Zalesińska M, Kulbacki M, Choromańska A. Nanosecond PEF Induces Oxidative Stress and Apoptosis via Proteasomal Activity Inhibition in Gastric Adenocarcinoma Cells with Drug Resistance. Int J Mol Sci 2022; 23:12943. [PMID: 36361727 PMCID: PMC9657809 DOI: 10.3390/ijms232112943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/06/2022] [Accepted: 10/18/2022] [Indexed: 08/01/2023] Open
Abstract
Nanosecond (ns) pulsed electric field (PEF) is a technology in which the application of ultra-short electrical pulses can be used to disrupt the barrier function of cell plasma and internal membranes. Disruptions of the membrane integrity cause a substantial imbalance in cell homeostasis in which oxidative stress is a principal component. In the present study, nsPEF-induced oxidative stress was investigated in two gastric adenocarcinoma cell lines (EPG85-257P and EPG85-257RDB) which differ by their sensitivity to daunorubicin. Cells were exposed to 200 pulses of 10 ns duration, with the amplitude and pulse repetition frequency at 1 kHz, with electric field intensity varying from 12.5 to 50 kV/cm. The electroporation buffer contained either 1 mM or 2 mM calcium chloride. CellMask DeepRed visualized cell plasma permeabilization, Fluo-4 was used to visualize internal calcium ions content, and F-actin was labeled with AlexaFluor®488 for the cytoskeleton. The cellular viability was determined by MTT assay. An alkaline and neutral comet assay was employed to detect apoptotic and necrotic cell death. The luminescent method estimated the modifications in GSSG/GSH redox potential and the imbalance of proteasomal activity (chymotrypsin-, trypsin- and caspase-like). The reactive oxygen species (ROS) level was measured by flow cytometry using dihydroethidium (DHE) dye. Morphological visualization indicated cell shrinkage, affected cell membranes (characteristic bubbles and changed cell shape), and the reorganization of actin fibers with sites of its dense concentration; the effect was more intense with the increasing electric field strength. The most significant decrease in cell viability and GSSG/GSH redox potential was noted at the highest amplitude of 50 kV/cm, and calcium ions amplified this effect. nsPEF, particularly with calcium ions, inhibited proteasomal activities, resulting in increased protein degradation. nsPEF increased the percentage of apoptotic cells and ROS levels. The EPG85-257 RDB cell line, which is resistant to standard chemotherapy, was more sensitive to applied nsPEF protocols. The applied nsPEF method disrupted the metabolism of cancer cells and induced apoptotic cell death. The nsPEF ability to cause apoptosis, oxidative stress, and protein degradation make the nsPEF methodology a suitable alternative to current anticancer pharmacological methods.
Collapse
Affiliation(s)
- Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-556 Wroclaw, Poland
| | - Nina Rembiałkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-556 Wroclaw, Poland
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-556 Wroclaw, Poland
- Department of Animal Developmental Biology, Faculty of Biological Sciences, University of Wroclaw, 50-335 Wroclaw, Poland
| | - Joanna Rossowska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 50-422 Wroclaw, Poland
| | - Małgorzata Drąg-Zalesińska
- Division of Histology and Embryology, Division of Human Morpholog and Embryology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Marek Kulbacki
- Polish-Japanese Academy of Information Technology, 02-008 Warsaw, Poland
- DIVE IN AI, 53-307 Wroclaw, Poland
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-556 Wroclaw, Poland
| |
Collapse
|
7
|
Electrochemotherapy: An Alternative Strategy for Improving Therapy in Drug-Resistant SOLID Tumors. Cancers (Basel) 2022; 14:cancers14174341. [PMID: 36077875 PMCID: PMC9454613 DOI: 10.3390/cancers14174341] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Chemotherapy is becoming an increasingly difficult antitumor therapy to practice due to the multiple mechanisms of drug resistance. To overcome the problem, it is possible to use alternative techniques, such as electrochemotherapy, which involves the simultaneous administration of the electrical pulse (electroporation) and the treatment with the drug in order to improve the effectiveness of the drug against the tumor. Electroporation has improved the efficacy of some chemotherapeutic agents, such bleomycin, cisplatin, mitomycin C, and 5-fluorouracil. The results of in vitro, veterinary, and clinical oncology studies are promising on various cancers, such as metastatic melanoma. The purpose of this review is to give an update on the state of the art of electrochemotherapy against the main solid tumors in the preclinical, clinical, and veterinary field. Abstract Electrochemotherapy (ECT) is one of the innovative strategies to overcome the multi drug resistance (MDR) that often occurs in cancer. Resistance to anticancer drugs results from a variety of factors, such as genetic or epigenetic changes, an up-regulated outflow of drugs, and various cellular and molecular mechanisms. This technology combines the administration of chemotherapy with the application of electrical pulses, with waveforms capable of increasing drug uptake in a non-toxic and well tolerated mechanical system. ECT is used as a first-line adjuvant therapy in veterinary oncology, where it improves the efficacy of many chemotherapeutic agents by increasing their uptake into cancer cells. The chemotherapeutic agents that have been enhanced by this technique are bleomycin, cisplatin, mitomycin C, and 5-fluorouracil. After their use, a better localized control of the neoplasm has been observed. In humans, the use of ECT was initially limited to local palliative therapy for cutaneous metastases of melanoma, but phase I/II studies are currently ongoing for several histotypes of cancer, with promising results. In this review, we described the preclinical and clinical use of ECT on drug-resistant solid tumors, such as head and neck squamous cell carcinoma, breast cancer, gynecological cancer and, finally, colorectal cancer.
Collapse
|
8
|
Lv L, Yang S, Zhu Y, Zhai X, Li S, Tao X, Dong D. Relationship between metabolic reprogramming and drug resistance in breast cancer. Front Oncol 2022; 12:942064. [PMID: 36059650 PMCID: PMC9434120 DOI: 10.3389/fonc.2022.942064] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Breast cancer is the leading cause of cancer death in women. At present, chemotherapy is the main method to treat breast cancer in addition to surgery and radiotherapy, but the process of chemotherapy is often accompanied by the development of drug resistance, which leads to a reduction in drug efficacy. Furthermore, mounting evidence indicates that drug resistance is caused by dysregulated cellular metabolism, and metabolic reprogramming, including enhanced glucose metabolism, fatty acid synthesis and glutamine metabolic rates, is one of the hallmarks of cancer. Changes in metabolism have been considered one of the most important causes of resistance to treatment, and knowledge of the mechanisms involved will help in identifying potential treatment deficiencies. To improve women's survival outcomes, it is vital to elucidate the relationship between metabolic reprogramming and drug resistance in breast cancer. This review analyzes and investigates the reprogramming of metabolism and resistance to breast cancer therapy, and the results offer promise for novel targeted and cell-based therapies.
Collapse
Affiliation(s)
- Linlin Lv
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian, China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanna Zhu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaohan Zhai
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuai Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Harvey KE, LaVigne EK, Dar MS, Salyer AE, Pratt EPS, Sample PA, Aryal UK, Gowher H, Hockerman GH. RyR2/IRBIT regulates insulin gene transcript, insulin content, and secretion in the insulinoma cell line INS-1. Sci Rep 2022; 12:7713. [PMID: 35562179 PMCID: PMC9095623 DOI: 10.1038/s41598-022-11276-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/31/2022] [Indexed: 12/01/2022] Open
Abstract
The role of ER Ca2+ release via ryanodine receptors (RyR) in pancreatic β-cell function is not well defined. Deletion of RyR2 from the rat insulinoma INS-1 (RyR2KO) enhanced IP3 receptor activity stimulated by 7.5 mM glucose, coincident with reduced levels of the protein IP3 Receptor Binding protein released with Inositol 1,4,5 Trisphosphate (IRBIT). Insulin content, basal (2.5 mM glucose) and 7.5 mM glucose-stimulated insulin secretion were reduced in RyR2KO and IRBITKO cells compared to controls. INS2 mRNA levels were reduced in both RyR2KO and IRBITKO cells, but INS1 mRNA levels were specifically decreased in RyR2KO cells. Nuclear localization of S-adenosylhomocysteinase (AHCY) was increased in RyR2KO and IRBITKO cells. DNA methylation of the INS1 and INS2 gene promotor regions was very low, and not different among RyR2KO, IRBITKO, and controls, but exon 2 of the INS1 and INS2 genes was more extensively methylated in RyR2KO and IRBITKO cells. Exploratory proteomic analysis revealed that deletion of RyR2 or IRBIT resulted in differential regulation of 314 and 137 proteins, respectively, with 41 in common. These results suggest that RyR2 regulates IRBIT levels and activity in INS-1 cells, and together maintain insulin content and secretion, and regulate the proteome, perhaps via DNA methylation.
Collapse
Affiliation(s)
- Kyle E Harvey
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Emily K LaVigne
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
- Interdisciplinary Life Sciences Program, Purdue University, West Lafayette, IN, USA
| | - Mohd Saleem Dar
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Amy E Salyer
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Evan P S Pratt
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
- Interdisciplinary Life Sciences Program, Purdue University, West Lafayette, IN, USA
| | - Paxton A Sample
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Uma K Aryal
- Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | - Humaira Gowher
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Gregory H Hockerman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
10
|
Electroporation and Electrochemotherapy in Gynecological and Breast Cancer Treatment. Molecules 2022; 27:molecules27082476. [PMID: 35458673 PMCID: PMC9026735 DOI: 10.3390/molecules27082476] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/14/2022] [Accepted: 04/10/2022] [Indexed: 12/24/2022] Open
Abstract
Gynecological carcinomas affect an increasing number of women and are associated with poor prognosis. The gold standard treatment plan is mainly based on surgical resection and subsequent chemotherapy with cisplatin, 5-fluorouracil, anthracyclines, or taxanes. Unfortunately, this treatment is becoming less effective and is associated with many side effects that negatively affect patients’ physical and mental well-being. Electroporation based on tumor exposure to electric pulses enables reduction in cytotoxic drugs dose while increasing their effectiveness. EP-based treatment methods have received more and more interest in recent years and are the subject of a large number of scientific studies. Some of them show promising therapeutic potential without using any cytotoxic drugs or molecules already present in the human body (e.g., calcium electroporation). This literature review aims to present the fundamental mechanisms responsible for the course of EP-based therapies and the current state of knowledge in the field of their application in the treatment of gynecological neoplasms.
Collapse
|
11
|
Hall FG, Liceaga AM. Isolation and proteomic characterization of tropomyosin extracted from edible insect protein. FOOD CHEMISTRY. MOLECULAR SCIENCES 2021; 3:100049. [PMID: 35415661 PMCID: PMC8991843 DOI: 10.1016/j.fochms.2021.100049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/20/2021] [Accepted: 10/31/2021] [Indexed: 11/25/2022]
Abstract
Edible insects are considered promising sustainable protein sources. Thermal treatments and proteolysis are commonly used to improve their safety and quality. However, their allergenicity remains mostly unexplored. Tropomyosin, a major insect pan-allergen, can be used to study processing effects on its immunoreactivity. In this study, selective precipitation was used to extract tropomyosin from heated and protease-treated crickets. Immunoinformatics predicted 31 epitope regions, while proteomic analysis suggested decreased amounts of intact epitope regions in microwave-heated/protease-treated crickets. Tropomyosin peptide sequences were identified in higher abundance in convection-heated samples. Finally, tropomyosin immunoreactivity by immunoblotting and ELISA, revealed that protease treatments under microwave heating had lower (p < 0.05) IgE and IgG reactivity. Based on results, processing insects using proteolysis and microwave-heating could be effective for generating hypoallergenic cricket protein ingredients. The use of proteomics and bioinformatics proved to be useful tools in understanding the impact of processing on allergenic reactivity of insect proteins.
Collapse
Affiliation(s)
- Felicia G. Hall
- Protein Chemistry and Bioactive Peptides Laboratory, Department of Food Science, Purdue University, 745 Agriculture Mall Dr., West Lafayette, IN 47907, USA
| | - Andrea M. Liceaga
- Protein Chemistry and Bioactive Peptides Laboratory, Department of Food Science, Purdue University, 745 Agriculture Mall Dr., West Lafayette, IN 47907, USA
| |
Collapse
|
12
|
Enhanced Antiproliferation Potency of Electrical Pulse-Mediated Metformin and Cisplatin Combination Therapy on MDA-MB-231 Cells. Appl Biochem Biotechnol 2021; 194:18-36. [PMID: 34741262 DOI: 10.1007/s12010-021-03723-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/08/2021] [Indexed: 10/19/2022]
Abstract
We investigated the combined potency of metformin and cisplatin on the MDA-MB-231, triple-negative breast cancer (TNBC) cells with the application of electrical pulses. There are no targeted therapies for this subset of breast cancer because of the absence of specific biomarkers. Cytotoxic chemotherapy is the mainstream mode of treatment for TNBC, and cisplatin is the most commonly used chemotherapeutic drug. While there is a good response initially, TNBC cells develop drug resistance eventually. Thus, there is a need for alternate therapies. Toward this, we studied the antiproliferation characteristics of electrical pulse-mediated combination therapy using metformin, the commonly used Type-2 diabetes drug, along with cisplatin. We used metformin, as it has various anticancer properties caused by repressing energy pathways in a cancer cell. Application of 8 pulses of 1000 V/cm, 100 µs, at 1 Hz frequency, enhanced the drug uptake leading to cell viability as low as 25.86% at 30 µM cisplatin and 5 mM metformin in a 24 h study. Also, the same studies were conducted on MCF10A, a non-cancerous human epithelial cell. It aided in comparing the result for both MDA-MB-231 and MCF10A cell lines while establishing a better understanding of the experimental outcomes. Overall, the various experimental results from colony-forming assay, reactive oxidative analysis, and the intracellular glucose metabolic assay indicate the possibility of the electrical pulses-based cisplatin and metformin drug combination as a potential alternative to TNBC treatment.
Collapse
|
13
|
Sun X, Luo H, Han C, Zhang Y, Yan C. Identification of a Hypoxia-Related Molecular Classification and Hypoxic Tumor Microenvironment Signature for Predicting the Prognosis of Patients with Triple-Negative Breast Cancer. Front Oncol 2021; 11:700062. [PMID: 34490098 PMCID: PMC8416750 DOI: 10.3389/fonc.2021.700062] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/31/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose The hypoxic tumor microenvironment was reported to be involved in different tumorigenesis mechanisms of triple-negative breast cancer (TNBC), such as invasion, immune evasion, chemoresistance, and metastasis. However, a systematic analysis of the prognostic prediction models based on multiple hypoxia-related genes (HRGs) has not been established in TNBC before in the literature. We aimed to develop and verify a hypoxia gene signature for prognostic prediction in TNBC patients. Methods The RNA sequencing profiles and clinical data of TNBC patients were generated from the TCGA, GSE103091, and METABRIC databases. The TNBC-specific differential HRGs (dHRGs) were obtained from differential expression analysis of hypoxia cultured TNBC cell lines compared with normoxic cell lines from the GEO database. Non-negative matrix factorization (NMF) method was then performed on the TNBC patients using the dHRGs to explore a novel molecular classification on the basis of the dHRG expression patterns. Prognosis-associated dHRGs were identified by univariate and multivariate Cox regression analysis to establish the prognostic risk score model. Results Based on the expressions of 205 dHRGs, all the patients in the TCGA training cohort were categorized into two subgroups, and the patients in Cluster 1 demonstrated worse OS than those in Cluster 2, which was validated in two independent cohorts. Additionally, the effects of somatic copy number variation (SCNV), somatic single nucleotide variation (SSNV), and methylation level on the expressions of dHRGs were also analyzed. Then, we performed Cox regression analyses to construct an HRG-based risk score model (3-gene dHRG signature), which could reliably discriminate the overall survival (OS) of high-risk and low-risk patients in TCGA, GSE103091, METABRIC, and BMCHH (qRT-PCR) cohorts. Conclusions In this study, a robust predictive signature was developed for patients with TNBC, indicating that the 3-gene dHRG model might serve as a potential prognostic biomarker for TNBC.
Collapse
Affiliation(s)
- Xiaoli Sun
- Department of Medical Oncology, Baoji Maternal and Child Health Hospital, Baoji, China
| | - Huan Luo
- Department of Breast Surgery, Baoji Maternal and Child Health Hospital, Baoji, China
| | - Chenbo Han
- Department of Breast Surgery, Baoji Maternal and Child Health Hospital, Baoji, China
| | - Yu Zhang
- Department of Breast Surgery, Baoji Maternal and Child Health Hospital, Baoji, China
| | - Cunli Yan
- Department of Breast Surgery, Baoji Maternal and Child Health Hospital, Baoji, China.,Department of General Surgery, Baoji Maternal and Child Health Hospital, Baoji, China
| |
Collapse
|
14
|
Condello M, D’Avack G, Vona R, Spugnini EP, Scacco L, Meschini S. Electrochemotherapy with Mitomycin C Potentiates Apoptosis Death by Inhibiting Autophagy in Squamous Carcinoma Cells. Cancers (Basel) 2021; 13:3867. [PMID: 34359775 PMCID: PMC8345561 DOI: 10.3390/cancers13153867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 11/16/2022] Open
Abstract
We investigated the chemosensitizing effect of electroporation (EP), which, using electrical pulses, permeabilizes cancer cells to drugs. The study involved two human hypopharyngeal and tongue carcinoma cell lines. The surface and intracytoplasmic expression of P-gp were evaluated by flow cytometry, demonstrating that both lines were intrinsically resistant. After establishing the optimal dose of mitomycin C (MMC) to be used, in combination with EP, we showed, by both MTT assay and optical and electron scanning microscopy, the potentiating cytotoxic effect of EP with MMC compared to single treatments. Flow cytometry showed that the cytotoxicity of EP + MMC was due to the induction of apoptosis. In addition to verifying the release of cytochrome C in EP + MMC samples, we performed an expression analysis of caspase-3, caspase-9, Akt, pAkt, HMGB1, LC3I, LC3II, p62, Beclin1, and associated proteins with both apoptotic and autophagic phenomena. Our results were confirmed by two veterinary patients in whom the EP + MMC combination was used to control margins after the resection of corneal squamous carcinoma. In conclusion, we affirmed that the effect for which EP enhances MMC treatment is due to the inhibition of the autophagic process induced by the drug in favor of apoptosis.
Collapse
Affiliation(s)
- Maria Condello
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy; (M.C.); (G.D.)
| | - Gloria D’Avack
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy; (M.C.); (G.D.)
| | - Rosa Vona
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy;
| | | | | | - Stefania Meschini
- National Center for Drug Research and Evaluation, National Institute of Health, 00161 Rome, Italy; (M.C.); (G.D.)
| |
Collapse
|
15
|
Deepak Singh D, Han I, Choi EH, Yadav DK. CRISPR/Cas9 based genome editing for targeted transcriptional control in triple-negative breast cancer. Comput Struct Biotechnol J 2021; 19:2384-2397. [PMID: 34025931 PMCID: PMC8120801 DOI: 10.1016/j.csbj.2021.04.036] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer (BC) is the most common type of cancer in women at the global level and the highest mortality rate has been observed with triple-negative breast cancer (TNBC). Accumulation of genetic lesions an aberrant gene expression and protein degradation are considered to underlie the onset of tumorigenesis and metastasis. Therefore, the challenge to identify the genes and molecules that could be potentially used as potent biomarkers for personalized medicine against TNBC with minimal or no associated side effects. Discovery of the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9) arrangement and an increasing repertoire of its new variants has provided a much-needed fillip towards editing TNBC genomes. In this review, we discuss the CRISPR/Cas9 genome editing, CRISPR Technology for diagnosis of (Triple-negative breast cancer) TNBC, Drug Resistance, and potential applications of CRISPR/Cas9 and its variants in deciphering or engineering intricate molecular and epigenetic mechanisms associated with TNBC. Furthermore, we have also explored the TNBC and CRISPR/Cas9 genome editing potential for repairing, genetic modifications in TNBC.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University, Seoul, Republic of Korea
| | - Eun-Ha Choi
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University, Seoul, Republic of Korea
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro 191, Yeonsu-gu, Incheon City, Republic of Korea
| |
Collapse
|
16
|
Electrochemotherapy Modulates Mammary Tumor Growth in Rats on a Western Diet Supplemented with Curcumin. Biomedicines 2020; 8:biomedicines8110498. [PMID: 33202747 PMCID: PMC7697715 DOI: 10.3390/biomedicines8110498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/18/2022] Open
Abstract
In the US, every 12 min, six women are diagnosed with breast cancer and one dies. This highlights a critical need for developing alternate therapies using natural compounds, which are cost effective and with less side effects. Curcumin, the yellow pigment of turmeric has been found to suppress initiation, progression, and metastasis of a variety of tumors. Multiple clinical trials highlight the efficacy of curcumin in treating breast cancer and other diseases. Our in vitro studies have demonstrated that the electrical pulse (EP) application can further enhance the effectiveness of curcumin against breast cancer cells in a therapy called electrochemotherapy (ECT). In a direct extension of these results, we studied the effect of ECT coupled with intratumoral curcumin administration (EP+Cur) on N-methyl-N-nitrosourea (MNU) induced mammary tumors in female Sprague Dawley rats. Beginning at the weaning and throughout the study, rats were fed either western diet (West) or western diet, supplemented with 1% curcumin (W+Cur). Our results showed that EP+Cur treatment led to a reduced growth rate in rats fed with W+Cur diet compared to West diet (57.14% vs. 16.67% in West diet). These results provide a foundation for further studies towards utilizing it in clinical practice.
Collapse
|
17
|
Mittal L, Camarillo IG, Varadarajan GS, Srinivasan H, Aryal UK, Sundararajan R. High-throughput, Label-Free Quantitative Proteomic Studies of the Anticancer Effects of Electrical Pulses with Turmeric Silver Nanoparticles: an in vitro Model Study. Sci Rep 2020; 10:7258. [PMID: 32350346 PMCID: PMC7190727 DOI: 10.1038/s41598-020-64128-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022] Open
Abstract
Triple negative breast cancer (TNBC) represents 15–20% of the over one million new breast cancer cases occurring each year. TNBC is an aggressive cancer phenotype, with low 5-year survival rates, high 3-year recurrence rates, and increased risk of metastasis. A lack of three commonly exploited hormone receptors renders TNBC resistant to endocrine therapies and lends to its critical absence of viable therapeutic targets. This necessitates the development of alternate and effective novel therapeutic strategies for TNBC. Towards this, our current work seeks to develop the technique of Electrical pulse (EP)-mediated Turmeric silver nanoparticles (TurNP) therapy, known as Electrochemotherapy (ECT), to effectively target TNBC cells. This technique involves the efficient delivery of natural bioactive molecules with anti-cancer effects via a biophysical means. In these experiments, the bioactive molecules are turmeric, a dried rhizome of Curcuma longa that has been used for centuries, both as a dietary supplement and as a medicine in Ayurveda (science of life) in the Indian subcontinent and in traditional Chinese medicine. Our results reveal the combined effect of TurNP + EP treatment in reducing MDA-MB-231 cell viability to as low as 9% at 12 h. Showing biological selectivity, this combination treatment has a substantially lower effect on non-tumorigenic mammary epithelial MCF10A cells (67% viability). To gain mechanistic insights into the actions of TurNP-based ECT treatment, we performed high-throughput, label-free quantitative proteomics studies. Proteomics results indicate that TurNP + EP treatment significantly influenced expression of a diverse list of proteins, including receptors, transcription factors, structural proteins, kinases, and metabolic enzymes. This include the downregulation of 25 proteins in PI3K-Akt signaling pathway (such as GRB2, EGFR, EPHA2, GNB1, GNB2, 14–3–3 family, and Integrin family proteins), and 12 proteins (AKR1A1, ALDOA, ALDOC, PGK1, PGM1, PGAM1, ENO1, ENO2, GAPDH, TPI1, LDHA, and LDHB) in the glycolytic pathway with concomitant reduction in metabolite levels (glucose uptake, and intracellular- lactate, glutamine, and glutamate). Compared to TurNP alone, TurNP + EP treatment upregulated 66 endoplasmic reticulum and 193 mitochondrial proteins, enhancing several processes and pathways, including Pyruvate Metabolism, Tricarboxylic acid (TCA) cycle, and Oxidative Phosphorylation (OXPHOS), which redirected the TNBC metabolism to mitochondria. This switch in the metabolism caused excessive production of H2O2 reactive oxygen species (ROS) to inflict cell death in MDA-MB-231 cells, demonstrating the potency of this treatment.
Collapse
Affiliation(s)
- Lakshya Mittal
- School of Engineering Technology, Purdue University, West Lafayette, IN, 47907, USA
| | - Ignacio G Camarillo
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA.,Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Gowri Sree Varadarajan
- Division of High Voltage Engineering, Dept. of Electrical & Electronics Engineering, College of Engineering, Anna University, Guindy, Chennai, TN, 600025, India
| | - Hemalatha Srinivasan
- School of Life Sciences, B. S. Abdur Rahman Crescent Institute of Science & Technology, Chennai, TN, 600048, India
| | - Uma K Aryal
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA.,Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Raji Sundararajan
- School of Engineering Technology, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
18
|
Chan H, Bhide KP, Vaidyam A, Hedrick V, Sobreira TJP, Sors TG, Grant RW, Aryal UK. Proteomic Analysis of 3T3-L1 Adipocytes Treated with Insulin and TNF-α. Proteomes 2019; 7:35. [PMID: 31635166 PMCID: PMC6958341 DOI: 10.3390/proteomes7040035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/19/2022] Open
Abstract
Insulin resistance is an indication of early stage Type 2 diabetes (T2D). Insulin resistant adipose tissues contain higher levels of insulin than the physiological level, as well as higher amounts of intracellular tumor necrosis factor-α (TNF-α) and other cytokines. However, the mechanism of insulin resistance remains poorly understood. To better understand the roles played by insulin and TNF-α in insulin resistance, we performed proteomic analysis of differentiated 3T3-L1 adipocytes treated with insulin (Ins), TNF-α (TNF), and both (Ins + TNF). Out of the 693 proteins identified, the abundances of 78 proteins were significantly different (p < 0.05). Carnitine parmitoyltransferase-2 (CPT2), acetyl CoA carboxylase 1 (ACCAC-1), ethylmalonyl CoA decarboxylase (ECHD1), and methylmalonyl CoA isomerase (MCEE), enzymes required for fatty acid β-oxidation and respiratory electron transport, and β-glucuronidase, an enzyme responsible for the breakdown of complex carbohydrates, were down-regulated in all the treatment groups, compared to the control group. In contrast, superoxide dismutase 2 (SOD2), protein disulfide isomerase (PDI), and glutathione reductase, which are the proteins responsible for cytoskeletal structure, protein folding, degradation, and oxidative stress responses, were up-regulated. This suggests higher oxidative stress in cells treated with Ins, TNF, or both. We proposed a conceptual metabolic pathway impacted by the treatments and their possible link to insulin resistance or T2D.
Collapse
Affiliation(s)
- Hayley Chan
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Ketaki P Bhide
- College of Agriculture, Purdue University, West Lafayette, IN 47907, USA.
| | - Aditya Vaidyam
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Victoria Hedrick
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.
| | | | - Thomas G Sors
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.
| | - Ryan W Grant
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Uma K Aryal
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA.
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|