1
|
Das U, Kundu J, Shaw P, Bose C, Ghosh A, Gupta S, Sarkar S, Bhadra J, Sinha S. Self-transfecting GMO-PMO chimera targeting Nanog enable gene silencing in vitro and suppresses tumor growth in 4T1 allografts in mouse. MOLECULAR THERAPY - NUCLEIC ACIDS 2023; 32:203-228. [PMID: 37078062 PMCID: PMC10106836 DOI: 10.1016/j.omtn.2023.03.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 03/16/2023] [Indexed: 04/05/2023]
Abstract
Phosphorodiamidate morpholino oligonucleotide (PMO)-based antisense reagents cannot enter cells without the help of a delivery technique, which limits their clinical applications. To overcome this problem, self-transfecting guanidinium-linked morpholino (GMO)-PMO or PMO-GMO chimeras have been explored as antisense agents. GMO facilitates cellular internalization and participates in Watson-Crick base pairing. Targeting NANOG in MCF7 cells resulted in decline of the whole epithelial to mesenchymal transition (EMT) and stemness pathway, evident through its phenotypic manifestations, all of which were promulgated in combination with Taxol due to downregulation of MDR1 and ABCG2. GMO-PMO-mediated knockdown of no tail gene resulted in desired phenotypes in zebrafish even upon delivery after 16-cell stages. In BALB/c mice, 4T1 allografts were found to regress via intra-tumoral administration of NANOG GMO-PMO antisense oligonucleotides (ASOs), which was associated with occurrence of necrotic regions. GMO-PMO-mediated tumor regression restored histopathological damage in liver, kidney, and spleen caused by 4T1 mammary carcinoma. Serum parameters of systemic toxicity indicated that GMO-PMO chimeras are safe. To the best of our knowledge, self-transfecting antisense reagent is the first report since the discovery of guanidinium-linked DNA (DNG), which could be useful as a combination cancer therapy and, in principle, can render inhibition of any target gene without using any delivery vehicle.
Collapse
|
2
|
Ortega-Rivera OA, Gallegos-Alcalá P, Jiménez M, Quintanar JL, Torres-Juarez F, Rivas-Santiago B, del Toro-Arreola S, Salinas E. Inhibition of Tumor Growth and Metastasis by Newcastle Disease Virus Strain P05 in a Breast Cancer Mouse Model. J Breast Cancer 2023; 26:186-200. [PMID: 37051644 PMCID: PMC10139849 DOI: 10.4048/jbc.2023.26.e9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/16/2022] [Accepted: 01/24/2023] [Indexed: 02/25/2023] Open
Abstract
PURPOSE Conventional therapies and surgery remain the standard treatment for breast cancer. However, combating the eventual development of metastasis is still a challenge. Newcastle disease virus (NDV) is one of the various species of viruses under clinical evaluation as a vector for oncolytic, gene-, and immune-stimulating therapies. The purpose of this study was to evaluate the antitumor activity of a recombinant NDV (rNDV-P05) in a breast cancer murine model. METHODS Tumors were induced by injecting the cellular suspension (4T1 cell line) subcutaneously. The virus strain P05 was applied three times at intervals of seven days, starting seven days after tumor induction, and was completed 21 days later. Determination of tumor weight, spleen index, and lung metastasis were done after sacrificing the mice. Serum levels of interferon (IFN)-α, IFN-γ, tumor necrosis factor (TNF)-α, and TNF-related apoptosis-inducing ligand (TRAIL) were quantified by enzyme-linked immunosorbent assay. CD8+ infiltrated cells were analyzed by immunofluorescence. RESULTS rNDV-P05 showed a route-of-administration-dependent effect, demonstrating that the systemic administration of the virus significantly reduces the tumor mass and volume, spleen index, and abundance of metastatic clonogenic colonies in lung tissue, and increases the inhibition rate of the tumor. The intratumoral administration of rNDV-P05 was ineffective for all the parameters evaluated. Antitumor and antimetastatic capability of rNDV-P05 is mediated, at least partially, through its immune-stimulatory effect on the upregulation of TNF-α, TRAIL, IFN-α, and IFN-γ, and its ability to recruit CD8+ T cells into tumor tissue. CONCLUSION Systemic treatment with rNDV-P05 decreases the tumoral parameters in the breast cancer murine model.
Collapse
Affiliation(s)
- Oscar Antonio Ortega-Rivera
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes, Mexico
- Department of NanoEngineering, University of California San Diego, La Jolla, USA
| | - Pamela Gallegos-Alcalá
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes, Mexico
| | - Mariela Jiménez
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes, Mexico
| | - J. Luis Quintanar
- Department of Physiology and Pharmacology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes, Mexico
| | - Flor Torres-Juarez
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security (IMSS), Zacatecas, Mexico
| | - Bruno Rivas-Santiago
- Medical Research Unit-Zacatecas, Mexican Institute for Social Security (IMSS), Zacatecas, Mexico
| | - Susana del Toro-Arreola
- Department of Physiology, CUCS, University of Guadalajara, Guadalajara, Mexico
- Institute of Research in Chronic Degenerative Diseases, Department of Molecular Biology and Genomic, CUCS, University of Guadalajara, Guadalajara, Mexico
| | - Eva Salinas
- Department of Microbiology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
3
|
Muhammed E, Erenso DB, Gao Y, Chen L, Kelley M, Vazquez C, Gale M, Nichols C, Crogman HT. Measurement of Charge and Refractive Indices in Optically Trapped and Ionized Living Cells. Tomography 2022; 9:70-88. [PMID: 36648994 PMCID: PMC9844322 DOI: 10.3390/tomography9010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
The post-ionization dynamics of chemo-treated and untreated 4T1 breast cancer cells ionized by laser trapping techniques are studied. We have determined each cell's charge and refractive index by developing a theoretical model for the forces determining the post-ionization dynamics. The shift in a cell's refractive index due to an intense oscillating electric field was studied, and the results are reported here. We observed that a trapped cell, as it becomes charged, will eventually exit the trap perpendicular to the beam's direction; this means that the electric force of the cell overcomes the trapping force. As a result, the cell's conductivity changes due to the oscillating field, causing a decrease in the cell's refractive index.
Collapse
Affiliation(s)
- Endris Muhammed
- Department of Physics, Addis Ababa University, Addis Ababa 1176, Ethiopia
| | - Daniel B. Erenso
- Department of Physics, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Ying Gao
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Li Chen
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Michele Kelley
- Department of Physics, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Carina Vazquez
- Department of Physics, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Mitchell Gale
- Department of Physics, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Cody Nichols
- Department of Physics, California State University Dominguez Hills, Carson, CA 90747, USA
| | - Horace T. Crogman
- Department of Physics, California State University Dominguez Hills, Carson, CA 90747, USA
- Correspondence:
| |
Collapse
|
4
|
Mimansa, Jamwal M, Das R, Shanavas A. High Drug Loading Nanoparticles Stabilized with Autologous Serum Proteins Passively Inhibits Tumor Growth. Biomacromolecules 2022; 23:5065-5073. [PMID: 36218374 DOI: 10.1021/acs.biomac.2c00907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We report drug nanocrystals stabilized with host-specific serum proteins with high loading (∼63% w/w). The human serum derived curcumin nanoparticles (Cur-NanoSera) showed superior in vitro anticancer efficiency compared to a free drug with substantial hemocompatibility. The preadsorbed protein coating impeded further protein corona formation, even with repeated serum exposures. Acute and subacute toxicity evaluations post single and dual injections of C57BL/6 mice indicated that Cur-NanoSera showed no prominent inflammatory response or organ damage in the in-bred mice. Passive accumulation of Cur-NanoSera in tumor tissue significantly suppressed its growth in a syngeneic breast tumor model in addition to controlling tumor burden associated splenomegaly.
Collapse
Affiliation(s)
- Mimansa
- Institute of Nano Science and Technology, Sector 81, Mohali, 140306, India
| | - Manu Jamwal
- Department of Haematology, Post Graduate Institute of Medical Education and Research (PGIMER), Madhya Marg, Sector 12, Chandigarh, 160012, India
| | - Reena Das
- Department of Haematology, Post Graduate Institute of Medical Education and Research (PGIMER), Madhya Marg, Sector 12, Chandigarh, 160012, India
| | - Asifkhan Shanavas
- Institute of Nano Science and Technology, Sector 81, Mohali, 140306, India
| |
Collapse
|
5
|
Zhang B, Zhang Y, Dang W, Xing B, Yu C, Guo P, Pi J, Deng X, Qi D, Liu Z. The anti-tumor and renoprotection study of E-[c(RGDfK)2]/folic acid co-modified nanostructured lipid carrier loaded with doxorubicin hydrochloride/salvianolic acid A. J Nanobiotechnology 2022; 20:425. [PMID: 36153589 PMCID: PMC9509648 DOI: 10.1186/s12951-022-01628-x] [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: 03/31/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Poor in vivo targeting of tumors by chemotherapeutic drugs reduces their anti-cancer efficacy in the clinic. The discovery of over-expressed components on the tumor cell surface and their specific ligands provide a basis for targeting tumor cells. However, the differences in the expression levels of these receptors on the tumor cell surface limit the clinical application of anti-tumor preparations modified by a single ligand. Meanwhile, toxicity of chemotherapeutic drugs leads to poor tolerance to anti-tumor therapy. The discovery of natural active products antagonizing these toxic side effects offers an avenue for relieving cancer patients’ pain during the treatment process. Since the advent of nanotechnology, interventions, such as loading appropriate drug combinations into nano-sized carriers and multiple tumor-targeting functional modifications on the carrier surface to enhance the anti-tumor effect and reduce toxic and side effects, have been widely used for treating tumors.
Results
Nanocarriers containing doxorubicin hydrochloride (DOX) and salvianolic acid A (Sal A) are spherical with a diameter of about 18 nm; the encapsulation efficiency of both DOX and salvianolic acid A is greater than 80%. E-[c(RGDfK)2]/folic acid (FA) co-modification enabled nanostructured lipid carriers (NLC) to efficiently target a variety of tumor cells, including 4T1, MDA-MB-231, MCF-7, and A549 cells in vitro. Compared with other preparations (Sal A solution, NLC-Sal A, DOX solution, DOX injection, Sal A/DOX solution, NLC-DOX, NLC-Sal A/DOX, and E-[c(RGDfK)2]/FA-NLC-Sal A/DOX) in this experiment, the prepared E-[c(RGDfK)2]/FA-NLC-Sal A/DOX had the best anti-tumor effect. Compared with the normal saline group, it had the highest tumor volume inhibition rate (90.72%), the highest tumor weight inhibition rate (83.94%), led to the highest proportion of apoptosis among the tumor cells (61.30%) and the lowest fluorescence intensity of proliferation among the tumor cells (0.0083 ± 0.0011). Moreover, E-[c(RGDfK)2]/FA-NLC-Sal A/DOX had a low level of nephrotoxicity, with a low creatinine (Cre) concentration of 52.58 μmoL/L in the blood of mice, and no abnormalities were seen on pathological examination of the isolated kidneys at the end of the study. Sal A can antagonize the nephrotoxic effect of DOX. Free Sal A reduced the Cre concentration of the free DOX group by 61.64%. In NLC groups, Sal A reduced the Cre concentration of the DOX group by 42.47%. The E-[c(RGDfK)2]/FA modification reduced the side effects of the drug on the kidney, and the Cre concentration was reduced by 46.35% compared with the NLC-Sal A/DOX group. These interventions can potentially improve the tolerance of cancer patients to chemotherapy.
Conclusion
The E-[c(RGDfK)2]/FA co-modified DOX/Sal A multifunctional nano-drug delivery system has a good therapeutic effect on tumors and low nephrotoxicity and is a promising anti-cancer strategy.
Graphical Abstract
Collapse
|
6
|
|
7
|
Luan F, Peng L, Lei Z, Jia X, Zou J, Yang Y, He X, Zeng N. Traditional Uses, Phytochemical Constituents and Pharmacological Properties of Averrhoa carambola L.: A Review. Front Pharmacol 2021; 12:699899. [PMID: 34475822 PMCID: PMC8407000 DOI: 10.3389/fphar.2021.699899] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/02/2021] [Indexed: 11/25/2022] Open
Abstract
Averrhoa carambola L. (star fruit) is an edible fruit that is extensively cultivated in southern China, Southeast Asia, India, and northern South America. It has a sweet and juicy taste and is frequently used in fruit salads and fruit platters, as a garnish in cocktail drinks and beverages, or squeezed into juice and served as a beverage. Traditionally, it has been used for treating diabetes and diabetic nephropathy, arthralgia, vomiting, lithangiuria, coughing, hangovers, and chronic paroxysmal headache for thousands of years. Currently, approximately 132 compounds have been isolated from A. carambola. Among them, flavonoids, benzoquinone, and their glycosides have been considered as biologically active substances, which are responsible for various biological activities. Pharmacological studies have revealed that crude extracts or monomeric compounds from A. carambola exhibit multiple bioactivities, such as anti-oxidant, anti-hyperglycemic, anti-obesity, anti-hyperlipidemic, anti-tumor, anti-inflammatory, hepatoprotective, cardioprotective, anti-hypertensive, neuroprotective, and others. Thus, A. carambola is a valuable treatment in Chinese medicine with therapeutic potential for multiple diseases, especially diabetes and diabetes-related diseases. Even though it is a very promising candidate in the development of functional food and the pharmaceutical industry, reports on its bioactivities have only been conducted in vivo and in vitro and there is a gap in research regarding clinical settings and safety. This review therefore provides a comprehensive and systematic overview of current progress on botany, ethnopharmacology, phytochemistry, pharmacology, and toxicity of A. carambola, providing a valuable reference for further developments and applications of A. carambola in the pharmaceutical industry and functional food.
Collapse
Affiliation(s)
- Fei Luan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lixia Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziqin Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiyu Jia
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junbo Zou
- Department of Pharmacology, College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yan Yang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Xirui He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Mokriani S, Tukmechi A, Harzandi N, Jabalameli L. In vivo murine breast cancer targeting by magnetic iron nanoparticles involving L. GG cytoplasmic fraction. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:682-689. [PMID: 34249271 PMCID: PMC8244610 DOI: 10.22038/ijbms.2021.54961.12322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/19/2021] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Use of chemical anti-cancer drugs frequently creates serious side effects. However, probiotics are natural and treat different kinds of cancer without undesired effects. MATERIALS AND METHODS In this study, a nano delivery system was planned to transport the Lactobacillus rhamnosus GG (L. GG) cytoplasmic fraction (Cf) to cancerous tissue in a mouse model. Magnetic iron nanoparticles (MINPs) were synthesized and loaded with L. GG-Cf(0, 0.312, 0.625, 1.25, and 2.5 mg/ml) and were administrated for three weeks to treat experimentally induced murine breast cancer in a constant magnetic field. At the end of the trial, the treating efficacy of this complex molecule was evaluated via western blotting, immunohistochemistry, and qPCR. RESULTS Results showed that MINPS can deliver and accumulate L. GG-Cf in cancer tissue, and reduce the size and volume of the tumors. Additionally, in cancer tissues of treated mice with 2.5 mg/ml of Cf-MINPs, significantly induced apoptosis was seen compared with untreated mice (control), and our data proved that this induction may be due to the caspase-3 pathway. CONCLUSION L. GG-Cf could treat murine breast cancer, and MINPs are a suitable candidate for drug delivery because of their safety, uniformity, and magnetic properties.
Collapse
Affiliation(s)
- Salar Mokriani
- Department of Microbiology, Faculty of Sciences, Karaj Branch, Islamic Azad University, Karaj, Alborz Province, Iran
| | - Amir Tukmechi
- Department of Microbiology, Faculty of Veterinary Medicine, Urmia University, Urmia, West Azarbaijan Province, Iran
| | - Naser Harzandi
- Department of Microbiology, Faculty of Veterinary Medicine, Karaj Branch, Islamic Azad University, Karaj, Alborz province, Iran
| | - Leila Jabalameli
- Department of Microbiology, Faculty of Sciences, Karaj Branch, Islamic Azad University, Karaj, Alborz Province, Iran
| |
Collapse
|
9
|
Wang L, Yang X, Song Q, Fu J, Wang W, Du K, Chen S, Cao J, Huang R, Zou C. Uncovering the Pharmacological Mechanism of 2-Dodecyl-6-Methoxycyclohexa-2,5 -Diene-1,4-Dione Against Lung Cancer Based on Network Pharmacology and Experimental Evaluation. Front Pharmacol 2021; 12:617555. [PMID: 33613291 PMCID: PMC7887632 DOI: 10.3389/fphar.2021.617555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/06/2021] [Indexed: 11/16/2022] Open
Abstract
Background: 2-Dodecyl-6-Methoxycyclohexa-2, 5-Diene-1,4-Dione (DMDD) was purified from the roots of Averrhoa carambola L. Previous research demonstrated that DMDD is a small molecular compound with significant therapeutic potential for tumors. However, the potential targets and pharmacological mechanism of DMDD to treat lung cancer has not been reported. Methods: We employed network pharmacology and experimental evaluation to reveal the pharmacological mechanism of DMDD against lung cancer. Potential therapeutic targets of DMDD were screened by PharmMapper. Differentially expressed genes (DEGs) in The Cancer Genome Atlas (TCGA) lung cancer data sets were extracted and analyzed by GEPIA2. The mechanism of DMDD against lung cancer was determined by PPI, gene ontology (GO) and KEGG pathway enrichment analysis. Survival analysis and molecular docking were employed to obtain the key targets of DMDD. Human lung cancer cell lines H1975 and PC9 were used to detect effects of DMDD treatment in vitro. The expression of key targets after DMDD treated was validated by Western Blot. Results: A total of 60 Homo sapiens potential therapeutic targets of DMDD and 3,545 DEGs in TCGA lung cancer datasets were identified. Gene ontology and pathway analysis revealed characteristic of the potential targets of DMDD and DEGs in lung cancer respectively. Cell cycle and pathways in cancer were overlapping with DMDD potential targets and lung cancer DEGs. Eight overlapping genes were found between DMDD potential therapeutic targets and lung cancer related DEGs. Survival analysis showed that high expression of DMDD potential targets CCNE1 and E2F1 was significantly related to poor patient survival in lung cancer. Molecular docking found that DMDD exhibited significant binding affinities within the active site of CCNE1 and E2F1. Further tests showed that DMDD inhibited the proliferation, migration and clone formation in lung cancer cell lines (H1975 and PC9) in a dose and time dependent manner. Mechanistically, DMDD treatment decreased the expression of CDK2, CCNE1, E2F1 proteins and induced cell cycle arrest at the G1/S phase in H1975 and PC9 cells. Conclusion: These results delineated that DMDD holds therapeutic potential that blocks tumorigenesis by cell cycle regulation in lung cancer, and may provide potential therapies for lung cancer.
Collapse
Affiliation(s)
- Lihui Wang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China.,Department of Pharmacology, Guangxi Medical University, Nanning, China
| | - Xin Yang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Qiong Song
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Jiejun Fu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Wenchu Wang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Kechen Du
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Shuai Chen
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Jinjin Cao
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Renbin Huang
- Department of Pharmacology, Guangxi Medical University, Nanning, China
| | - Chunlin Zou
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Center for Translational Medicine and School of Preclinical Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
10
|
Nagai K, Sato T, Kojima C. Design of a dendrimer with a matrix metalloproteinase-responsive fluorescence probe and a tumor-homing peptide for metastatic tumor cell imaging in the lymph node. Bioorg Med Chem Lett 2021; 33:127726. [PMID: 33316406 DOI: 10.1016/j.bmcl.2020.127726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/18/2020] [Accepted: 11/27/2020] [Indexed: 02/01/2023]
Abstract
Fluorescence imaging is a noninvasive technique for cancer diagnosis. Dendrimers are regularly branched macromolecules with highly controllable size and structure that are a potent multifunctional nanoparticle. Anionic-terminal polyamidoamine (PAMAM) dendrimers were previously found to be accumulated in the lymph node, which is one of the main routes of tumor metastasis. In this study, we designed and synthesized a dendrimeric imaging probe for lymph node-resident tumor cell imaging. A matrix metalloproteinase-2 (MMP-2)-responsive fluorescence peptide probe and a tumor-homing peptide were conjugated to the carboxy-terminal dendrimer. The dendrimeric imaging probe treatment showed fluorescence signals inside some tumor cells (e.g., human fibrosarcoma HT-1080 and breast cancer 4T1 cells), depending on the MMP activity, but not in macrophage-like RAW264 cells.
Collapse
Affiliation(s)
- Kento Nagai
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan
| | - Tatsumi Sato
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan
| | - Chie Kojima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-2 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan.
| |
Collapse
|
11
|
Tasdemir SS, Sanlier N. An insight into the anticancer effects of fermented foods: A review. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
12
|
Alhudaithi SS, Almuqbil RM, Zhang H, Bielski ER, Du W, Sunbul FS, Bos PD, da Rocha SRP. Local Targeting of Lung-Tumor-Associated Macrophages with Pulmonary Delivery of a CSF-1R Inhibitor for the Treatment of Breast Cancer Lung Metastases. Mol Pharm 2020; 17:4691-4703. [PMID: 33170724 DOI: 10.1021/acs.molpharmaceut.0c00983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The lungs are major sites of metastases for several cancer types, including breast cancer (BC). Prognosis and quality of life of BC patients that develop pulmonary metastases are negatively impacted. The development of strategies to slow the growth and relieve the symptoms of BC lung metastases (BCLM) is thus an important goal in the management of BC. However, systemically administered first line small molecule chemotherapeutics have poor pharmacokinetic profiles and biodistribution to the lungs and significant off-target toxicity, severely compromising their effectiveness. In this work, we propose the local delivery of add-on immunotherapy to the lungs to support first line chemotherapy treatment of advanced BC. In a syngeneic murine model of BCLM, we show that local pulmonary administration (p.a.) of PLX-3397 (PLX), a colony-stimulating factor 1 receptor inhibitor (CSF-1Ri), is capable of overcoming physiological barriers of the lung epithelium, penetrating the tumor microenvironment (TME), and decreasing phosphorylation of CSF-1 receptors, as shown by the Western blot of lung tumor nodules. That inhibition is accompanied by an overall decrease in the abundance of protumorigenic (M2-like) macrophages in the TME, with a concomitant increase in the amount of antitumor (M1-like) macrophages when compared to the vehicle-treated control. These effects with PLX (p.a.) were achieved using a much smaller dose (1 mg/kg, every other day) compared to the systemic doses typically used in preclinical studies (40-800 mg/kg/day). As an additive in combination with intravenous (i.v.) administration of paclitaxel (PTX), PLX (p.a.) leads to a decrease in tumor burden without additional toxicity. These results suggested that the proposed immunochemotherapy, with regional pulmonary delivery of PLX along with the i.v. standard of care chemotherapy, may lead to new opportunities to improve treatment, quality of life, and survival of patients with BCLM.
Collapse
Affiliation(s)
- Sulaiman S Alhudaithi
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Rashed M Almuqbil
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Hanming Zhang
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Elizabeth R Bielski
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Wei Du
- Department of Pathology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Fatemah S Sunbul
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Paula D Bos
- Department of Pathology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States.,VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Sandro R P da Rocha
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298, United States.,VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
13
|
Tan X, Chen W, Jiao C, Liang H, Yun H, He C, Chen J, Ma X, Xie Y. Anti-tumor and immunomodulatory activity of the aqueous extract of Sarcodon imbricatus in vitro and in vivo. Food Funct 2020; 11:1110-1121. [PMID: 31825431 DOI: 10.1039/c9fo01230c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sarcodon imbricatus (S. imbricatus), a well-known edible mushroom, is one of the most commonly consumed wild mushrooms in China because of its nutritional value. Previous studies have demonstrated that S. imbricatus has immunoregulatory activity. We previously described the potential anti-tumor activity of several types of mushrooms, including S. imbricatus. In this study, the results demonstrate that an aqueous extract of S. imbricatus (SIE) effectively inhibits the growth, migration, and invasion properties of breast cancer cells in vitro and reduces tumor growth in vivo. In addition, the SIE increased serum concentrations of interleukin (IL)-2, IL-6 and tumor necrosis factor-α, natural killer cell activity and the viability of splenocytes and reduced the expression of programmed cell death-Ligand 1 (PD-L1) in 4T1 tumor-bearing mice. Collectively, these results are the first demonstration that the SIE has anti-tumor and immunomodulatory effects in the 4T1 mouse breast cancer model. These findings provide a scientific rationale for the potential therapeutic use of S. imbricatus in breast cancer patients.
Collapse
Affiliation(s)
- Xupeng Tan
- Guangdong Yuewei Edible Fungi Technology Co., Ltd., Guangzhou, 510663, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Yao WZ, Veeraperumal S, Qiu HM, Chen XQ, Cheong KL. Anti-cancer effects of Porphyra haitanensis polysaccharides on human colon cancer cells via cell cycle arrest and apoptosis without causing adverse effects in vitro. 3 Biotech 2020; 10:386. [PMID: 32832336 PMCID: PMC7419411 DOI: 10.1007/s13205-020-02379-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
In this study, the anticancer effects of Porphyra haitanensis polysaccharides (PHPs) on human colon cancer cells and non-cancerous cells were evaluated. PHP was extracted by an ultrasonic/microwave-assisted method, and three fractions of polysaccharides (PHP-F1, PHP-F2 and PHP-F3) were obtained through a DEAE-52 cellulose ion-exchange column. The results of the cytotoxicity test showed that all of the PHP fractions had inhibitory effects on the growth of colon cancer cells HT-29, LoVo and SW-480, but no toxic effects on the normal human cells HaCaT. The fractions PHP-F2 and PHP-F3 had the most significant cytotoxicity on HT-29 cells. Studies on intracellular reactive oxygen species (ROS) levels, cell apoptosis, the apoptosis index (using Hoechst 33342 staining) and analysis of cell cycle arrest using flow cytometry revealed that the fractions PHP-F2 and PHP-F3 could apparently induce oxidative stress and apoptosis in HT-29 cells and cause cell G0-G1 phase arrest. These findings suggest that polysaccharides from P. haitanensis have anticancer effects on human colon cancer cells and therefore might be regarded as new candidates for the prevention and treatment of colon cancers.
Collapse
Affiliation(s)
- Wan-Zi Yao
- Guangdong Provincial Key Laboratory of Marine Biotechnology, STU-UNIVPM Joint Algal Research Center, Institute of Marine Sciences, Shantou University, Shantou, 515063 Guangdong People’s Republic of China
| | - Suresh Veeraperumal
- Guangdong Provincial Key Laboratory of Marine Biotechnology, STU-UNIVPM Joint Algal Research Center, Institute of Marine Sciences, Shantou University, Shantou, 515063 Guangdong People’s Republic of China
| | - Hua-Mai Qiu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, STU-UNIVPM Joint Algal Research Center, Institute of Marine Sciences, Shantou University, Shantou, 515063 Guangdong People’s Republic of China
| | - Xian-Qiang Chen
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200 Guangxi China
| | - Kit-Leong Cheong
- Guangdong Provincial Key Laboratory of Marine Biotechnology, STU-UNIVPM Joint Algal Research Center, Institute of Marine Sciences, Shantou University, Shantou, 515063 Guangdong People’s Republic of China
| |
Collapse
|
15
|
Zhou X, Wu X, Qin L, Lu S, Zhang H, Wei J, Chen L, Jiang L, Wu Y, Chen C, Huang R. Anti-Breast Cancer Effect of 2-Dodecyl-6-Methoxycyclohexa-2,5-Diene-1,4-Dione in vivo and in vitro Through MAPK Signaling Pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2667-2684. [PMID: 32764871 PMCID: PMC7369253 DOI: 10.2147/dddt.s237699] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/07/2020] [Indexed: 12/18/2022]
Abstract
Background 2-Dodecyl-6-methoxycyclohexa-2,5-diene-1,4-dione (DMDD) has been reported to inhibit a variety of cancer cell lines. The purpose of this study was to investigate the effects of DMDD on 4T1 breast cancer cells and the effects of DMDD on 4T1 breast cancer in mice and its molecular mechanisms. Methods 4T1 breast cancer cells were treated with different concentrations of DMDD, and their proliferation, apoptosis, cell-cycle distribution, migration, and invasion were detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT, Acridine orange and ethidium bromide dual staining analysis (AO/EB) dual staining, flow cytometry, scratch test, and the Transwell assay. Relative quantitative real-time qPCR analysis and Western blot were applied to examine the expression levels of related genes and proteins. In animal experiments, we established a xenograft model to assess the anti-breast cancer effects of DMDD by evaluating the inhibition rate. The apoptotic activity of DMDD was evaluated by hematoxylin-eosin (HE) staining, transmission electron microscope (TEM) analysis and TdT-mediated dUTP nick end labeling (TUNEL) assays. The mRNA expression levels of MAPK pathway components were detected by relative quantitative real-time qPCR. In addition, the protein expression levels of MAPK pathway components were assessed through immunohistochemical assays and Western blotting. Results Experiments showed that DMDD could inhibit the proliferation, migration, invasion of 4T1 cells and induce cellular apoptosis and G1 cell cycle arrest. Moreover, DMDD down-regulated the mRNA expressions of raf1, mek1, mek2, erk1, erk2, bcl2, and up-regulated the mRNA expression of bax. DMDD reduced the protein expressions of p-raf1, p-mek, p-erk, p-p38, Bcl2, MMP2, MMP9 and increased the protein expressions of Bax and p-JNK. The results showed that DMDD can effectively reduce the tumor volume and weight of breast cancer in vivo, up-regulate the expression of IL-2, down-regulate the expression of IL-4 and IL-10, induce the apoptosis of breast cancer cells in mice, and regulate the expression of genes and proteins of the MAPK pathway. Conclusion Our study indicates that DMDD can inhibit proliferation, migration, and invasion and induces apoptosis and cell-cycle arrest of 4T1 breast cancer cells. Also, our findings indicate that DMDD induces the apoptosis of breast cancer cells and inhibits the growth in mice. Its mechanism may be related to the MAPK pathway.
Collapse
Affiliation(s)
- Xing Zhou
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Xingchun Wu
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Luhui Qin
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Shunyu Lu
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Hongliang Zhang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Jinbin Wei
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Lixiu Chen
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Luhui Jiang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Yani Wu
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Chunxia Chen
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| | - Renbin Huang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, Guangxi, People's Republic of China
| |
Collapse
|
16
|
Wu Y, Liu J, Movahedi F, Gu W, Xu T, Xu ZP. Enhanced Prevention of Breast Tumor Metastasis by Nanoparticle-Delivered Vitamin E in Combination with Interferon-Gamma. Adv Healthc Mater 2020; 9:e1901706. [PMID: 32052565 DOI: 10.1002/adhm.201901706] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/16/2020] [Indexed: 12/15/2022]
Abstract
Preventing cancer metastasis is one of the remaining challenges in cancer therapy. As an efficient natural product, alpha-tocopheryl succinate (α-TOS), the most effective form of vitamin E, holds great anticancer potential. To improve its efficacy and bioavailability, lipid-coated calcium carbonate/phosphate (LCCP) nanoparticles (NPs) with folic acid and PEG modification are synthesized for efficient delivery of α-TOS to 4T1 cancer cells. The optimized LCCP-FA NPs (NP-TOS15) show an α-TOS loading efficiency of around 60%, and enhanced uptake by 4T1 metastatic cancer cells. Consequently, NP-TOS15 significantly enhance the anticancer effect in combination with interferon-gamma (IFN-γ) in terms of apoptosis facilitation and migration inhibition. Importantly, NP-TOS15 upregulate the anticancer immunity via downregulating program death ligand 1 (PD-L1) expression that is initially induced by IFN-γ, and remarkably prevent the lung metastasis, particularly in combination with IFN-γ. Further investigation reveals that this combination therapy also modulates the cytotoxic lymphocyte infiltration into the tumor microenvironment for tumor elimination. Taken together, the NP delivery of α-TOS in combination with IFN-γ provides an applicable strategy for cancer therapy.
Collapse
Affiliation(s)
- Yilun Wu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Jianping Liu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Fatemeh Movahedi
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| | - Tiefeng Xu
- The First Affiliated Hospital of Hainan Medical University Cancer Institute of Hainan Medical University Haikou Hainan 570102 China
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology The University of Queensland St Lucia QLD 4072 Australia
| |
Collapse
|
17
|
Muhammed E, Chen L, Gao Y, Erenso D. Chemo-treated 4T1 breast cancer cells radiation response measured by single and multiple cell ionization using infrared laser trap. Sci Rep 2019; 9:17547. [PMID: 31772194 PMCID: PMC6879764 DOI: 10.1038/s41598-019-53821-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/25/2019] [Indexed: 01/28/2023] Open
Abstract
We present a study that uses a laser trapping technique for measurement of radiation sensitivity of untreated and chemo-treated cancer cells. We used a human mammary tumor cell line (4T1) treated by an antitumor compound, 2-Dodecyl-6-methoxycyclohexa-2, 5-diene-1,4-dione (DMDD), which was extracted from the root of Averrhoa carambola L. The untreated control group, and both 2-hour and 24-hour treated groups of 4T1 cells were used in this study. The absorbed threshold ionization energy (TIE) and the threshold radiation dose (TRD) were determined using a high-power infrared laser (at 1064 nm) trap by single and multiple cells trapping and ionization. The results were analyzed using descriptive and t-statistics. The relation of the TIE and TRD to the mass of the individual cells were also analyzed for different hours of treatment in comparison with the control group. Both TIE and TRD decrease with increasing treatment periods. However, the TRD decreases with mass regardless of the treatment. Analyses of the TRD for single vs multiple cells ionizations within each group have also consistently showed this same behavior regardless of the treatment. The underlying factors for these observed relations are explained in terms of radiation, hyperthermia, and chemo effects.
Collapse
Affiliation(s)
- Endris Muhammed
- Department of Physics, Addis Ababa University, Addis Ababa, 1176, Addis Ababa, Ethiopia
| | - Li Chen
- Department of Pharmacology, College of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi, 545006, China
| | - Ying Gao
- International Ginseng Institute, School of Agriculture, Middle Tennessee State University, Murfreesboro, TN, 37132, USA
| | - Daniel Erenso
- Department of Physics, Middle Tennessee State University, Murfreesboro, TN, 37132, USA.
| |
Collapse
|
18
|
Mendez Utz VE, Perdigón G, de Moreno de LeBlanc A. Oral administration of milk fermented by Lactobacillus casei CRL431 was able to decrease metastasis from breast cancer in a murine model by modulating immune response locally in the lungs. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.01.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
19
|
de Almeida Schirmer BG, de Araujo MR, Silveira MB, Pereira JM, Vieira LC, Alves CG, Mbolela WT, Ferreira AV, Silva-Cunha A, Fialho SL, da Silva JB, Malamut C. Comparison of [ 18F]Fluorocholine and [ 18F]Fluordesoxyglucose for assessment of progression, lung metastasis detection and therapy response in murine 4T1 breast tumor model. Appl Radiat Isot 2018; 140:278-288. [PMID: 30081351 DOI: 10.1016/j.apradiso.2018.07.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/23/2018] [Accepted: 07/25/2018] [Indexed: 10/28/2022]
Abstract
The [18F]Fluorocholine ([18F]FCH) tracer for PET imaging has been proven to be effective for several malignances. However, there are only a few studies related to its breast tumor applicability and they are still limited. The aim of this study was investigate the efficacy of [18F]FCH/PET compared to [18F]FDG/PET in a murine 4T1 mammary carcinoma model treated and nontreated. [18F]FCH/PET showed its applicability for primary tumor and lung metastasis detection and their use for response monitoring of breast cancer therapeutics at earlier stages.
Collapse
Affiliation(s)
| | - Marina Rios de Araujo
- Unidade de Pesquisa e Produção de Radiofármacos, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Belo Horizonte, Brazil
| | - Marina Bicalho Silveira
- Unidade de Pesquisa e Produção de Radiofármacos, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Belo Horizonte, Brazil
| | - Jousie Michel Pereira
- Unidade de Pesquisa e Produção de Radiofármacos, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Belo Horizonte, Brazil
| | - Lorena Carla Vieira
- Faculdade de Farmácia - Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; Fundação Ezequiel Dias (FUNED), Belo Horizonte, Brazil
| | - Clarice Gregório Alves
- Unidade de Pesquisa e Produção de Radiofármacos, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Belo Horizonte, Brazil
| | - William Tshisuaka Mbolela
- Unidade de Pesquisa e Produção de Radiofármacos, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Belo Horizonte, Brazil
| | - Andrea Vidal Ferreira
- Unidade de Pesquisa e Produção de Radiofármacos, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Belo Horizonte, Brazil
| | - Armando Silva-Cunha
- Faculdade de Farmácia - Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Juliana Batista da Silva
- Unidade de Pesquisa e Produção de Radiofármacos, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Belo Horizonte, Brazil
| | - Carlos Malamut
- Unidade de Pesquisa e Produção de Radiofármacos, Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Belo Horizonte, Brazil.
| |
Collapse
|
20
|
Li X, Singh K, Luo Z, Mejia-Cordova M, Jamalpour M, Lindahl B, Zhang G, Sandler S, Welsh M. Pro-tumoral immune cell alterations in wild type and Shb-deficient mice in response to 4T1 breast carcinomas. Oncotarget 2018; 9:18720-18733. [PMID: 29721156 PMCID: PMC5922350 DOI: 10.18632/oncotarget.24643] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 02/21/2018] [Indexed: 01/21/2023] Open
Abstract
To assess mechanisms responsible for breast carcinoma metastasis, 4T1 breast carcinomas were grown orthotopically in wild type or Shb knockout mice. Tumor growth, metastasis, vascular characteristics and immune cell properties were analyzed. Absence of Shb did not affect tumor growth although it increased lung metastasis. Shb knockout mouse tumors showed decreased redness and less developed vascular plexa located at the periphery of the tumors. No difference in overall tumor vascular density, leakage or pericyte coverage was noted between the genotypes although the average vessel size was smaller in the knockout. Tumors induced an increase of CD11b+ cells in spleen, lymph node, thymus, bone marrow and blood. Numbers of Shb knockout CD11b/CD8+ cells were decreased in lymph nodes and bone marrow of tumor bearing mice. Mice with tumors had reduced numbers of CD4+ lymphocytes in blood/lymphoid organs, whereas in most of these locations the proportion of CD4+ cells co-expressing FoxP3 was increased, suggesting a relative increase in Treg cells. This finding was reinforced by increased blood interleukin-35 (IL-35) in wild type tumor bearing mice. Shb knockout blood showed in addition an increased proportion of IL-35 expressing Treg cells, supporting the notion that absence of Shb further promotes tumor evasion from immune cell recognition. This could explain the increased number of lung metastases observed under these conditions. In conclusion, 4T1 tumors alter immune cell responses that promote tumor expansion, metastasis and escape from T cell recognition in an Shb dependent manner.
Collapse
Affiliation(s)
- Xiujuan Li
- Department of Medical Cell Biology, Uppsala University, Uppsala 75123, Sweden.,Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Kailash Singh
- Department of Medical Cell Biology, Uppsala University, Uppsala 75123, Sweden
| | - Zhengkang Luo
- Department of Medical Cell Biology, Uppsala University, Uppsala 75123, Sweden
| | | | - Maria Jamalpour
- Department of Medical Cell Biology, Uppsala University, Uppsala 75123, Sweden
| | - Björn Lindahl
- Department of Medical Cell Biology, Uppsala University, Uppsala 75123, Sweden
| | - Ganlin Zhang
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala 75123, Sweden
| | - Stellan Sandler
- Department of Medical Cell Biology, Uppsala University, Uppsala 75123, Sweden
| | - Michael Welsh
- Department of Medical Cell Biology, Uppsala University, Uppsala 75123, Sweden
| |
Collapse
|
21
|
Chen C, Chen W, Li Y, Dong Y, Teng X, Nong Z, Pan X, Lv L, Gao Y, Wu G. Hyperbaric oxygen protects against myocardial reperfusion injury via the inhibition of inflammation and the modulation of autophagy. Oncotarget 2017; 8:111522-111534. [PMID: 29340072 PMCID: PMC5762340 DOI: 10.18632/oncotarget.22869] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/27/2017] [Indexed: 01/01/2023] Open
Abstract
Our previous study demonstrated that hyperbaric oxygen (HBO) preconditioning protected against myocardial ischemia reperfusion injury (MIRI) and improved myocardial infarction. However, HBO’s effect on MIRI-induced inflammation and autophagy remains unclear. In this study, we investigate the potential impact and underlying mechanism of HBO preconditioning on an MIRI-induced inflammatory response and autophagy using a ligation of the left anterior descending (LAD) coronary artery rat model. Our results showed that HBO restored myocardial enzyme levels and decreased the apoptosis of cardiomyocytes, which were induced by MIRI. Moreover, HBO significantly suppressed MIRI-induced inflammatory cytokines. This effect was associated with the inhibition of the TLR4-nuclear factor kappa-B (NF-κB) pathway. Interestingly, lower expression levels of microtubule-associated protein 1 light chain 3B (LC3B) and Beclin-1 were observed in the HBO-treatment group. Furthermore, we observed that HBO reduced excessive autophagy by activating the mammalian target of the rapamycin (mTOR) pathway, as evidenced by higher expression levels of threonine protein kinase (Akt) and phosphorylated-mTOR. In conclusion, HBO protected cardiomocytes during MIRI by attenuating inflammation and autophagy. Our results provide a new mechanistic insight into the cardioprotective role of HBO against MIRI.
Collapse
Affiliation(s)
- Chunxia Chen
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Wan Chen
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Yaoxuan Li
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Yanling Dong
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Xiaoming Teng
- Department of Neurology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Zhihuan Nong
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, Guangxi 530022, P. R. China
| | - Xiaorong Pan
- Department of Hyperbaric Oxygen, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Liwen Lv
- Department of Emergency, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| | - Ying Gao
- Department of Biology and Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Guangwei Wu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P. R. China
| |
Collapse
|