1
|
Gueiros LA, Gobbo M, Santos-Silva AR, Merigo E, Miranda-Silva W, Fregnani ER, Ottaviani G, Kauark-Fontes E, Bensadoun RJ, Arany P. Underexplored Areas of Photobiomodulation in Oral Oncology: An Expert Analysis. Photobiomodul Photomed Laser Surg 2024; 42:609-619. [PMID: 39422592 DOI: 10.1089/photob.2023.0155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Objective: This study aimed to review the current body of literature on underexplored areas of photobiomodulation (PBM) for preventing and/or treating oral adverse events. Background: Recent studies suggest that PBM may offer potential benefits in managing cancer-related toxicities other than oral mucositis. Nevertheless, further research to establish conclusive evidence is still missing. Methods: A panel of specialists conducted a narrative review to evaluate the evidence on PBM therapy for oral mucositis, xerostomia, dysgeusia, dysphagia, and trismus/fibrosis. Each topic was reviewed by two specialists who discussed treatment rationale, summarized current evidence, evaluated risk/benefit ratio, and identified future research directions. Results: The current evidence suggests promising outcomes in nonroutine uses of PBM for xerostomia, dysgeusia, odynophagia, oral mucositis (extraoral PBM and the pediatric population), and trismus/fibrosis. However, the primary studies are often small and may have biases that require further evaluation, particularly regarding treatment safety. Conclusion: Despite the overall positive impression of PBM therapy for oral adverse events of cancer treatment, robust evidence from large multicentered studies is necessary to support its widespread clinical use.
Collapse
Affiliation(s)
- Luiz Alcino Gueiros
- Departamento de Clínica e Odontologia Preventiva & Oral Medicine Unit, Hospital das Clínicas, Universidade Federal de Pernambuco, Recife, Brazil
| | - Margherita Gobbo
- Unit of Oral and Maxillofacial Surgery, Ca' Foncello Hospital, Treviso, Italy
| | - Alan Roger Santos-Silva
- Oral Diagnosis Department, Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | | | - Wanessa Miranda-Silva
- Molecular Oncology Center, Instituto de Ensino e Pesquisa, Hospital Sírio-Libanês, São Paulo, Brazil
| | | | - Giulia Ottaviani
- Department of Surgical, Medical and Health Sciences, University of Trieste, Trieste, Italy
| | - Elisa Kauark-Fontes
- Department of Propaedeutic and Integrated Clinic, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Praveen Arany
- Oral Biology, Surgery and Biomedical Engineering, University at Buffalo, New York, New York, USA
| |
Collapse
|
2
|
Ibrahim HA, Suardi N, Khaniabadi PM, Zulbaharin SFM, Taggo A. The cytotoxicity of breast cancer mcf-7 cell line treated with different wavelength of low-level laser. Lasers Med Sci 2024; 39:238. [PMID: 39307856 DOI: 10.1007/s10103-024-04187-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024]
Abstract
Breast cancer remains a significant global health challenge, spurring ongoing investigations into innovative treatment approaches. Low-level laser therapy (LLLT) has emerged as a promising non-invasive therapeutic avenue of interest. This research delves into the impact of LLLT on the cytotoxicity of the MCF-7 breast cancer cell line, employing lasers emitting various wavelengths. The objective is to assess whether diverse LLLT wavelengths elicit disparate cytotoxic responses, shedding light on LLLT's potential as a targeted breast cancer treatment. MCF-7 cell cultures were subjected to lasers of varying wavelengths, including blue (473 nm), red (660 nm), and near-infrared (780 nm). Each wavelength was delivered at four different power levels: 10, 25, 45, and 65 mW, with exposure durations of 60, 300, 600, and 900 s. Cellular responses, encompassing factors such as cell viability, and cytotoxicity were assessed using WST-1 assays technique. Statistical analysis was performed to discern the wavelength-specific impacts of low-level laser therapy (LLLT) on MCF-7 cells. The study revealed that the blue laser had the least noticeable adverse impact on MCF-7 breast cancer cell lines, leading to the highest cell survival rate of 107.62% after 24 h. The most severe toxicity occurred when the laser was used at 45 mW for 900 s, resulting in cell viability ranging from 81.85% to 107.62%. As for cell viability after exposure to the red laser, the mildest harmful effect was observed at 45 mW power for 60 s, resulting in a cell survival rate of 147.62%. Conversely, the most significant toxic response occurred at 10 mW power for 60 s, resulting in a cell viability of 91.56%. In contrast, when employing infrared laser irradiation, the least substantial cytotoxic effect on MCF-7 cells was observed at 10 mW power for 600 s, resulting in the highest cell viability of 109.37% after 24 h. The most pronounced cytotoxic effect was observed by infrared laser (780 nm) at 25 mW power for 900 s, leading to the lowest viability of 32.53%.
Collapse
Affiliation(s)
- Habibu Ahmad Ibrahim
- School of Physics, Universiti Sains Malaysia, 11800, Penang, Malaysia
- Department of Physics, Kano University of Science and Technology Wudil, Kano, Nigeria
| | - Nursakinah Suardi
- School of Physics, Universiti Sains Malaysia, 11800, Penang, Malaysia.
| | - Pegah Moradi Khaniabadi
- Community and Health Research Unit (CaHRU), School of Health & Social Care, University of Lincoln, Lincoln, LN5 7AY, United Kingdom
| | | | - Aijesta Taggo
- School of Physics, Universiti Sains Malaysia, 11800, Penang, Malaysia
| |
Collapse
|
3
|
Kang M, Lee Y, Lee Y, Kim E, Jo J, Shin H, Choi J, Oh J, Yoon H, Kang HW. Wavelength-dependent photobiomodulation (PBM) for proliferation and angiogenesis of melanoma tumor in vitro and in vivo. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 258:112990. [PMID: 39032372 DOI: 10.1016/j.jphotobiol.2024.112990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Photobiomodulation (PBM) has widely been used to effectively treat complications associated with cancer treatment, including oral mucositis, radiation dermatitis, and surgical wounds. However, the safety of PBM against cancer still needs to be validated as the effects of PBM on cancer cells and their mechanisms are unclear. The current study investigated the wavelength-dependent PBM effects by examining four different laser wavelengths (405, 532, 635, and 808 nm) on B16F10 melanoma tumor cells. In vitro tests showed that PBM with 808 nm promoted both proliferation and migration of B16F10 cells. In vivo results demonstrated that PBM with 808 nm significantly increased the relative tumor volume and promoted angiogenesis with overexpression of VEGF and HIF-1α. In addition, PBM induced the phosphorylation of factors closely related to cancer cell proliferation and tumor growth and upregulated the related gene expression. The current result showed that compared to the other wavelengths, 808 nm yielded a significant tumor-stimulating effect the malignant melanoma cancer. Further studies will investigate the in-depth molecular mechanism of PBM on tumor stimulation in order to warrant the safety of PBM for clinical cancer treatment.
Collapse
Affiliation(s)
- Myungji Kang
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine-Integrated Biomedical Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
| | - Yeachan Lee
- Center for Advanced Models for Translational Sciences and Therapeutics and Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yuri Lee
- School of Life Science, Handong Global University, Pohang 37554, Republic of Korea
| | - Eunjung Kim
- School of Life Science, Handong Global University, Pohang 37554, Republic of Korea
| | - Jihye Jo
- Research Center for Marine-Integrated Biomedical Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare and Digital Healthcare Research Center, College of Information Technology and Convergence, Pukyong National University, Busan 48513, Republic of Korea
| | - Hwarang Shin
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine-Integrated Biomedical Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea
| | - Jaeyeop Choi
- Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Junghwan Oh
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare and Digital Healthcare Research Center, College of Information Technology and Convergence, Pukyong National University, Busan 48513, Republic of Korea; Smart Gym-Based Translational Research Center for Active Senior's Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Hongsup Yoon
- School of Life Science, Handong Global University, Pohang 37554, Republic of Korea.
| | - Hyun Wook Kang
- Industry 4.0 Convergence Bionics Engineering, Department of Biomedical Engineering, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine-Integrated Biomedical Technology, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare and Digital Healthcare Research Center, College of Information Technology and Convergence, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
4
|
Seo KJ, Yoon JH, Chung BY, Lee HK, Park WS, Chae HS. Effects of photobiomodulation on colon cancer cell line HT29 according to mitochondria. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 257:112966. [PMID: 38970968 DOI: 10.1016/j.jphotobiol.2024.112966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND/AIM Although photobiomodulation therapy (PBMt) is available to alleviate post-operative side effects of malignant diseases, its application is still controversial due to some potential of cancer recurrence and occurrence of a secondary malignancy. We investigated effect of PBMt on mitochondrial function in HT29 colon cancer cells. METHODS HT29 cell proliferation was determined with MTT assay after PBMt. Immunofluorescent staining was performed to determine mitochondrial biogenesis and reactive oxygen species (ROS). Mitochondrial membrane potential was measured with Mitotracker. Western blotting was executed to determine expression of fission, fusion, UCP2, and cyclin B1 and D1 proteins. In vivo study was performed by subcutaneously inoculating cancer cells into nude mice and immunohistochemistry was done to determine expression of FIS1, MFN2, UCP2, and p-AKT. RESULTS The proliferation and migration of HT29 cells reached maximum with PBMt (670 nm, light emitting diode, LED) at 2.0 J/cm2 compared to control (P < 0.05) with more expression of cyclin B1 and cyclin D1 (P < 0.05). Immunofluorescent staining showed that ROS and mitochondrial membrane potential were enhanced after PBMt compared to control. ATP synthesis of mitochondria was also higher in the PBMt group than in the control (P < 0.05). Expression levels of fission and fusion proteins were significantly increased in the PBMt group than in the control (P < 0.05). Electron microscopy revealed that the percentage of mitochondria showing fission was not significantly different between the two groups. Oncometabolites including D-2-hydoxyglutamate in the supernatant of cell culture were higher in the PBMt group than in the control with increased UCP2 expression (P < 0.05). Both tumor size and weight of xenograft in nude mice model were bigger and heavier in the PBMt group than in the control (P < 0.05). Immunohistologically, mitochondrial biogenesis proteins UCP2 and p-AKT in xenograft of nude mice were expressed more in the PBMt group than in the control (P < 0.05). CONCLUSIONS Treatment with PBM using red light LED may induce proliferation and progression of HT29 cancer cells by increasing mitochondrial activity and fission.
Collapse
Affiliation(s)
- Kyung Jin Seo
- Department of Pathology, Uijongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Hwan Yoon
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Bom Yee Chung
- Department of Internal Medicine, Uijongbu St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hae Kyung Lee
- Department of Laboratory Medicine, Uijongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Won Sang Park
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Hiun Suk Chae
- Department of Internal Medicine, Uijongbu St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Razack SA, Lee Y, Bose S, Shin H, Jung WK, Kang HW. Photo-triggered caffeic acid delivery via psyllium polysaccharide- gellan gum-based injectable bionanogel for epidermoid carcinoma treatment. Int J Biol Macromol 2024; 267:131166. [PMID: 38582464 DOI: 10.1016/j.ijbiomac.2024.131166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/16/2024] [Accepted: 03/25/2024] [Indexed: 04/08/2024]
Abstract
Here, the simultaneous effect of chemo- and photothermal therapy against epidermoid carcinoma (EC) was investigated. A novel hydrogel, termed bionanogel (BNG), was designed using psyllium mucilage polysaccharide and bacterial gellan gum, incorporated with nanocomplex carrying caffeic acid (CA) and IR-820, and further characterized. The dual effect of BNG and 808 nm laser (BNG + L) on EC was investigated. Staining and scratch assays were performed to analyze their therapeutic effect on EC. In vivo evaluations of BNG + L in xenograft models were performed. Rapid transition, limited swelling, degradability and high tensile strength indicated BNG stability and sustained drug release. Irradiation with 808 nm laser light at 1.25 W /cm2 for 4 min resulted in a temperature increase of 53 °C and facilitated cell ablation. The in vitro studies showed that BNG + L suppressed cancer progression via a late apoptotic effect. The in vivo study showed that the slow release of CA from BNG + L significantly attenuated EC with low mitotic index and downregulation of proteins involved in cancer proliferation such as EGFR, AKT, PI3K, ERK, mTOR and HIF-1α. Thus, BNG could be a novel medium for targeted and controlled drug delivery for the treatment of epidermoid cancer when triggered by NIR light.
Collapse
Affiliation(s)
- Sirajunnisa Abdul Razack
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
| | - Yeachan Lee
- Center for Advanced Models for Translational Sciences and Therapeutics and Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sivakumar Bose
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea
| | - Hwarang Shin
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea; Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea; Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information, Pukyong National University, Busan, Republic of Korea
| | - Hyun Wook Kang
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, Republic of Korea; Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information, Pukyong National University, Busan, Republic of Korea.
| |
Collapse
|
6
|
Shakhova M, Elagin V, Plekhanov A, Khilov A, Kurakina D, Kamensky V, Kirillin M. Post-Operational Photodynamic Therapy of the Tumor Bed: Comparative Analysis for Cold Knife and Laser Scalpel Resection. Biomedicines 2024; 12:291. [PMID: 38397893 PMCID: PMC11154242 DOI: 10.3390/biomedicines12020291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
In this paper, we report on a study regarding the efficiency of the post-operational phototherapy of the tumor bed after resection with both a cold knife and a laser scalpel in laboratory mice with CT-26 tumors. Post-operational processing included photodynamic therapy (PDT) with a topically applied chlorin-based photosensitizer (PS), performed at wavelengths of 405 or 660 nm, with a total dose of 150 J/cm2. The selected design of the tumor model yielded zero recurrence in the laser scalpel group and 92% recurrence in the cold knife group without post-processing, confirming the efficiency of the laser scalpel in oncology against the cold knife. The application of PDT after the cold knife resection decreased the recurrence rate to 70% and 42% for the 405 nm and 660 nm procedures, respectively. On the other hand, the application of PDT after the laser scalpel resection induced recurrence rates of 18% and 30%, respectively, for the considered PDT performance wavelengths. The control of the penetration of PS into the tumor bed by fluorescence confocal microscopy indicated the deeper penetration of PS in the case of the cold knife, which presumably provided deeper PDT action, while the low-dose light exposure of deeper tissues without PS, presumably, stimulated tumor recurrence, which was also confirmed by the differences in the recurrence rate in the 405 and 660 nm groups. Irradiation-only light exposures, in all cases, demonstrated higher recurrence rates compared to the corresponding PDT cases. Thus, the PDT processing of the tumor bed after resection could only be recommended for the cold knife treatment and not for the laser scalpel resection, where it could induce tumor recurrence.
Collapse
Affiliation(s)
- Maria Shakhova
- Department of Ear, Nose and Throat Diseases, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia;
| | - Vadim Elagin
- Institute of Experimental Oncology and Biomedical Technologies, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia; (A.P.); (V.K.)
| | - Anton Plekhanov
- Institute of Experimental Oncology and Biomedical Technologies, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia; (A.P.); (V.K.)
| | - Aleksandr Khilov
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| | - Daria Kurakina
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| | - Vladislav Kamensky
- Institute of Experimental Oncology and Biomedical Technologies, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia; (A.P.); (V.K.)
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| | - Mikhail Kirillin
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| |
Collapse
|
7
|
da Silva TG, Rodrigues JA, Siqueira PB, Dos Santos Soares M, Mencalha AL, de Souza Fonseca A. Effects of photobiomodulation by low-power lasers and LEDs on the viability, migration, and invasion of breast cancer cells. Lasers Med Sci 2023; 38:191. [PMID: 37610503 DOI: 10.1007/s10103-023-03858-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/16/2023] [Indexed: 08/24/2023]
Abstract
Among the malignant tumors, breast cancer is the most commonly diagnosed worldwide, being the most prevalent in women. Photobiomodulation has been used for wound healing, swelling and pain reduction, and muscle repair. The application of photobiomodulation in cancer patients has been controversial. Therefore, a better understanding of radiation-induced effects involved in photobiomodulation on cancer cells is needed. Thus, this study aimed to investigate the effects of exposure to low-power lasers and LEDs on cell viability, migration, and invasion in human breast cancer cells. MCF-7 and MDA-MB-231 cells were irradiated with a low-power red laser (23, 46, and 69 J/cm2, 0.77 W/cm2) and blue LED (160, 321, and 482 J/cm2, 5.35 W/cm2), alone or in combination. Cell viability was assessed using the WST-1 assay, cell migration was evaluated using the wound healing assay, and cell invasion was performed using the Matrigel transwell assay. Viability and migration were not altered in MCF-7 and MDA-MB-231 cultures after exposure to low-power red laser and blue LED. However, there was a decrease in cell invasion from the cultures of the two cell lines evaluated. The results suggest that photobiomodulation induced by low-power red laser and blue LED does not alter cell viability and migration but decreases cell invasion in human breast cancer cells.
Collapse
Affiliation(s)
- Thayssa Gomes da Silva
- Departamento de Biofísica E Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade Do Estado Do Rio de Janeiro, Boulevard 28 de Setembro, 87, PAPC, 4Th Floor, CEP: 20.551-030, Vila Isabel, Rio de Janeiro, Brazil.
| | - Juliana Alves Rodrigues
- Departamento de Biofísica E Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade Do Estado Do Rio de Janeiro, Boulevard 28 de Setembro, 87, PAPC, 4Th Floor, CEP: 20.551-030, Vila Isabel, Rio de Janeiro, Brazil
| | - Priscyanne Barreto Siqueira
- Departamento de Biofísica E Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade Do Estado Do Rio de Janeiro, Boulevard 28 de Setembro, 87, PAPC, 4Th Floor, CEP: 20.551-030, Vila Isabel, Rio de Janeiro, Brazil
| | - Márcia Dos Santos Soares
- Departamento de Biofísica E Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade Do Estado Do Rio de Janeiro, Boulevard 28 de Setembro, 87, PAPC, 4Th Floor, CEP: 20.551-030, Vila Isabel, Rio de Janeiro, Brazil
| | - Andre Luiz Mencalha
- Departamento de Biofísica E Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade Do Estado Do Rio de Janeiro, Boulevard 28 de Setembro, 87, PAPC, 4Th Floor, CEP: 20.551-030, Vila Isabel, Rio de Janeiro, Brazil
| | - Adenilson de Souza Fonseca
- Departamento de Biofísica E Biometria, Instituto de Biologia Roberto Alcantara Gomes, Universidade Do Estado Do Rio de Janeiro, Boulevard 28 de Setembro, 87, PAPC, 4Th Floor, CEP: 20.551-030, Vila Isabel, Rio de Janeiro, Brazil
- Departamento de Ciências Fisiológicas, Instituto Biomédico, Universidade Federal Do Estado Do Rio de Janeiro, Rua Frei Caneca, 94, Rio de Janeiro, 20211040, Brazil
| |
Collapse
|
8
|
Kim YJ, Song J, Lee DH, Um SH, Bhang SH. Suppressing cancer by damaging cancer cell DNA using LED irradiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 243:112714. [PMID: 37084656 DOI: 10.1016/j.jphotobiol.2023.112714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND High-energy irradiation eliminates cancer cells by destroying their genetic components. However, there are several side effects from doing this, such as fatigue, dermatitis, and hair loss, which remain obstacles to this treatment. Here, we propose a moderate method that uses low-energy white light from a light-emitting diode (LED) to selectively inhibit cancer cell proliferation without affecting normal cells. METHODS The association between LED irradiation and cancer cell growth arrest was evaluated based on cell proliferation, viability, and apoptotic activity. Immunofluorescence, polymerase chain reaction, and western blotting were performed in vitro and in vivo to identify the metabolism related to the inhibition of HeLa cell proliferation. RESULTS LED irradiation aggravated the defective p53 signaling pathway and induced cell growth arrest in cancer cells. Consequently, cancer cell apoptosis was induced by the increased DNA damage. Additionally, LED irradiation inhibited the proliferation of cancer cells by suppressing the MAPK pathway. Furthermore, the suppression of cancer growth by the regulation of p53 and MAPK was observed in cancer-bearing mice irradiated with LED. CONCLUSIONS Our findings suggest that LED irradiation can suppress cancer cell activity and may contribute to preventing the proliferation of cancer cells after medical surgery without causing side effects.
Collapse
Affiliation(s)
- Yu-Jin Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Jihun Song
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
9
|
Di Gregorio E, Israel S, Staelens M, Tankel G, Shankar K, Tuszyński JA. The distinguishing electrical properties of cancer cells. Phys Life Rev 2022; 43:139-188. [PMID: 36265200 DOI: 10.1016/j.plrev.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
Abstract
In recent decades, medical research has been primarily focused on the inherited aspect of cancers, despite the reality that only 5-10% of tumours discovered are derived from genetic causes. Cancer is a broad term, and therefore it is inaccurate to address it as a purely genetic disease. Understanding cancer cells' behaviour is the first step in countering them. Behind the scenes, there is a complicated network of environmental factors, DNA errors, metabolic shifts, and electrostatic alterations that build over time and lead to the illness's development. This latter aspect has been analyzed in previous studies, but how the different electrical changes integrate and affect each other is rarely examined. Every cell in the human body possesses electrical properties that are essential for proper behaviour both within and outside of the cell itself. It is not yet clear whether these changes correlate with cell mutation in cancer cells, or only with their subsequent development. Either way, these aspects merit further investigation, especially with regards to their causes and consequences. Trying to block changes at various levels of occurrence or assisting in their prevention could be the key to stopping cells from becoming cancerous. Therefore, a comprehensive understanding of the current knowledge regarding the electrical landscape of cells is much needed. We review four essential electrical characteristics of cells, providing a deep understanding of the electrostatic changes in cancer cells compared to their normal counterparts. In particular, we provide an overview of intracellular and extracellular pH modifications, differences in ionic concentrations in the cytoplasm, transmembrane potential variations, and changes within mitochondria. New therapies targeting or exploiting the electrical properties of cells are developed and tested every year, such as pH-dependent carriers and tumour-treating fields. A brief section regarding the state-of-the-art of these therapies can be found at the end of this review. Finally, we highlight how these alterations integrate and potentially yield indications of cells' malignancy or metastatic index.
Collapse
Affiliation(s)
- Elisabetta Di Gregorio
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Simone Israel
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Michael Staelens
- Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada
| | - Gabriella Tankel
- Department of Mathematics & Statistics, McMaster University, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada
| | - Karthik Shankar
- Department of Electrical & Computer Engineering, University of Alberta, 9211 116 Street NW, Edmonton, T6G 1H9, AB, Canada
| | - Jack A Tuszyński
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada; Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, T6G 1Z2, AB, Canada.
| |
Collapse
|
10
|
Yang J, Fu Q, Jiang H, Li Y, Liu M. Progress of phototherapy for osteosarcoma and application prospect of blue light photobiomodulation therapy. Front Oncol 2022; 12:1022973. [PMID: 36313662 PMCID: PMC9606592 DOI: 10.3389/fonc.2022.1022973] [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: 08/19/2022] [Accepted: 09/20/2022] [Indexed: 12/02/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor that mainly affects the pediatric and adolescent population; limb salvage treatment has become one of the most concerned and expected outcomes of OS patients recently. Phototherapy (PT), as a novel, non-invasive, and efficient antitumor therapeutic approach including photodynamic therapy (PDT), photothermal therapy (PTT), and photobiomodulation therapy (PBMT), has been widely applied in superficial skin tumor research and clinical treatment. OS is the typical deep tumor, and its phototherapy research faces great limitations and challenges. Surprisingly, pulse mode LED light can effectively improve tissue penetration and reduce skin damage caused by high light intensity and has great application potential in deep tumor research. In this review, we discussed the research progress and related molecular mechanisms of phototherapy in the treatment of OS, mainly summarized the status quo of blue light PBMT in the scientific research and clinical applications of tumor treatment, and outlooked the application prospect of pulsed blue LED light in the treatment of OS, so as to further improve clinical survival rate and prognosis of OS treatment and explore corresponding cellular mechanisms.
Collapse
Affiliation(s)
- Jiali Yang
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Qiqi Fu
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Hui Jiang
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Yinghua Li
- Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
- *Correspondence: Yinghua Li, ; Muqing Liu,
| | - Muqing Liu
- School of Information Science and Technology, Fudan University, Shanghai, China
- Zhongshan Fudan Joint Innovation Center, Zhongshan, China
- *Correspondence: Yinghua Li, ; Muqing Liu,
| |
Collapse
|
11
|
Staelens M, Di Gregorio E, Kalra AP, Le HT, Hosseinkhah N, Karimpoor M, Lim L, Tuszyński JA. Near-Infrared Photobiomodulation of Living Cells, Tubulin, and Microtubules In Vitro. FRONTIERS IN MEDICAL TECHNOLOGY 2022; 4:871196. [PMID: 35600165 PMCID: PMC9115106 DOI: 10.3389/fmedt.2022.871196] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 11/29/2022] Open
Abstract
We report the results of experimental investigations involving photobiomodulation (PBM) of living cells, tubulin, and microtubules in buffer solutions exposed to near-infrared (NIR) light emitted from an 810 nm LED with a power density of 25 mW/cm2 pulsed at a frequency of 10 Hz. In the first group of experiments, we measured changes in the alternating current (AC) ionic conductivity in the 50–100 kHz range of HeLa and U251 cancer cell lines as living cells exposed to PBM for 60 min, and an increased resistance compared to the control cells was observed. In the second group of experiments, we investigated the stability and polymerization of microtubules under exposure to PBM. The protein buffer solution used was a mixture of Britton-Robinson buffer (BRB aka PEM) and microtubule cushion buffer. Exposure of Taxol-stabilized microtubules (~2 μM tubulin) to the LED for 120 min resulted in gradual disassembly of microtubules observed in fluorescence microscopy images. These results were compared to controls where microtubules remained stable. In the third group of experiments, we performed turbidity measurements throughout the tubulin polymerization process to quantify the rate and amount of polymerization for PBM-exposed tubulin vs. unexposed tubulin samples, using tubulin resuspended to final concentrations of ~ 22.7 μM and ~ 45.5 μM in the same buffer solution as before. Compared to the unexposed control samples, absorbance measurement results demonstrated a slower rate and reduced overall amount of polymerization in the less concentrated tubulin samples exposed to PBM for 30 min with the parameters mentioned above. Paradoxically, the opposite effect was observed in the 45.5 μM tubulin samples, demonstrating a remarkable increase in the polymerization rates and total polymer mass achieved after exposure to PBM. These results on the effects of PBM on living cells, tubulin, and microtubules are novel, further validating the modulating effects of PBM and contributing to designing more effective PBM parameters. Finally, potential consequences for the use of PBM in the context of neurodegenerative diseases are discussed.
Collapse
Affiliation(s)
- Michael Staelens
- Department of Physics, University of Alberta, Edmonton, AB, Canada
| | | | - Aarat P. Kalra
- Scholes Lab, Department of Chemistry, Princeton University, Princeton, NJ, United States
| | - Hoa T. Le
- Department of Physics, University of Alberta, Edmonton, AB, Canada
| | | | | | - Lew Lim
- Vielight Inc., Toronto, ON, Canada
| | - Jack A. Tuszyński
- Department of Physics, University of Alberta, Edmonton, AB, Canada
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Jack A. Tuszyński
| |
Collapse
|
12
|
Gholami L, Khorsandi K, Taghdiri Nooshabadi V, Shahabi S, Jazaeri M, Esfahani H, Rabiei Faradonbeh D, Veisi Malekshahi Z, Afsartala Z, Mostafa N. Effect of Photobiomodulation on Structure and Function of Extracellular Vesicle Secreted from Mesenchymal Stem Cells. Photochem Photobiol 2022; 98:1447-1458. [DOI: 10.1111/php.13633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/02/2022] [Accepted: 04/03/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Leila Gholami
- Department of periodontics, Dental Research Center Hamadan University of Medical Sciences Hamadan Iran
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry University of British Columbia Canada
| | - Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center Yara Institute ACECR Tehran Iran
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences The George Washington University Washington DC 20037 USA
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine Semnan University of Medical Science Iran
| | - Shiva Shahabi
- Student Research Committee, School of Dentistry Hamadan University of Medical Sciences Iran
| | - Marzieh Jazaeri
- Student Research Committee, School of Dentistry Hamadan University of Medical Sciences Iran
| | - HomaSadat Esfahani
- Department of Photodynamic, Medical Laser Research Center Yara Institute ACECR Tehran Iran
| | - Davood Rabiei Faradonbeh
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine Tehran University of Medical Sciences Tehran Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine Tehran University of Medical Sciences Tehran Iran
| | - Zohreh Afsartala
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute Tehran University of Medical Science Tehran Iran
| | - Nesrine Mostafa
- Department of Oral Health Sciences, Faculty of Dentistry University of British Columbia Canada
| |
Collapse
|
13
|
Baczewska M, Stępień P, Mazur M, Krauze W, Nowak N, Szymański J, Kujawińska M. Method to analyze effects of low-level laser therapy on biological cells with a digital holographic microscope. APPLIED OPTICS 2022; 61:B297-B306. [PMID: 35201152 DOI: 10.1364/ao.445337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
Low-level laser therapy (LLLT) is a therapeutic tool that uses the photobiochemical interaction between light and tissue. Its effectiveness is controversial due to a strong dependence on dosimetric parameters. In this work, we demonstrate that digital holographic microscopy is an effective label-free imaging technique to analyze the effects of LLLT on biological cells, and we propose the full methodology to create correct synthetic aperture phase maps for further extensive, highly accurate statistical analysis. The proposed methodology has been designed to provide a basis for many other biological experiments using quantitative phase imaging. We use SHSY-5Y and HaCaT cells irradiated with different doses of red light for the experiment. The analysis shows quantitative changes in cell dry mass density and the projected cell surface in response to different radiation doses.
Collapse
|
14
|
Xia Y, Yu W, Cheng F, Rao T, Ruan Y, Yuan R, Ning J, Zhou X, Lin F, Zheng D. Photobiomodulation With Blue Laser Inhibits Bladder Cancer Progression. Front Oncol 2021; 11:701122. [PMID: 34733776 PMCID: PMC8558536 DOI: 10.3389/fonc.2021.701122] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/30/2021] [Indexed: 01/22/2023] Open
Abstract
Blue lasers are becoming more widely used in the diagnosis and treatment of bladder cancer; however, their photobiomodulation effects on bladder cancer cells remains unclear. The purpose of the current study was to explore the photobiomodulation effect of blue laser irradiation on bladder cancer progression and the associated mechanisms. The human uroepithelial cell line SV-HUC-1 and human bladder cancer cell lines T24 and EJ were exposed to blue laser irradiation (450 nm) at various energy densities, and cell proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), and the levels of the proteins associated with the MAPK pathway proteins were determined. A significant decrease in cell viability was observed in a density-dependent manner after blue laser irradiation at > 4 J/cm2 in both bladder cancer cell lines. However, the blue laser did not reduce cell viability in SV-HUC-1 cells until the energy density exceeded 16 J/cm2. Meanwhile, Ki67 levels, reflecting cell proliferation and senescence, were also significantly decreased after blue laser irradiation at 4 J/cm2 and 8 J/cm2 in the absence of cell cycle arrest. Moreover, blue laser irradiation at 4 J/cm2 and 8 J/cm2 caused a reduction in cell migration and invasion and also reduced the expression levels of MMP-2, MMP-9, Snail, N-cadherin, phospho-MEK and phospho-ERK, and elevated the expression levels of E-cadherin. Meanwhile ERK activator(tBHQ) significantly reversed the irradiation-induced suppression of proliferation, migration and invasion in T24 and EJ cell lines. The present study showed that blue laser irradiation inhibited bladder cancer proliferation in a density-dependent manner and inhibited bladder cancer progression by suppressing migration, invasion, and the EMT process in T24 and EJ cell lines. This inhibition was possibly mediated via suppression of the MAPK/MEK/ERK pathway. Thus, the use of a low-energy blue laser in the diagnosis and treatment of bladder cancer is possibly safe and may have an anti-tumor effect.
Collapse
Affiliation(s)
- Yuqi Xia
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuan Ruan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Run Yuan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jinzhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Di Zheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Fractionated low-level laser irradiation on breast cancer (MCF 7 cells) treatment. Lasers Med Sci 2021; 37:1265-1271. [PMID: 34331605 DOI: 10.1007/s10103-021-03384-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
Breast cancer is responsible for one of the top leading causes of cancer deaths among women. Radiotherapy (RT) uses high energy radiation to kill cancer cells, but this method has been reportedly linked to risks of toxicity. Post-therapeutic relapse from RT believed to be caused by its toxicity is one of the challenges encountered during tumour therapy. Therefore, further attention should be devoted to developing novel anti-tumour therapeutic approaches. The role of low-level laser therapy (LLLT) in breast cancer management is to alleviate the side effects arising from RT, instead of acting against the tumour cells directly. This study investigated the effects of low-level laser (532 nm), as well as single and fractionated irradiation, on breast cancer MCF 7 cell line. Additionally, this study assessed the most effective laser parameter for fractionated irradiation. The MCF 7 cells were irradiated with green laser power at 1.5, 45.0, and 100.0 mW with a spot size diameter of 0.7 mm for 1, 5, 10, and 15 min. The irradiation was carried out in single, double, and triple fractionation separated by 5- and 10-min intervals in between the fractional regimes. The laser output of 100 mW showed a promising potential in killing cells with single fractionation. However, as the irradiation was fractionated into two, power of 1.5 mW appeared to be more effective in cell death, which contributed to the lowest percentage cells viable of 31.4% recorded in the study. It was proven that fractionated regime was more successful in tumour cell death.
Collapse
|
16
|
Multi-Lens Arrays (MLA)-Assisted Photothermal Effects for Enhanced Fractional Cancer Treatment: Computational and Experimental Validations. Cancers (Basel) 2021; 13:cancers13051146. [PMID: 33800182 PMCID: PMC7962441 DOI: 10.3390/cancers13051146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Colorectal cancer is one of the most common cancers and the third leading cause of cancer-related deaths in the United States. As a non- or minimally invasive cancer treatment, photothermal therapy (PTT) has been widely used to generate irreversible thermal injuries in tumors. However, conventional PTT employs an end-firing flat fiber to deliver laser energy, leading to the incomplete removal of tumor tissues due to an uneven beam distribution over the tumor surface. Multi-lens arrays (MLA) generate multiple micro-beams to uniformly distribute laser energy on the tissue surface. Therefore, the application of MLA for PTT in cancer affords a spatially enhanced distribution of micro-beams and laser-induced temperature in the tumor. The purpose of the current study is to computationally and experimentally demonstrate the therapeutic benefits of MLA-assisted fractional PTT on colorectal cancer, in comparison to flat fiber-based PTT. Abstract Conventional photothermal therapy (PTT) for cancer typically employs an end-firing flat fiber (Flat) to deliver laser energy, leading to the incomplete treatment of target cells due to a Gaussian-shaped non-uniform beam profile. The purpose of the current study is to evaluate the feasibility of multi-lens arrays (MLA) for enhanced PTT by delivering laser light in a fractional micro-beam pattern. Computational and experimental evaluations compare the photothermal responses of gelatin phantoms and aqueous dye solutions to irradiations with Flat and MLA. In vivo colon cancer models have been developed to validate the therapeutic capacity of MLA-assisted irradiation. MLA yields 1.6-fold wider and 1.9-fold deeper temperature development in the gelatin phantom than Flat, and temperature monitoring identified the optimal treatment condition at an irradiance of 2 W/cm2 for 180 s. In vivo tests showed that the MLA group was accompanied by complete tumor eradication, whereas the Flat group yielded incomplete removal and significant tumor regrowth 14 days after PTT. The proposed MLA-assisted PTT spatially augments photothermal effects with the fractional micro-beams on the tumor and helps achieve complete tumor removal without recurrence. Further investigations are expected to optimize treatment conditions with various wavelengths and photosensitizers to warrant treatment efficacy and safety for clinical translation.
Collapse
|
17
|
Hosseinpour S, Xu C, Walsh LJ. Impact of photobiomodulation using four diode laser wavelengths of on cationic liposome gene transfection into pre-osteoblast cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 215:112108. [PMID: 33418241 DOI: 10.1016/j.jphotobiol.2020.112108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 11/21/2020] [Accepted: 12/18/2020] [Indexed: 11/16/2022]
Abstract
Gene therapy can be an effective treatment modality for some severe genetic diseases. Despite efforts to improve their performance, non-viral gene delivery methods remain inefficient and costly. As an alternative to viral vectors, cationic liposomes have a good safety profile and low immunogenicity, but relatively low transfection efficiency. They may also be toxic to cells at high concentrations. Given these challenges, the present study explored the impact of photobiomodulation (PBM) on cationic liposome plasmid DNA transfection in terms of its efficiency and toxicity, using Lipofectamine 2000 to carry green fluorescent protein (GFP) encoding plasmid DNA, with the pre-osteoblast MC3T3-E1 cell line as the target. Cultures were irradiated using diode lasers (445, 685, 810, or 970 nm) at 200 mW using pulsed mode (50 Hz), with a power density of 104.64 mW/cm2, and irradiance from 6 to 18 joules. To determine transfection efficiency, expression of GFP was assessed using confocal laser scanning microscopy and flow cytometry. Cell viability was evaluated using the MTT assay. PBM using 810 nm and 970 nm lasers significantly enhanced transfection efficiency for GFP, indicating more efficient uptake of plasmid DNA. Conversely, laser irradiation at 445 nm and 685 nm wavelengths reduced the GFP transfection efficiency. Treatment using 685, 810, and 970 nm lasers at 12 J maintained cell viability and prevented toxicity of cationic liposomes. Overall, these findings support the concept that PBM using near infrared laser wavelengths can enhance transfection efficiency and support cell viability when cationic liposomes are used as the vector in gene therapy.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, The University of Queensland, Herston, QLD, 4006, Australia.
| | - Chun Xu
- School of Dentistry, The University of Queensland, Herston, QLD, 4006, Australia.
| | - Laurence J Walsh
- School of Dentistry, The University of Queensland, Herston, QLD, 4006, Australia.
| |
Collapse
|
18
|
Günaydın A, Çakıcı EB. Effect of Photobiomodulation Therapy following Direct Pulp Capping on Postoperative Sensitivity by Thermal Stimulus: A Retrospective Study. Med Princ Pract 2021; 30:347-354. [PMID: 33827095 PMCID: PMC8436640 DOI: 10.1159/000516342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/05/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The aim of this study was to investigate the effect of photobiomodulation therapy (PBMT) on postoperative pain provoked by thermal stimulation in direct pulp capping (DPC). SUBJECTS AND METHODS A retrospective study was performed using the records of patients who received DPC using mineral trioxide aggregate. Teeth irradiated with a laser were assigned as the PBMT group, and nonirradiated teeth were considered as the control group. Before treatment and 6 h, 1 day, and 7 days after treatment, tooth sensitivity to a cold stimulus was recorded using a visual analog scale. RESULTS From a total of 123 documented DPC procedures, only 72 directly capped permanent teeth met the inclusion criteria. Age, gender, and tooth location were comparable between the groups. A statistically significant difference was found in sensitivity to cold stimulus between groups on day 7 (p = 0.007), but no difference was found at the preoperative, 6 h, and day 1 time points (p = 0.055, p = 0.132, and p = 0.100, respectively). In the intragroup evaluation, a significantly greater decrease in sensitivity to cold stimulus was detected in the PBMT group than that in the control group, although both groups showed a reduction in discomfort throughout the follow-up period (p = 0.000). CONCLUSION PBMT is an effective method for enhancing patient comfort by reducing thermal sensitivity following DPC procedures.
Collapse
|
19
|
Golovynska I, Golovynskyi S, Stepanov YV, Stepanova LI, Qu J, Ohulchanskyy TY. Red and near-infrared light evokes Ca 2+ influx, endoplasmic reticulum release and membrane depolarization in neurons and cancer cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 214:112088. [PMID: 33278762 DOI: 10.1016/j.jphotobiol.2020.112088] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/26/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Low level light therapy uses light of specific wavelengths in red and near-infrared spectral range to treat various pathological conditions. This light is able to modulate biochemical cascade reactions in cells that can have important health implications. In this study, the effect of low intensity light at 650, 808 and 1064 nm on neurons and two types of cancer cells (neuroblastoma and HeLa) is reported, with focus on the photoinduced change of intracellular level of Ca2+ ions and corresponding signaling pathways. The obtained results show that 650 and 808 nm light promotes intracellular Ca2+ elevation regardless of cell type, but with different dynamics due to the specificities of Ca2+ regulation in neurons and cancer cells. Two origins responsible for Ca2+ elevation are determined to be: influx of exogenous Ca2+ ions into cells and Ca2+ release from endoplasmic reticulum. Our investigation of the related cellular processes shows that light-induced membrane depolarization is distinctly involved in the mechanism of Ca2+ influx. Ca2+ release from endoplasmic reticulum activated by reactive oxygen species generation is considered as a possible light-dependent signaling pathway. In contrast to the irradiation with 650 and 808 nm light, no effects are observed under 1064 nm irradiation. We believe that the obtained insights are of high significance and can be useful for the development of drug-free phototherapy.
Collapse
Affiliation(s)
- Iuliia Golovynska
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, PR China
| | - Sergii Golovynskyi
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, PR China
| | - Yurii V Stepanov
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, PR China
| | - Liudmyla I Stepanova
- Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Junle Qu
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, PR China.
| | - Tymish Y Ohulchanskyy
- Center for Biomedical Photonics, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, PR China.
| |
Collapse
|
20
|
In Vitro Cytological Responses against Laser Photobiomodulation for Periodontal Regeneration. Int J Mol Sci 2020; 21:ijms21239002. [PMID: 33256246 PMCID: PMC7730548 DOI: 10.3390/ijms21239002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 02/06/2023] Open
Abstract
Periodontal disease is a chronic inflammatory disease caused by periodontal bacteria. Recently, periodontal phototherapy, treatment using various types of lasers, has attracted attention. Photobiomodulation, the biological effect of low-power laser irradiation, has been widely studied. Although many types of lasers are applied in periodontal phototherapy, molecular biological effects of laser irradiation on cells in periodontal tissues are unclear. Here, we have summarized the molecular biological effects of diode, Nd:YAG, Er:YAG, Er,Cr:YSGG, and CO2 lasers irradiation on cells in periodontal tissues. Photobiomodulation by laser irradiation enhanced cell proliferation and calcification in osteoblasts with altering gene expression. Positive effects were observed in fibroblasts on the proliferation, migration, and secretion of chemokines/cytokines. Laser irradiation suppressed gene expression related to inflammation in osteoblasts, fibroblasts, human periodontal ligament cells (hPDLCs), and endothelial cells. Furthermore, recent studies have revealed that laser irradiation affects cell differentiation in hPDLCs and stem cells. Additionally, some studies have also investigated the effects of laser irradiation on endothelial cells, cementoblasts, epithelial cells, osteoclasts, and osteocytes. The appropriate irradiation power was different for each laser apparatus and targeted cells. Thus, through this review, we tried to shed light on basic research that would ultimately lead to clinical application of periodontal phototherapy in the future.
Collapse
|
21
|
Bensadoun RJ, Epstein JB, Nair RG, Barasch A, Raber-Durlacher JE, Migliorati C, Genot-Klastersky MT, Treister N, Arany P, Lodewijckx J, Robijns J. Safety and efficacy of photobiomodulation therapy in oncology: A systematic review. Cancer Med 2020; 9:8279-8300. [PMID: 33107198 PMCID: PMC7666741 DOI: 10.1002/cam4.3582] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022] Open
Abstract
We performed a systematic review of the current literature addressing the safety and efficacy of photobiomodulation therapy (PBMT) in cancer patients. In this systematic review, the Preferred Reporting Items for Systematic Reviews and Meta‐Analyses (PRISMA) guidelines were used. In vitro, in vivo, and clinical studies, which investigated the effect of PBMT on cell proliferation/differentiation, tumor growth, recurrence rate, and/or overall survival were included. The Medline/PubMed, EMBASE, and Scopus databases were searched through April 2020. A total of 67 studies met the inclusion criteria with 43 in vitro, 15 in vivo, and 9 clinical studies identified. In vitro studies investigating the effect of PBMT on a diverse range of cancer cell lines demonstrated conflicting results. This could be due to the differences in used parameters and the frequency of PBM applications. In vivo studies and clinical trials with a follow‐up period demonstrated that PBMT is safe with regards to tumor growth and patient advantage in the prevention and treatment of specific cancer therapy‐related complications. Current human studies, supported by most animal studies, show safety with PBMT using currently recommended clinical parameters, including in Head & Neck cancer (HNC) in the area of PBMT exposure. A significant and growing literature indicates that PBMT is safe and effective, and may even offer a benefit in patient overall survival. Nevertheless, continuing research is indicated to improve understanding and provide further elucidation of remaining questions regarding PBM use in oncology.
Collapse
Affiliation(s)
| | - Joel B Epstein
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA.,Cedars-Sinai Health System, Los Angeles, CA, USA
| | - Raj G Nair
- Oral Medicine/Oral Oncology, Griffith University and Haematology and Oncology, Gold Coast University Hospital, Queensland Health, Gold Coast, QLD, Australia
| | - Andrei Barasch
- Harvard School of Dental Medicine, Cambridge Health Alliance, Cambridge, MA, USA
| | - Judith E Raber-Durlacher
- Department of Oral Medicine, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Oral and Maxillofacial Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Cesar Migliorati
- College of Dentistry, Department of Oral and Maxillofacial Diagnostic Sciences, University of Florida, Gainesville, FL, USA
| | | | - Nathaniel Treister
- Department of Oral Medicine, Harvard School of Dental Medicine, Boston, MA, USA
| | - Praveen Arany
- School of Dental Medicine, University of Buffalo, Buffalo, NY, USA
| | - Joy Lodewijckx
- Faculty of Medicine and Life Sciences, UHasselt, Hasselt, Belgium
| | - Jolien Robijns
- Faculty of Medicine and Life Sciences, UHasselt, Hasselt, Belgium
| | | |
Collapse
|
22
|
Cherkasova E, Babak K, Belotelov A, Labutina J, Yusupov V, Vorobieva N, Nerush A, Maslennikova A. Effects of photobiomodulation in relation to HeLa Kyoto tumor cells exposed to ionizing radiation. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 209:111936. [DOI: 10.1016/j.jphotobiol.2020.111936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 01/19/2023]
|
23
|
Tam SY, Tam VCW, Ramkumar S, Khaw ML, Law HKW, Lee SWY. Review on the Cellular Mechanisms of Low-Level Laser Therapy Use in Oncology. Front Oncol 2020; 10:1255. [PMID: 32793501 PMCID: PMC7393265 DOI: 10.3389/fonc.2020.01255] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
Photobiomodulation (PBM) using low-level laser therapy (LLLT) is a treatment that is increasingly used in oncology. Studies reported enhancement of wound healing with reduction in pain, tissue swelling and inflammatory conditions such as radiation dermatitis, oral mucositis, and lymphedema. However, factors such as wavelength, energy density and irradiation frequency influence the cellular mechanisms of LLLT. Moreover, the effects of LLLT vary according to cell types. Thus, controversy arose as a result of poor clinical response reported in some studies that may have used inadequately planned treatment protocols. Since LLLT may enhance tumor cell proliferation, these will also need to be considered before clinical use. This review aims to summarize the current knowledge of the cellular mechanisms of LLLT by considering its effects on cell proliferation, metabolism, angiogenesis, apoptosis and inflammation. With a better understanding of the cellular mechanisms, bridging findings from laboratory studies to clinical application can be improved.
Collapse
Affiliation(s)
- Shing Yau Tam
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Victor C W Tam
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Shanmugasundaram Ramkumar
- Department of Clinical Oncology, NHS Foundation Trust, University Hospital Southampton, Southampton, United Kingdom
| | - May Ling Khaw
- Tasmanian School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Helen K W Law
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Shara W Y Lee
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| |
Collapse
|
24
|
Tian Y, Kim H, Kang HW. In vitro anti-tumor effect of high-fluence low-power laser light on apoptosis of human colorectal cancer cells. Lasers Med Sci 2020; 36:513-520. [PMID: 32462231 DOI: 10.1007/s10103-020-03050-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/22/2020] [Indexed: 11/24/2022]
Abstract
Colorectal cancer is the third most common malignancy all over the world, along with high morbidity and mortality. As a treatment, high-fluence low-power laser irradiation (HF-LPLI) has reported that its biostimulatory activity can suppress or even destruct tumor growth in neoplastic diseases. The aim of the present study is to examine a therapeutic capacity of HF-LPLI for colorectal cancer treatment by using human colon cancer cell (HT29) model. The in vitro cancer cell model was used to analyze the underlying mechanism of laser-induced apoptosis. Laser irradiation was performed five times (once a day for five consecutive days) with 635 nm laser light for 8 and 16 min (fluence = 128 and 256 J/cm2), respectively. The efficiency of the HF-LPLI treatment was evaluated by MTT, fluorescence staining, cell wound healing, and western blot test during the 5-day period. Experiment data showed that HF-LPLI had a dose-dependent stimulating effect on cell viability, migration, and apoptosis of HT29 cells. The inhibition effect of laser treatment at 256 J/cm2 on cell viability was statistically significant. Meanwhile, the wound healing and western blot tests also confirmed that HF-LPLI could inhibit cell migration and induce cell apoptosis. The current research results demonstrate that 635 nm HF-LPLI can be an alternative treatment option for colorectal cancer by increasing the expression of caspase-3 and inducing HT29 tumor cell apoptosis through activation of the mitochondrial pathway.
Collapse
Affiliation(s)
- Ye Tian
- Interdisciplinary Program of Marine-Bio, Electrical & Mechanical Engineering, Pukyong National University, Busan, South Korea
| | - Hyejin Kim
- Interdisciplinary Program of Marine-Bio, Electrical & Mechanical Engineering, Pukyong National University, Busan, South Korea
| | - Hyun Wook Kang
- Interdisciplinary Program of Marine-Bio, Electrical & Mechanical Engineering, Pukyong National University, Busan, South Korea. .,Department of Biomedical Engineering and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, 48513, South Korea.
| |
Collapse
|
25
|
Cheng W, Yao M, Sun K, Li W. Progress in Photobiomodulation for Bone Fractures: A Narrative Review. PHOTOBIOMODULATION PHOTOMEDICINE AND LASER SURGERY 2020; 38:260-271. [PMID: 32427551 DOI: 10.1089/photob.2019.4732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Objective: The aim of this article is to examine current concepts and the future direction of implementing photobiomodulation (PBM) for fracture treatment. Background data: The effectiveness of PBM for bone regeneration has been demonstrated throughout in vitro studies and animal models. Yet, insufficient clinical trials have been reported on treating fractures with PBM. Materials and methods: A narrative review was composed on the basis of a literary search. Inclusion criteria consisted of studies between 2000 and 2019 using animal or human fracture models. Exclusion criteria consisted of studies that did not pertain to complete fractures or used other forms of intervention. Results: Ten animal studies on rats and rabbits and four clinical trials were found on using PBM for complete fractures. Conclusions: Based on positive outcomes in animal trials, parameter optimization of PBM for human fractures still requires extensive research on factors such as dosage, wavelength, penetration depth, treatment frequency, and the use of pulsed waves.
Collapse
Affiliation(s)
- Weyland Cheng
- Department of Orthopaedics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Manye Yao
- Department of Orthopaedics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Keming Sun
- Department of Orthopaedics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Weili Li
- Department of Orthopaedics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
26
|
Tam SY, Wu VWC, Law HKW. Hypoxia-Induced Epithelial-Mesenchymal Transition in Cancers: HIF-1α and Beyond. Front Oncol 2020; 10:486. [PMID: 32322559 PMCID: PMC7156534 DOI: 10.3389/fonc.2020.00486] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 03/18/2020] [Indexed: 01/10/2023] Open
Abstract
Metastasis is the main cause of cancer-related mortality. Although the actual process of metastasis remains largely elusive, epithelial-mesenchymal transition (EMT) has been considered as a major event in metastasis. Besides, hypoxia is common in solid cancers and has been considered as an important factor for adverse treatment outcomes including metastasis. Since EMT and hypoxia potentially share several signaling pathways, many recent studies focused on investigate the issue of hypoxia-induced EMT. Among all potential mediators of hypoxia-induced EMT, hypoxia-inducible factor-1α (HIF-1α) has been studied extensively. Moreover, there are other potential mediators that may also contribute to the process. This review aims to summarize the recent reports on hypoxia-induced EMT by HIF-1α or other potential mediators and provide insights for further investigations on this issue. Ultimately, better understanding of hypoxia-induced EMT may allow us to develop anti-metastatic strategies and improve treatment outcomes.
Collapse
Affiliation(s)
- Shing Yau Tam
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Vincent W C Wu
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Helen K W Law
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| |
Collapse
|
27
|
Hu J, Shi J, Gao Y, Yang W, Liu P, Liu Q, He F, Wang C, Li T, Xie R, Zhu J, Yang P. 808 nm Near-Infrared Light-Excited UCNPs@mSiO 2-Ce6-GPC3 Nanocomposites For Photodynamic Therapy In Liver Cancer. Int J Nanomedicine 2019; 14:10009-10021. [PMID: 31908456 PMCID: PMC6929933 DOI: 10.2147/ijn.s221496] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/02/2019] [Indexed: 12/23/2022] Open
Abstract
Background It is important to explore effective treatment for liver cancer. Photodynamic therapy (PDT) is a novel technique to treat liver cancer, but its clinical application is obstructed by limited depth of visible light penetration into tissue. The near-infrared (NIR) photosensitizer is a potential solution to the limitations of PDT for deep tumor tissue treatment. Purpose We aimed to investigate 808 nm NIR light-excited UCNPs@mSiO2-Ce6-GPC3 nanocomposites for PDT in liver cancer. Methods In our study, 808 nm NIR light-excited upconversion nanoparticles (UCNPs) were simultaneously loaded with the photosensitizer chlorin e6 (Ce6) and the antibody glypican-3 (GPC3), which is overexpressed in hepatocellular carcinoma cells. The multitasking UCNPs@mSiO2-Ce6-GPC3 nanoparticles under 808 nm laser irradiation with enhanced depth of penetration would enable the effective targeting of PDT. Results We found that the UCNPs@mSiO2-Ce6-GPC3 nanoparticles had good biocompatibility, low toxicity, excellent cell imaging in HepG2 cancer cells and high anti-tumor effect in vitro and in vivo. Conclusion We believe that the utilization of 808 nm NIR excited UCNPs@mSiO2-Ce6-GPC3 nanoparticles for PDT is a safe and potential therapeutic option for liver cancer.
Collapse
Affiliation(s)
- Jiahe Hu
- Department of Digestive Internal Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Jialan Shi
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin 150001, People's Republic of China.,Department of Surgery, VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 12132, USA
| | - Yingqian Gao
- Department of Digestive Internal Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Wei Yang
- Department of Digestive Internal Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Ping Liu
- Department of Digestive Internal Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Qinghao Liu
- Department of Digestive Internal Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Fei He
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin 150001, People's Republic of China
| | - Chunxu Wang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin 150001, People's Republic of China
| | - Tao Li
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin 150001, People's Republic of China
| | - Rui Xie
- Department of Digestive Internal Medicine, Harbin Medical University Cancer Hospital, Harbin 150081, People's Republic of China
| | - Jiuxin Zhu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratories of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin 150001, People's Republic of China
| |
Collapse
|
28
|
Ban D, Hua S, Zhang W, Shen C, Miao X, Liu W. Costunolide reduces glycolysis-associated activation of hepatic stellate cells via inhibition of hexokinase-2. Cell Mol Biol Lett 2019; 24:52. [PMID: 31428167 PMCID: PMC6694499 DOI: 10.1186/s11658-019-0179-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/08/2019] [Indexed: 02/06/2023] Open
Abstract
Background Hepatic stellate cell (HSC) activation is a central event during hepatic fibrosis. Aerobic glycolysis is one of its metabolic hallmarks. Blocking glycolysis is a novel therapeutic option for liver fibrosis. This study investigated the effects of costunolide, a natural product demonstrated to have hepatoprotective effects, on HSC activation and glycolysis. Methods Primary HSCs were isolated from rats and cultured through 5 to 6 passages. Cell viability, activation markers, and glycolytic metabolism were examined in primary HSCs using various cellular and molecular approaches. Results At 30 μM, costunolide reduced the viability of HSCs and inhibited the expression of α-smooth muscle actin and collagen I, two key markers of HSC activation. It also decreased glucose uptake and consumption, and reduced the intracellular levels of lactate in HSCs. At 10 mM, the glycolysis inhibitor 2-DG had a similar impact to costunolide at 30 μM: it significantly downregulated the expression of HSC activation markers. The combination of the two compounds produced more remarkable effects. Furthermore, costunolide repressed the expression and activity of hexokinase 2 (HK2), a pivotal rate-limiting enzyme that regulates glycolysis. However, overexpression of HK2 via plasmid transfection significantly reversed the costunolide-mediated downregulation of activation markers in HSCs, indicating that suppression of HK2 was required for costunolide to inhibit glycolysis-associated HSC activation. Conclusions Our results show that costunolide can suppress HSC activation, and this is associated with inhibition of HK2, which blocks aerobic glycolysis. This suggests that costunolide is an antifibrotic candidate with potential for further development.
Collapse
Affiliation(s)
- Dujing Ban
- 1Nanjing University of Chinese Medicine, Nanjing, 210023 China.,3The Nanjing Integrative Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210014 China
| | - Shangbo Hua
- 2The Kunshan Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan, 215300 China
| | - Wen Zhang
- 3The Nanjing Integrative Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210014 China
| | - Chao Shen
- 3The Nanjing Integrative Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210014 China
| | - Xuehua Miao
- 3The Nanjing Integrative Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210014 China
| | - Wensheng Liu
- 3The Nanjing Integrative Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210014 China
| |
Collapse
|
29
|
da Silva JL, Silva-de-Oliveira AFS, Andraus RAC, Maia LP. Effects of low level laser therapy in cancer cells—a systematic review of the literature. Lasers Med Sci 2019; 35:523-529. [DOI: 10.1007/s10103-019-02824-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/01/2019] [Accepted: 05/30/2019] [Indexed: 12/17/2022]
|
30
|
Jeanneau C, Giraud T, Laurent P, About I. BioRoot RCS Extracts Modulate the Early Mechanisms of Periodontal Inflammation and Regeneration. J Endod 2019; 45:1016-1023. [PMID: 31160081 DOI: 10.1016/j.joen.2019.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION The balance between periapical tissue inflammation and regeneration after the removal of necrotic/infected tissues is pivotal in determining the success of endodontic treatment. This study was designed to investigate the effect of silicate-based root canal sealer BioRoot RCS (BRCS; Septodont, Saint-Maur-des-Fossés, France) on modulating the inflammatory mechanisms and early steps of regeneration initiated by human periodontal ligament (PDL) fibroblasts. METHODS Samples of BRCS and Pulp Canal Sealer (PCS; SybronEndo, Orange, CA) were incubated in culture medium to obtain material extracts. To simulate bacterial infection and endodontic sealer use, PDL fibroblasts were stimulated with lipopolysaccharides and cultured with material extracts. The secretion of proinflammatory cytokine (interleukin 6) and growth factor (transforming growth factor beta 1) were quantified by enzyme-linked immunosorbent assay. Inflammatory cell recruitment sequence was investigated using a human inflammatory monocytic cell line (THP-1) that can be activated into macrophage-like cells. The adhesion of THP-1 to endothelial cells (human umbilical vein endothelial cells) was studied using fluorescent THP-1, their migration using Boyden chambers, and their activation into macrophage-like cells using a cell adhesion assay. The proliferation of PDL fibroblasts was quantified by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, whereas the migration of PDL stem cells was investigated using Boyden chambers after immunofluorescence and reverse transcription polymerase chain reaction characterization. RESULTS Interleukin 6 secretion decreased with BRCS, whereas it increased with PCS. Transforming growth factor beta 1 secretion significantly increased only with BRCS. The material extracts did not affect THP-1 adhesion to human umbilical vein endothelial cells, but only BRCS inhibited their migration. Moreover, activation of THP-1 decreased with BRCS and to a lesser extent with PCS. Finally, BRCS increased PDL fibroblast proliferation without affecting PDL stem cell migration. By contrast, PCS decreased PDL fibroblast proliferation and PDL stem cell migration. CONCLUSIONS This work shows that the endodontic sealers modulate the PDL inflammatory and regeneration potentials in vitro. It demonstrates that BRCS has anti-inflammatory effects and the potential to promote tissue regeneration.
Collapse
Affiliation(s)
- Charlotte Jeanneau
- Aix Marseille University, Centre National de la Recherche Scientifique ISM, Institut des Sciences du Mouvement, Marseille, France
| | - Thomas Giraud
- Aix Marseille University, Centre National de la Recherche Scientifique ISM, Institut des Sciences du Mouvement, Marseille, France; Assistance Publique - Hôpitaux de Marseille, Hôpital Timone, Service d'Odontologie, Marseille, France
| | - Patrick Laurent
- Aix Marseille University, Centre National de la Recherche Scientifique ISM, Institut des Sciences du Mouvement, Marseille, France; Assistance Publique - Hôpitaux de Marseille, Hôpital Timone, Service d'Odontologie, Marseille, France
| | - Imad About
- Aix Marseille University, Centre National de la Recherche Scientifique ISM, Institut des Sciences du Mouvement, Marseille, France.
| |
Collapse
|
31
|
Bamps M, Dok R, Nuyts S. Low-Level Laser Therapy Stimulates Proliferation in Head and Neck Squamous Cell Carcinoma Cells. Front Oncol 2018; 8:343. [PMID: 30211121 PMCID: PMC6122283 DOI: 10.3389/fonc.2018.00343] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/06/2018] [Indexed: 12/15/2022] Open
Abstract
Objectives: Low-level laser therapy (LLLT) is a promising non-invasive treatment option for oropharyngeal mucositis, which is a common side effect of many oncological treatments. LLLT is known for its wound healing properties due to the stimulation of cellular processes, such as proliferation, migration and differentiation. Controversy exists on the possible stimulatory effect of LLLT on head and neck cancer (HNSCC) cells in patients treated with radiotherapy. The aim of this study was to evaluate the biostimulatory effect together with the underlying mechanisms of LLLT on HNSCC cancer cells and normal epithelial cells. Materials and methods: HNSCC cell lines (SCC154, SQD9, and SCC61) and human tonsil epithelial cells were exposed to a Gallium-Aluminum-Arsenide diode laser (830 nm, 150 mW) with energy densities of 0, 1, and 2 J/cm2. The proliferation potential of the cells was assessed by Sulforhodamine B assay, immunoblotting (mitogenic pathways), immunocytochemistry (Ki67), and flow cytometry (PI cell cycle staining). Results: Cell proliferation was increased in HNSCC cell lines after laser irradiation with 1 J/cm2, whereas no significant increase was seen after laser irradiation with 2 J/cm2. In contrast, no effect on cell proliferation was seen in the human tonsil epithelial cells after laser irradiation with any of the energy densities. The increased proliferation was associated with elevated levels of pAKT, pERK, and Ki67 protein expression and cell cycle progression. Conclusion: Our results show that LLLT increases cell proliferation in a dose-dependent manner in HNSCC cells but not in normal epithelial tonsil cells. These results suggest that LLLT has to be used with caution when treating oropharyngeal mucositis in HNSCC patients since tumor cells present in the LLLT irradiation field could be triggered by LLLT.
Collapse
Affiliation(s)
- Marieke Bamps
- Laboratory of Experimental Radiotherapy, Department of Oncology, KU Leuven Leuven, Belgium
| | - Rüveyda Dok
- Laboratory of Experimental Radiotherapy, Department of Oncology, KU Leuven Leuven, Belgium
| | - Sandra Nuyts
- Laboratory of Experimental Radiotherapy, Department of Oncology, KU Leuven Leuven, Belgium.,Department of Radiation Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Kara C, Selamet H, Gökmenoğlu C, Kara N. Low level laser therapy induces increased viability and proliferation in isolated cancer cells. Cell Prolif 2017; 51:e12417. [PMID: 29160001 DOI: 10.1111/cpr.12417] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/01/2017] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Low level laser therapy (LLLT), which stimulates natural biological processes in the application region, is frequently used in dental treatments. The aim of our study was to evaluate the effects of LLLT which could activate precancerous cells or increase existing cancerous tissue in case of clinically undetectable situations. MATERIALS AND METHODS Saos-2 osteoblast-like osteosarcoma cells and A549 human lung carcinoma cells were used. Twenty-four hours after preparation of cell culture plates, laser irradiation was performed 1, 2 and 3 times according to the test groups using Nd:YAG laser with the power output 0.5, 1, 2 and 3 W. Cell proliferation analysis was performed by MTT assay at the 24th hour following the last laser applications. RESULTS Generally, it was observed that the proliferation rates increased as the number of applications increased, when compared to the controls, especially in those cases in which the irradiation was performed 2 or 3 times more. CONCLUSION The findings of this study have led to the conclusion that LLLT increases cancer cell proliferation, depending on the power output level of the laser and the number of applications. In addition to the proliferation and mitotic activity of the cancer tissue cells, we concluded that LLLT, which is frequently used in dental practice, could activate precancerous cells or increase existing cancerous tissue.
Collapse
Affiliation(s)
- C Kara
- Department of Periodontology, Faculty of Dentistry, Ordu University, Ordu, Turkey
| | - H Selamet
- Department of Periodontology, Faculty of Dentistry, Ordu University, Ordu, Turkey
| | - C Gökmenoğlu
- Department of Periodontology, Faculty of Dentistry, Ordu University, Ordu, Turkey
| | - N Kara
- Department of Pedodontics, Faculty of Dentistry, Ordu University, Ordu, Turkey
| |
Collapse
|