1
|
Aasy NKA, Sallam MA, Ragab D, Abdelmonsif DA, Aly RG, Abdelfattah EZA, Elkhodairy KA. CD44-targeted hyaluronic acid coated imiquimod lipid nanocapsules foster the efficacy against skin cancer: Attempt to conquer unfavorable side effects. Int J Biol Macromol 2024; 290:138895. [PMID: 39701268 DOI: 10.1016/j.ijbiomac.2024.138895] [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: 05/18/2024] [Revised: 12/03/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
This study was executed to mitigate imiquimod (IMQ)-side effects and promote its anticancer potential against skin cancer via encapsulation in hyaluronic acid-coated lipid nanocapsules (HA-LNCs) for targeted topical delivery. The LNCs were prepared using the phase inversion technique. Optimized LNCs formulation was gained following 22 factorial design experiment to adjust the IMQ and CTAB concentrations. The two variables were found to significantly influence the dependent responses. The encapsulation efficiency of IMQ exceeded 97 %. HA coating provided a sustained release of IMQ from LNCs, with 63.81 ± 2.45 % of IMQ released after 24 h. Moreover, the ex-vivo human skin permeation study showed that 7.9-fold more IMQ was localized in all skin layers than that permeated. In vitro anticancer activity indicated that IMQ-HA-LNCs had higher cytotoxicity (IC50 = 22.39 μg/mL) compared to free IMQ (IC50 = 97.94 μg/mL). Further, in vivo studies revealed that encapsulation of IMQ in HA-LNCs enhanced its immunostimulatory potential and promoted its anti-tumor activity in competing skin cancer even in low doses compared to the untreated group and group treated with a brand product with no topical or systemic toxicity. The present study suggested that HA-LNCs with their mixed polymeric/lipophilic nature epitomize a promising strategy for safe topical delivery of poorly water-soluble candidates.
Collapse
Affiliation(s)
- Noha Khalifa Abo Aasy
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| | - Marwa A Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Doaa Ragab
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Doaa A Abdelmonsif
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, Egypt; Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, University of Alexandria, Egypt
| | - Rania G Aly
- Department of Surgical Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | | | - Kadria A Elkhodairy
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
2
|
Niculet E, Radaschin DS, Arbune M, Bobeica C, Craescu M, Onisor C, Gurau G, Busila C, Vasile CI, Tatu AL. Basal Cell Carcinoma Cleft: The Missing Piece of the Puzzle. Cureus 2024; 16:e71244. [PMID: 39525230 PMCID: PMC11550456 DOI: 10.7759/cureus.71244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND This study aims to explore the tumor-stroma separation or the cleft characterizing basal cell carcinoma (BCC). METHODOLOGY In this retrospective cohort investigation, we enrolled 244 patients who received a confirmed diagnosis of BCC through histopathological examination in the period of 2019-2020 at the Pathology Laboratory of the "Sfântul Apostol Andrei" Emergency Clinical Hospital located in Galați, Romania. The identification of patients was accomplished by utilizing electronic health records, and relevant clinical, demographic, and histopathological data were retrieved from the physical database of the Pathology Laboratory. Key tumor characteristics were gathered, and an in-depth analysis of case slides was performed. RESULTS The average tumor-stroma cleft's width measurement was 48.136 µm, while its respective tumor island's width was on average 952.587 µm. The cleft's width and its respective tumor island's width are dependent on the BCC subtype, just like the ratio between the tumor island's measurement and its cleft's width are, being larger in basosquamous BCC, micronodular BCC, infiltrative BCC, and morpheaform BCC. CONCLUSION The BCC tumor islands were found to have a minimal approximately equal measurement to the tumor-stroma separation cleft, but they were always larger than the latter. Large clefts and their respective tumor islands were found in specific tumor subtypes such as basosquamous BCC, micronodular BCC, infiltrative BCC, and morpheaform BCC, but in nodular BCC also.
Collapse
Affiliation(s)
- Elena Niculet
- Department of Pathology, "Sfantul Apostol Andrei" Emergency Clinical Hospital of Galati, Romania, Galati, ROU
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR), "Dunărea de Jos" University, Galati, ROU
| | - Diana S Radaschin
- Department of Dermatology, Saint Parascheva Infectious Disease Clinical Hospital, Galati, ROU
- Biomedical Doctoral School, "Dunărea de Jos" University, Galati, ROU
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR), "Dunărea de Jos" University, Galati, ROU
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Manuela Arbune
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Carmen Bobeica
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Mihaela Craescu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Cristian Onisor
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Gabriela Gurau
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Camelia Busila
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galați, ROU
| | - Claudiu I Vasile
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| | - Alin L Tatu
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR), "Dunărea de Jos" University, Galati, ROU
- Department of Dermatology, "Sf. Cuvioasa Parascheva" Clinical Hospital of Infectious Diseases, Galati, ROU
- Department of Clinical Medicine, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galati, ROU
| |
Collapse
|
3
|
Chang SH, Chuang KC, Li ZY, Chang MC, Liu KT, Hsu CS, Huang SW, Chung MC, Wang SC, Chen YJ, Shieh JJ. The Protective Effects of Mcl-1 on Mitochondrial Damage and Oxidative Stress in Imiquimod-Induced Cancer Cell Death. Cancers (Basel) 2024; 16:3060. [PMID: 39272918 PMCID: PMC11394135 DOI: 10.3390/cancers16173060] [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: 08/07/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Mitochondria, vital organelles that generate ATP, determine cell fate. Dysfunctional and damaged mitochondria are fragmented and removed through mitophagy, a mitochondrial quality control mechanism. The FDA-approved drug IMQ, a synthetic agonist of Toll-like receptor 7, exhibits antitumor activity against various skin malignancies. We previously reported that IMQ promptly reduced the level of the antiapoptotic Mcl-1 protein and that Mcl-1 overexpression attenuated IMQ-triggered apoptosis in skin cancer cells. Furthermore, IMQ profoundly disrupted mitochondrial function, promoted mitochondrial fragmentation, induced mitophagy, and caused cell death by generating high levels of ROS. However, whether Mcl-1 protects mitochondria from IMQ treatment is still unknown. In this study, we demonstrated that Mcl-1 overexpression induced resistance to IMQ-induced apoptosis and reduced both IMQ-induced ROS generation and oxidative stress in cancer cells. Mcl-1 overexpression maintained mitochondrial function and integrity and prevented mitophagy in IMQ-treated cancer cells. Furthermore, IL-6 protected against IMQ-induced apoptosis by increasing Mcl-1 expression and attenuating IMQ-induced mitochondrial fragmentation. Mcl-1 overexpression ameliorates IMQ-induced ROS generation and mitochondrial fragmentation, thereby increasing mitochondrial stability and ultimately attenuating IMQ-induced cell death. Investigating the roles of Mcl-1 in mitochondria is a potential strategy for cancer therapy development.
Collapse
Affiliation(s)
- Shu-Hao Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Kai-Cheng Chuang
- Department of Life Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Zheng-Yi Li
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Mao-Chia Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
| | - Kuang-Ting Liu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Pathology & Laboratory Medicine, Taoyuan Armed Forces General Hospital, Taoyuan 325208, Taiwan
| | - Chien-Sheng Hsu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Frontier Molecular Medical Research Center in Children, Changhua Christian Children Hospital, Changhua 500209, Taiwan
| | - Shi-Wei Huang
- Center for Cell Therapy and Translation Research, China Medical University Hospital, Taichung 404327, Taiwan
| | - Mu-Chi Chung
- Division of Nephrology, Department of Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- PhD Program in Translational Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Biotechnology, Asia University, Taichung 413305, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Shih-Chung Wang
- Division of Pediatric Hematology/Oncology, Changhua Christian Children Hospital, Changhua 500209, Taiwan
| | - Yi-Ju Chen
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Dermatology, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Jeng-Jer Shieh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Education and Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| |
Collapse
|
4
|
Bopp L, Martinez ML, Schumacher C, Seitz R, Arana MH, Klapproth H, Lukas D, Oh JH, Neumayer D, Lackmann JW, Mueller S, von Stebut E, Brachvogel B, Brodesser S, Klein Geltink RI, Fabri M. Glutamine promotes human CD8 + T cells and counteracts imiquimod-induced T cell hyporesponsiveness. iScience 2024; 27:109767. [PMID: 38736545 PMCID: PMC11088342 DOI: 10.1016/j.isci.2024.109767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 02/24/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
T cells protect tissues from cancer. Although investigations in mice showed that amino acids (AA) critically regulate T cell immunity, this remains poorly understood in humans. Here, we describe the AA composition of interstitial fluids in keratinocyte-derived skin cancers (KDSCs) and study the effect of AA on T cells using models of primary human cells and tissues. Gln contributed to ∼15% of interstitial AAs and promoted interferon gamma (IFN-γ), but not granzyme B (GzB) expression, in CD8+ T cells. Furthermore, the Toll-like receptor 7 agonist imiquimod (IMQ), a common treatment for KDSCs, down-regulated the metabolic gatekeepers c-MYC and mTORC1, as well as the AA transporter ASCT2 and intracellular Gln, Asn, Ala, and Asp in T cells. Reduced proliferation and IFN-γ expression, yet increased GzB, paralleled IMQ effects on AA. Finally, Gln was sufficient to promote IFN-γ-production in IMQ-treated T cells. Our findings indicate that Gln metabolism can be harnessed for treating KDSCs.
Collapse
Affiliation(s)
- Luisa Bopp
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
| | - Maria Lopéz Martinez
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
| | - Clara Schumacher
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
| | - Robert Seitz
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
| | - Manuel Huerta Arana
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
| | - Henning Klapproth
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
| | - Dominika Lukas
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
| | - Ju Hee Oh
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- University of British Columbia, Vancouver, BC, Canada
| | - Daniela Neumayer
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
| | - Jan W. Lackmann
- CECAD Cluster of Excellence, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany
| | - Stefan Mueller
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Esther von Stebut
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
| | - Bent Brachvogel
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Biochemistry, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - Susanne Brodesser
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany
| | - Ramon I. Klein Geltink
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- University of British Columbia, Vancouver, BC, Canada
| | - Mario Fabri
- Department of Dermatology and Venereology, University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
5
|
Li ZY, Chang SH, Liu KT, Wu AE, Hsu CS, Huang SW, Chung MC, Wang SC, Kao JK, Chen YJ, Shieh JJ. Low-dose imiquimod induces melanogenesis in melanoma cells through an ROS-mediated pathway. J Dermatol Sci 2024; 113:18-25. [PMID: 38185543 DOI: 10.1016/j.jdermsci.2023.12.005] [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: 10/06/2022] [Revised: 12/09/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Melanogenesis is the process of melanin maturation which not only protects skin from UV radiation but also plays an important role in antigenicity of melanomas. Imiquimod (IMQ) is a toll-like receptor 7 (TLR7) agonist that exhibits antiviral and anticancer activity. OBJECTIVE To explore whether IMQ could induce melanogenesis in melanoma cells. METHODS The mouse melanoma cell line B16F10, the mouse immortalized melanocyte Melan-A, and human melanoma cell lines MNT-1, C32 and A375 were utilized in this study. The pigmented level was observed by the centrifuged cell pellet. The intracellular and extracellular melanin levels were examined in the absorbance in NaOH-extracted cell lysate and cell-cultured medium, respectively. The expression of melanogenesis related proteins was examined by immunoblotting. The intracellular cyclic AMP amount was evaluated by the cAMP Glo assay kit. The activity of phosphodiesterase 4B (PDE4B) was investigated by CREB reporter assay with overexpressed PDE4B or not. RESULTS We demonstrated that a low dose of IMQ could trigger melanogenesis in B16F10 cells. IMQ induced microphthalmia-associated transcription factor (MITF) nuclear translocation, upregulated the expression of melanogenesis-related proteins, increased tyrosinase (TYR) activity, and led to pigmentation in B16F10 cells. Next, we found that IMQ-induced melanogenesis was activated by excessive intracellular cAMP accumulation, which was regulated through IMQ-mediated PDE4B inhibition. Finally, IMQ-induced ROS production was found to be involved in melanogenesis by its control of PDE4B activity. CONCLUSIONS Low dose of IMQ could activate melanogenesis through the ROS/PDE4B/PKA pathway in melanoma cells.
Collapse
Affiliation(s)
- Zheng-Yi Li
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Hao Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Kuang-Ting Liu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan; Department of Pathology & Laboratory Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Alaina Edelie Wu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chien-Sheng Hsu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan; Frontier Molecular Medical Research Center in Children, Changhua Christian Children Hospital, Changhua County, Taiwan
| | - Shi-Wei Huang
- Center for Cell Therapy and Translation Research, China Medical University Hospital, Taichung, Taiwan
| | - Mu-Chi Chung
- Division of Nephrology, Department of Medicine, Taichung Veterans General Hospital, Taichung, Taiwan; PhD Program in Translational Medicine, National Chung Hsing University, Taichung, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Shih-Chung Wang
- Division of Pediatric Hematology/Oncology, Changhua Christian Children Hospital, Changhua County, Taiwan
| | - Jun-Kai Kao
- Frontier Molecular Medical Research Center in Children, Changhua Christian Children Hospital, Changhua County, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ju Chen
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan; Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jeng-Jer Shieh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan; Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan.
| |
Collapse
|
6
|
Chang SH, Lin PY, Wu TK, Hsu CS, Huang SW, Li ZY, Liu KT, Kao JK, Chen YJ, Wong TW, Wu CY, Shieh JJ. Imiquimod-induced ROS production causes lysosomal membrane permeabilization and activates caspase-8-mediated apoptosis in skin cancer cells. J Dermatol Sci 2022; 107:142-150. [PMID: 36075780 DOI: 10.1016/j.jdermsci.2022.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 08/12/2022] [Accepted: 08/23/2022] [Indexed: 10/15/2022]
Abstract
BACKGROUND Lysosomal cell death is induced by lysosomal membrane permeabilization (LMP) and the subsequent release of lysosomal proteolytic enzymes, including cathepsins (CTSs), which results in mitochondrial dysfunction and apoptosis. Imiquimod (IMQ), a synthetic TLR7 ligand, has both antiviral and antitumor activity against various skin malignancies in clinical treatment. Previously, we demonstrated IMQ not only caused lysosomal dysfunction but also triggered lysosome biogenesis to achieve lysosomal adaptation in cancer cells. OBJECTIVE To determine whether lysosomes are involved in IMQ-induced apoptosis. METHODS The human skin cancer cell lines BCC, A375 and mouse melanoma cell line B16F10 were used in all experiments. Cell death was determined by the Cell Counting Kit-8 (CCK-8) assay and DNA content assay. Protein expression was determined by immunoblotting. Caspase-8 activity was assessed using a fluorescence caspase-8 kit and determined by flow cytometry and confocal microscopy. RESULTS IMQ not only induced lysosome damage but also abrogated lysosome function in skin cancer cells. IMQ-induced caspase-8 activation contributed to the processes of lysosomal cell death. Moreover, the use of ROS scavengers significantly abolished caspase-8 activation and inhibited IMQ-induced LMP. Additionally, pharmacological inhibition of CTSD not only abrogated caspase-8 activation but also rescued IMQ-induced cell death. Finally, lysosome-alkalizing agents enhanced the cytotoxicity of IMQ in vitro and in vivo. CONCLUSIONS IMQ-induced ROS accumulation promotes LMP, releases CTSs into the cytosol, stimulates caspase-8 activation and finally causes lysosomal cell death. Lysosomal cell death and the CTSD/caspase-8 axis may play a crucial role in IMQ-induced cell death.
Collapse
Affiliation(s)
- Shu-Hao Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Pei-Ying Lin
- Center for Cell Therapy and Translation Research, China Medical University Hospital, Taichung, Taiwan
| | - Tsai-Kun Wu
- Division of Renal Medicine, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan; Post Baccalaureate Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chien-Sheng Hsu
- Frontier Molecular Medical Research Center in Children, Changhua Christian Children Hospital, Changhua County, Taiwan
| | - Shi-Wei Huang
- Center for Cell Therapy and Translation Research, China Medical University Hospital, Taichung, Taiwan
| | - Zheng-Yi Li
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Kuang-Ting Liu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan; Department of Pathology & Laboratory Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Jun-Kai Kao
- Post Baccalaureate Medicine, National Chung Hsing University, Taichung, Taiwan; Frontier Molecular Medical Research Center in Children, Changhua Christian Children Hospital, Changhua County, Taiwan
| | - Yi-Ju Chen
- Post Baccalaureate Medicine, National Chung Hsing University, Taichung, Taiwan; Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tak-Wah Wong
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Ying Wu
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Jeng-Jer Shieh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan; Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
7
|
Sun X, Huang X, Sun X, Chen S, Zhang Z, Yu Y, Zhang P. Oxidative Stress-Related lncRNAs Are Potential Biomarkers for Predicting Prognosis and Immune Responses in Patients With LUAD. Front Genet 2022; 13:909797. [PMID: 35754800 PMCID: PMC9214656 DOI: 10.3389/fgene.2022.909797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/04/2022] [Indexed: 12/24/2022] Open
Abstract
Lung adenocarcinoma is increasingly harmful to society and individuals as cancer with an inferior prognosis and insensitive to chemotherapy. Previous studies have demonstrated that oxidative stress and lncRNAs play a vital role in many biological processes. Therefore, we explored the role of lncRNAs associated with oxidative stress in the prognosis and survival of LUAD patients. We examined the expression profiles of lncRNAs and oxidative stress genes in this study. A prognosis prediction model and a nomogram were built based on oxidative stress-related lncRNAs. Functional and drug sensitivity analyses were also performed depending on oxidative stress-related lncRNA signature. Moreover, we investigated the relationship between immune response and immunotherapy. The results showed that a risk scoring model based on 16 critical oxidative stress lncRNAs was able to distinguish the clinical status of LUAD and better predict the prognosis and survival. Additionally, the model demonstrated a close correlation with the tumor immune system, and these key lncRNAs also revealed the relationship between LUAD and chemotherapeutic drug sensitivity. Our work aims to provide new perspectives and new ideas for the treatment and management of LUAD.
Collapse
Affiliation(s)
- Xinti Sun
- Department of Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xingqi Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaojuan Sun
- Department of Oncology, Qingdao University Affiliated Hospital, Qingdao, China
| | - Si Chen
- Department of Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Zeyang Zhang
- Department of Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yao Yu
- Department of Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Zhang
- Department of Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
8
|
Rostamizadeh L, Molavi O, Rashid M, Ramazani F, Baradaran B, Lavasanaifar A, Lai R. Recent advances in cancer immunotherapy: Modulation of tumor microenvironment by Toll-like receptor ligands. BIOIMPACTS : BI 2022; 12:261-290. [PMID: 35677663 PMCID: PMC9124882 DOI: 10.34172/bi.2022.23896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/18/2022]
Abstract
![]()
Immunotherapy is considered a promising approach for cancer treatment. An important strategy for cancer immunotherapy is the use of cancer vaccines, which have been widely used for cancer treatment. Despite the great potential of cancer vaccines for cancer treatment, their therapeutic effects in clinical settings have been limited. The main reason behind the lack of significant therapeutic outcomes for cancer vaccines is believed to be the immunosuppressive tumor microenvironment (TME). The TME counteracts the therapeutic effects of immunotherapy and provides a favorable environment for tumor growth and progression. Therefore, overcoming the immunosuppressive TME can potentially augment the therapeutic effects of cancer immunotherapy in general and therapeutic cancer vaccines in particular. Among the strategies developed for overcoming immunosuppression in TME, the use of toll-like receptor (TLR) agonists has been suggested as a promising approach to reverse immunosuppression. In this paper, we will review the application of the four most widely studied TLR agonists including agonists of TLR3, 4, 7, and 9 in cancer immunotherapy.
Collapse
Affiliation(s)
- Leila Rostamizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ommoleila Molavi
- Biotechnology Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ramazani
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Lavasanaifar
- Faculty of Pharmacy and Pharmaceutical Science, University of Alberta, Edmonton, Canada
| | - Raymond Lai
- Department of Laboratory Medicine & Pathology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
9
|
Excited-State Dynamics of Imiquimod in Aqueous Solutions. J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2022.113998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
10
|
Akesolo O, Buey B, Beltrán-Visiedo M, Giraldos D, Marzo I, Latorre E. Toll-like receptors: new targets for multiple myeloma treatment? Biochem Pharmacol 2022; 199:114992. [DOI: 10.1016/j.bcp.2022.114992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 02/08/2023]
|
11
|
Yamaguchi R, Sakamoto A, Yamaguchi R, Haraguchi M, Narahara S, Sugiuchi H, Yamaguch Y. IL-23 production in human macrophages is regulated negatively by tumor necrosis factor α-induced protein 3 and positively by specificity protein 1 after stimulation of the toll-like receptor 7/8 signaling pathway. Heliyon 2022; 8:e08887. [PMID: 35198762 PMCID: PMC8850731 DOI: 10.1016/j.heliyon.2022.e08887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/07/2021] [Accepted: 01/30/2022] [Indexed: 11/19/2022] Open
Abstract
The IL-23/IL-17 axis plays an important role in the development of autoimmune diseases, but the mechanism regulating IL-23 production is mainly unknown. We investigated how TNFAIP3 and Sp1 affect IL-23 production by human macrophages after exposure to resiquimod, a TLR7/8 agonist. IL-23 production was significantly upregulated by resiquimod but only slightly by LPS (a TLR4 agonist). Interestingly, IL-23 levels were significantly attenuated after sequential stimulation with LPS and resiquimod, but IL-12p40 and IL-18 levels were not. TLR4-related factors induced by LPS may regulate IL-23 expression via TLR7/8 signaling. LPS significantly enhanced TNFAIP3 and IRAK-M levels but reduced Sp1 levels. After exposure to resiquimod, RNA interference of TNFAIP3 upregulated IL-23 significantly more than siRNA transfection of IRAK-M did. In contrast, knockdown of Sp1 by RNA interference significantly attenuated IL-23 production. Transfection with siRNA for TNFAIP3 enhanced IL-23 expression significantly. After stimulation with resiquimod, GW7647—an agonist for PPARα (an inducer of NADHP oxidase)—and siRNA for UCP2 (a negative regulator of mitochondrial ROS generation) enhanced TNFAIP3 and reduced IL-23. siRNA for p22phox and gp91phox slightly increased Sp1 levels. However, after exposure to resiquimod siRNA-mediated knockout of DUOX1/2 significantly enhanced Sp1 and IL-23 levels, and decreased TNFα-dependent COX-2 expression. Concomitantly, TNFAIP3 levels was attenuated by DUOX1/2 siRNA. TNFAIP3 and Sp1 levels are reciprocally regulated through ROS generation. In conclusion, after stimulation of the TLR7/8 signaling pathway IL-23 production in human macrophages is regulated negatively by TNFAIP3.
Collapse
Affiliation(s)
- Rui Yamaguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kitaku Izumi-machi 325, Kumamoto 861-5598, Japan
| | - Arisa Sakamoto
- Graduate School of Medical Science, Kumamoto Health Science University, Kitaku Izumi-machi 325, Kumamoto 861-5598, Japan
| | - Reona Yamaguchi
- Department of Neuroscience, Graduate School of Medicine and Faculty of Medicine, Kyoto University, Yoshida-konoe-cho Sakyo-ku Kyoto 606-8501, Japan
| | - Misa Haraguchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kitaku Izumi-machi 325, Kumamoto 861-5598, Japan
| | - Shinji Narahara
- Graduate School of Medical Science, Kumamoto Health Science University, Kitaku Izumi-machi 325, Kumamoto 861-5598, Japan
| | - Hiroyuki Sugiuchi
- Graduate School of Medical Science, Kumamoto Health Science University, Kitaku Izumi-machi 325, Kumamoto 861-5598, Japan
| | - Yasuo Yamaguch
- Graduate School of Medical Science, Kumamoto Health Science University, Kitaku Izumi-machi 325, Kumamoto 861-5598, Japan
- Corresponding author.
| |
Collapse
|
12
|
Rocco R, Cambindo Botto AE, Muñoz MJ, Reingruber H, Wainstok R, Cochón A, Gazzaniga S. Early redox homeostasis disruption contributes to the differential cytotoxicity of imiquimod on transformed and normal endothelial cells. Exp Dermatol 2021; 31:608-614. [PMID: 34758172 DOI: 10.1111/exd.14499] [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: 03/26/2021] [Revised: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 11/28/2022]
Abstract
The off-label use of imiquimod (IQ) for hemangioma treatment has shown clinical benefits. We have previously reported a selective direct IQ-cytotoxic effect on transformed (H5V) vs. normal (1G11) endothelial cells (EC). In the present study, we investigated the mechanism underlying this selective cytotoxicity in terms of TLR7/8 receptor expression, NF-κB signalling and time-dependent modifications of oxidative stress parameters (ROS: reactive oxygen species, catalase and superoxide dismutase activities, GSH/GSSG and lipid peroxidation). TLR7/8 level was extremely low in both cell lines, and IQ did not upregulate TLR7/8 expression or activate NF-κB signalling. IQ significantly induced ROS in H5V after 2 h and strongly affected antioxidant defenses. After 12 h, enzyme activities were restored to baseline levels but a robust drop in GSH/GSSG persisted together with increased lipid peroxidation levels and a marked mitochondrial dysfunction. Although in normal IQ-treated EC some oxidative stress parameters were affected after 4 h, mitochondrial health and GSH/GSSG ratio remained notably unaffected after 12 h. Therefore, the early alterations (0-2 h) in transformed EC breached redox homeostasis as strongly as to enhance their susceptibility to IQ. This interesting facet of IQ as redox disruptor could broaden its therapeutic potential for other skin malignancies, alone or in adjuvant schemes.
Collapse
Affiliation(s)
- Rodrigo Rocco
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adrián E Cambindo Botto
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - Manuel J Muñoz
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina.,Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy.,Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Hernán Reingruber
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rosa Wainstok
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adriana Cochón
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvina Gazzaniga
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
13
|
Li Y, Gao S, Du X, Ji J, Xi Y, Zhai G. Advances in autophagy as a target in the treatment of tumours. J Drug Target 2021; 30:166-187. [PMID: 34319838 DOI: 10.1080/1061186x.2021.1961792] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Autophagy is a multi-step lysosomal degradation process, which regulates energy and material metabolism and has been used to maintain homeostasis. Autophagy has been shown to be involved in the regulation of health and disease. But at present, there is no consensus on the relationship between autophagy and tumour, and we consider that it plays a dual role in the occurrence and development of tumour. That is to say, under certain conditions, it can inhibit the occurrence of tumour, but it can also promote the process of tumour. Therefore, autophagy could be used as a target for tumour treatment. The regulation of autophagy plays a synergistic role in the radiotherapy, chemotherapy, phototherapy and immunotherapy of tumour, and nano drug delivery system provides a promising strategy for improving the efficacy of autophagy regulation. This review summarised the progress in the regulatory pathways and factors of autophagy as well as nanoformulations as carriers for the delivery of autophagy modulators.
Collapse
Affiliation(s)
- Yingying Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Shan Gao
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xiyou Du
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Yanwei Xi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
14
|
Bhagchandani S, Johnson JA, Irvine DJ. Evolution of Toll-like receptor 7/8 agonist therapeutics and their delivery approaches: From antiviral formulations to vaccine adjuvants. Adv Drug Deliv Rev 2021; 175:113803. [PMID: 34058283 PMCID: PMC9003539 DOI: 10.1016/j.addr.2021.05.013] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 02/07/2023]
Abstract
Imidazoquinoline derivatives (IMDs) and related compounds function as synthetic agonists of Toll-like receptors 7 and 8 (TLR7/8) and one is FDA approved for topical antiviral and skin cancer treatments. Nevertheless, these innate immune system-activating drugs have potentially much broader therapeutic utility; they have been pursued as antitumor immunomodulatory agents and more recently as candidate vaccine adjuvants for cancer and infectious disease. The broad expression profiles of TLR7/8, poor pharmacokinetic properties of IMDs, and toxicities associated with systemic administration, however, are formidable barriers to successful clinical translation. Herein, we review IMD formulations that have advanced to the clinic and discuss issues related to biodistribution and toxicity that have hampered the further development of these compounds. Recent strategies aimed at enhancing safety and efficacy, particularly through the use of bioconjugates and nanoparticle formulations that alter pharmacokinetics, biodistribution, and cellular targeting, are described. Finally, key aspects of the biology of TLR7 signaling, such as TLR7 tolerance, that may need to be considered in the development of new IMD therapeutics are discussed.
Collapse
Affiliation(s)
- Sachin Bhagchandani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Jeremiah A Johnson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
15
|
In vitro study on immune response modifiers as novel medical treatment options for cholesteatoma. Int J Pediatr Otorhinolaryngol 2021; 145:110743. [PMID: 33933986 DOI: 10.1016/j.ijporl.2021.110743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/24/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVES To investigate cytokine profile of cholesteatoma and to collect information about important intercellular signaling pathways by establishing two different cell culture models, to block important intercellular signaling pathways in cholesteatoma by applying immune system modifier drugs to develop alternative medical therapy options for cholesteatoma. METHODS To observe the pathogenesis of cholesteatoma and to apply the immunomodulatory drugs, cholesteatoma tissue culture models were constituted with HEKa cells and cholesteatoma keratinocytes, which were obtained from 3 patients who underwent operations for cholesteatoma. Medicines including 5-fluorourasil, imiquimod, cyclosporine, and tacrolimus were applied on both cholesteatoma keratinocytes and HEKa cells. After 48 h of incubation, IL-1, IL-6, IL-8, IL-10, TNF-α, and Ki67 levels were measured to determine cell viability rates. RESULTS In the cholesteatoma control group, IL-6 and TNF-α levels were found higher than in the HEKa control group. All repurposed drugs in the study demonstrated anti-inflammatory, anti-proliferative, and cytotoxic effects on cholesteatoma. Imiquimod and tacrolimus in particular are potential treatment prospects for cholesteatoma due to their strong anti-inflammatory and cytotoxic effects. CONCLUSION Medical therapy options for cholesteatoma are still missing and surgery is not the ultimate solution. We have focused on intercellular inflammatory processes, which play significant roles in the pathogenesis of cholesteatoma in our paper. Inflammation and proliferation of cholesteatoma decreased after all repurposed drug applications in our study. Anti-inflammatory and anti-proliferative effects of tacrolimus and imiquimod was more significant than other drugs in the study. For this reason, tacrolimus and imiquimod should be examined in depth with in vivo studies in terms of efficacy and safety for medical treatment of cholesteatoma.
Collapse
|
16
|
Chang SH, Wu CY, Chuang KC, Huang SW, Li ZY, Wang ST, Lai ZL, Chang CC, Chen YJ, Wong TW, Kao JK, Shieh JJ. Imiquimod Accelerated Antitumor Response by Targeting Lysosome Adaptation in Skin Cancer Cells. J Invest Dermatol 2021; 141:2219-2228.e8. [PMID: 33744296 DOI: 10.1016/j.jid.2021.01.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/01/2021] [Accepted: 01/11/2021] [Indexed: 01/01/2023]
Abstract
Lysosomal adaptation is a cellular physiological process in which the number and function of lysosomes are regulated at the transcriptional and post-transcriptional levels in response to extracellular and/or intracellular cues or lysosomal damage. Imiquimod (IMQ), a synthetic toll-like receptor 7 ligand with hydrophobic and weak basic properties, exhibits both antitumor and antiviral activity against various skin malignancies as a clinical treatment. Interestingly, IMQ has been suggested to be highly concentrated in the lysosomes of plasmacytoid dendritic cells, indicating that IMQ could modulate lysosome function after sequestration in the lysosome. In this study, we found that IMQ not only induced lysosomal membrane permeabilization and dysfunction but also increased lysosome biogenesis to achieve lysosomal adaptation in cancer cells. IMQ-induced ROS production but not lysosomal sequestration of IMQ was the major cause of lysosomal adaptation. Moreover, IMQ-induced lysosomal adaptation occurred through lysosomal calcium ion release and activation of the calcineurin/TFEB axis to promote lysosome biogenesis. Finally, depletion of TFEB sensitized skin cancer cells to IMQ-induced apoptosis in vitro and in vivo. In summary, a disruption of lysosomal adaptation might represent a therapeutic strategy for synergistically enhancing the cytotoxicity of IMQ in skin cancer cells.
Collapse
Affiliation(s)
- Shu-Hao Chang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Ying Wu
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kai-Cheng Chuang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shi-Wei Huang
- Center for Cell Therapy and Translation Research, China Medical University Hospital, Taichung, Taiwan
| | - Zheng-Yi Li
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Sin-Ting Wang
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Zi-Lun Lai
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, Taiwan
| | - Cheng-Chung Chang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ju Chen
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Tak-Wah Wong
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jun-Kai Kao
- Department of Pediatrics, Children's Hospital, Changhua Christian Hospital, Changhua, Taiwan
| | - Jeng-Jer Shieh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan; Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
17
|
Ernst O, Failayev H, Athamna M, He H, Tsfadia Y, Zor T. A dual and conflicting role for imiquimod in inflammation: A TLR7 agonist and a cAMP phosphodiesterase inhibitor. Biochem Pharmacol 2020; 182:114206. [DOI: 10.1016/j.bcp.2020.114206] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/10/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022]
|
18
|
Hübbe ML, Jæhger DE, Andresen TL, Andersen MH. Leveraging Endogenous Dendritic Cells to Enhance the Therapeutic Efficacy of Adoptive T-Cell Therapy and Checkpoint Blockade. Front Immunol 2020; 11:578349. [PMID: 33101304 PMCID: PMC7546347 DOI: 10.3389/fimmu.2020.578349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/26/2020] [Indexed: 01/15/2023] Open
Abstract
Adoptive cell therapy (ACT), based on treatment with autologous tumor infiltrating lymphocyte (TIL)-derived or genetically modified chimeric antigen receptor (CAR) T cells, has become a potentially curative therapy for subgroups of patients with melanoma and hematological malignancies. To further improve response rates, and to broaden the applicability of ACT to more types of solid malignancies, it is necessary to explore and define strategies that can be used as adjuvant treatments to ACT. Stimulation of endogenous dendritic cells (DCs) alongside ACT can be used to promote epitope spreading and thereby decrease the risk of tumor escape due to target antigen downregulation, which is a common cause of disease relapse in initially responsive ACT treated patients. Addition of checkpoint blockade to ACT and DC stimulation might further enhance response rates by counteracting an eventual inactivation of infused and endogenously primed tumor-reactive T cells. This review will outline and discuss therapeutic strategies that can be utilized to engage endogenous DCs alongside ACT and checkpoint blockade, to strengthen the anti-tumor immune response.
Collapse
Affiliation(s)
- Mie Linder Hübbe
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Copenhagen, Denmark
| | - Ditte Elisabeth Jæhger
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Copenhagen, Denmark
| |
Collapse
|
19
|
Recent Advances in Signaling Pathways Comprehension as Carcinogenesis Triggers in Basal Cell Carcinoma. J Clin Med 2020; 9:jcm9093010. [PMID: 32961989 PMCID: PMC7565128 DOI: 10.3390/jcm9093010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 12/14/2022] Open
Abstract
Basal cell carcinoma (BCC) is the most common malignant skin tumor. BCC displays a different behavior compared with other neoplasms, has a slow evolution, and metastasizes very rarely, but sometimes it causes an important local destruction. Chronic ultraviolet exposure along with genetic factors are the most important risk factors involved in BCC development. Mutations in the PTCH1 gene are associated with Gorlin syndrome, an autosomal dominant disorder characterized by the occurrence of multiple BCCs, but are also the most frequent mutations observed in sporadic BCCs. PTCH1 encodes for PTCH1 protein, the most important negative regulator of the Hedgehog (Hh) pathway. There are numerous studies confirming Hh pathway involvement in BCC pathogenesis. Although Hh pathway has been intensively investigated, it remains incompletely elucidated. Recent studies on BCC tumorigenesis have shown that in addition to Hh pathway, there are other signaling pathways involved in BCC development. In this review, we present recent advances in BCC carcinogenesis.
Collapse
|
20
|
Siddiqui SS, Rahman S, Rupasinghe HV, Vazhappilly CG. Dietary Flavonoids in p53-Mediated Immune Dysfunctions Linking to Cancer Prevention. Biomedicines 2020; 8:biomedicines8080286. [PMID: 32823757 PMCID: PMC7460013 DOI: 10.3390/biomedicines8080286] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
The p53 protein plays a central role in mediating immune functioning and determines the fate of the cells. Its role as a tumor suppressor, and in transcriptional regulation and cytokine activity under stress conditions, is well defined. The wild type (WT) p53 functions as a guardian for the genome, while the mutant p53 has oncogenic roles. One of the ways that p53 combats carcinogenesis is by reducing inflammation. WT p53 functions as an anti-inflammatory molecule via cross-talk activity with multiple immunological pathways, such as the major histocompatibility complex I (MHCI) associated pathway, toll-like receptors (TLRs), and immune checkpoints. Due to the multifarious roles of p53 in cancer, it is a potent target for cancer immunotherapy. Plant flavonoids have been gaining recognition over the last two decades to use as a potential therapeutic regimen in ameliorating diseases. Recent studies have shown the ability of flavonoids to suppress chronic inflammation, specifically by modulating p53 responses. Further, the anti-oxidant Keap1/Nrf2/ARE pathway could play a crucial role in mitigating oxidative stress, leading to a reduction of chronic inflammation linked to the prevention of cancer. This review aims to discuss the pharmacological properties of plant flavonoids in response to various oxidative stresses and immune dysfunctions and analyzes the cross-talk between flavonoid-rich dietary intake for potential disease prevention.
Collapse
Affiliation(s)
- Shoib Sarwar Siddiqui
- Department of Biotechnology, American University of Ras Al Khaimah, Ras Al Khaimah PO Box 10021, UAE;
| | - Sofia Rahman
- School of Natural Sciences and Mathematics, The University of Texas at Dallas, Richardson, TX 75080, USA;
| | - H.P. Vasantha Rupasinghe
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada;
- Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Cijo George Vazhappilly
- Department of Biotechnology, American University of Ras Al Khaimah, Ras Al Khaimah PO Box 10021, UAE;
- Correspondence:
| |
Collapse
|
21
|
Identification of Potential Biomarkers Associated with Basal Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2073690. [PMID: 32382535 PMCID: PMC7189327 DOI: 10.1155/2020/2073690] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 02/02/2020] [Accepted: 03/26/2020] [Indexed: 12/20/2022]
Abstract
Purpose This work is aimed at identifying several molecular markers correlated with the diagnosis and development of basal cell carcinoma (BCC). Methods The available microarray datasets for BCC were obtained from the Gene Expression Omnibus (GEO) database, and differentially expressed genes (DEGs) were identified between BCC and healthy controls. Afterward, the functional enrichment analysis and protein-protein interaction (PPI) network analysis of these screened DEGs were performed. An external validation for the DEG expression level was also carried out, and receiver operating characteristic curve analysis was used to evaluate the diagnostic values of DEGs. Result In total, five microarray datasets for BCC were downloaded and 804 DEGs (414 upregulated and 390 downregulated genes) were identified. Functional enrichment analysis showed that these genes including CYFIP2, HOXB5, EGFR, FOXN3, PTPN3, CDC20, MARCKSL1, FAS, and PTCH1 were closely correlated with the cell process and PTCH1 played central roles in the BCC signaling pathway. Moreover, EGFR was a hub gene in the PPI network. The expression changes of six genes (CYFIP2, HOXB5, FOXN3, PTPN3, MARCKSL1, and FAS) were validated by an external GSE74858 dataset analysis. Finally, ROC analysis revealed that CYFIP2, HOXB5, PTPN3, MARCKSL1, PTCH1, and CDC20 could distinguish BCC and healthy individuals. Conclusion Nine gene signatures (CYFIP2, HOXB5, EGFR, FOXN3, PTPN3, CDC20, MARCKSL1, FAS, and PTCH1) may serve as promising targets for BCC detection and development.
Collapse
|
22
|
Chuang KC, Chang CR, Chang SH, Huang SW, Chuang SM, Li ZY, Wang ST, Kao JK, Chen YJ, Shieh JJ. Imiquimod-induced ROS production disrupts the balance of mitochondrial dynamics and increases mitophagy in skin cancer cells. J Dermatol Sci 2020; 98:152-162. [PMID: 32376151 DOI: 10.1016/j.jdermsci.2020.03.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/10/2020] [Accepted: 03/31/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Mitochondrial homeostasis is a highly dynamic process involving continuous fission and fusion cycles and mitophagy to maintain mitochondrial functionality. Imiquimod (IMQ), a Toll-like receptor (TLR) 7 ligand, is used to treat various skin malignancies. IMQ also induces apoptotic and autophagic cell death in various cancers through a TLR7-independent pathway. OBJECTIVE To investigate whether IMQ-induced ROS production is involved in mitochondrial dysfunction, mitochondrial fragmentation and mitophagy in skin cancer cells. METHODS BCC/KMC-1, B16F10 and A375 skin cancer cells, AGS gastric cancer cells and primary human keratinocytes were treated with 50 μg/mL IMQ. After 4 h, ROS were detected by CM-H2DCFDA, DHE, and MitoSOX Red staining. After 24 h, cell viability and the mitochondrial membrane potential were evaluated by a CCK-8 assay and JC-1 staining, respectively. Oxygen consumption was assessed with an Oroboros instrument. Mitochondrial morphology and mitophagy were evaluated by MitoTracker and LysoTracker staining. Mitochondrial dynamics markers, including MFN-1, DRP-1 and OPA1, and mitophagy markers, including LC3, S65-phosphorylated ubiquitin, PINK1 and TOM20, were detected by immunoblotting. RESULTS IMQ not only induced severe ROS production but also resulted in increased mitochondrial membrane potential loss, mitochondrial fission and mitophagy and decreased oxygen consumption in skin cancer cells compared with normal keratinocytes. Pretreatment with the antioxidant NAC reduced IMQ-induced ROS production and attenuated IMQ-induced mitochondrial fission and mitophagy in skin cancer cells. CONCLUSIONS IMQ-induced ROS might be associated with mitochondrial dysfunction, mitochondrial fission and mitophagy in cancer cells. Alleviating IMQ-induced ROS production would reduce mitochondrial fission-to-fusion skewing and further reduce IMQ-induced mitophagy.
Collapse
Affiliation(s)
- Kai-Cheng Chuang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chuang-Rung Chang
- Institute of Biotechnology, National Tsing Hua University, Hsin Chu, Taiwan
| | - Shu-Hao Chang
- Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan
| | - Shi-Wei Huang
- Center for Cell Therapy and Translation Research, China Medical University Hospital, Taichung, Taiwan
| | - Show-Mei Chuang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Zheng-Yi Li
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Sin-Ting Wang
- Division of Translational Research and Center of Excellence for Cancer Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jun-Kai Kao
- Department of Pediatrics, Children's Hospital, Changhua Christian Hospital, Changhua, Taiwan; School of Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Yi-Ju Chen
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jeng-Jer Shieh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan; Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan; Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
23
|
Yang Y, Feng R, Wang YZ, Sun HW, Zou QM, Li HB. Toll-like receptors: Triggers of regulated cell death and promising targets for cancer therapy. Immunol Lett 2020; 223:1-9. [PMID: 32311408 DOI: 10.1016/j.imlet.2020.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/15/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023]
Abstract
Toll-like receptors (TLRs) belong to a family of pattern recognition receptors (PRRs). It is well known that TLRs play an essential role in activating innate and adaptive immune responses. TLRs are involved in mediating inflammatory responses and maintaining epithelial barrier homeostasis, and they are highly likely to activate various signalling pathways during cancer chemotherapy. For a long time, much research focused on the immune modulating function of TLRs in cancer genesis, pathology and therapeutic strategies. However, recent reports have suggested that except for the innate and adaptive immune responses that they initiate, TLRs can signal to induce regulated cell death (RCD), which also plays an important role in the antitumor process. TLR agonists also have been investigated as cancer therapeutic agents under clinical evaluation. In this review, we focused on the mechanism of RCD induced by TLR signals and the important role that they play in anticancer therapy combined with recent experimental and clinical trial data to discuss the possibility of TLRs as promising targets for cancer therapy. TLRs represent triggers of regulated cell death and targets for cancer therapy. The molecular mechanisms of TLR-induced RCD and relationship between TLR-signalling pathways and cancer remain to be investigated by further studies.
Collapse
Affiliation(s)
- Yun Yang
- National Engineering Research Center of Immunological Products & Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, PR China
| | - Rang Feng
- National Engineering Research Center of Immunological Products & Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, PR China
| | - Yuan-Zhong Wang
- City of Hope National Medical Center and Beckman Research Institute, 1500 East Duarte Road, Duarte, CA 91010, USA
| | - Hong-Wu Sun
- National Engineering Research Center of Immunological Products & Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, PR China
| | - Quan-Ming Zou
- National Engineering Research Center of Immunological Products & Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, PR China.
| | - Hai-Bo Li
- National Engineering Research Center of Immunological Products & Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, PR China.
| |
Collapse
|
24
|
Imiquimod Exerts Antitumor Effects by Inducing Immunogenic Cell Death and Is Enhanced by the Glycolytic Inhibitor 2-Deoxyglucose. J Invest Dermatol 2020; 140:1771-1783.e6. [PMID: 32035924 DOI: 10.1016/j.jid.2019.12.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/20/2019] [Accepted: 12/11/2019] [Indexed: 12/31/2022]
Abstract
The induction of immunogenic cell death (ICD) in cancer cells triggers specific immune responses against the same cancer cells. Imiquimod (IMQ) is a synthetic ligand of toll-like receptor 7 that exerts antitumor activity by stimulating cell-mediated immunity or by directly inducing apoptosis. Whether IMQ causes tumors to undergo ICD and elicits a specific antitumor immune response is unknown. We demonstrated that IMQ-induced ICD-associated features, including the surface exposure of calreticulin and the secretion of adenosine triphosphate and HMGB1, were mediated by ROS and endoplasmic reticulum stress. In a B16F10 melanoma mouse model, vaccinating mice with IMQ-induced ICD cell lysate or directly injecting IMQ in situ reduced tumor growth that was mediated by inducing tumor-specific T-cell proliferation, promoting tumor-specific cytotoxic killing by CD8+ T cells, and increasing the infiltration of various immune cells into tumor lesions. The ICD-associated features were crucial in the induction of specific antitumor immunity in vivo. The glycolytic inhibitor 2-deoxyglucose enhanced IMQ-induced ICD-associated features and strengthened the antitumor immunity mediated by IMQ-induced ICD cell lysate in p53-mutant cancer cells, which were IMQ-resistant in vitro. We conclude that IMQ is an authentic ICD inducer and provide a concept connecting IMQ-induced cancer cell death and antitumor immune responses.
Collapse
|
25
|
Frank LA, Gazzi RP, Mello PA, Chaves P, Peña F, Beck RCR, Buffon A, Pohlmann AR, Guterres SS. Anti-HPV Nanoemulsified-Imiquimod: A New and Potent Formulation to Treat Cervical Cancer. AAPS PharmSciTech 2020; 21:54. [PMID: 31907712 DOI: 10.1208/s12249-019-1558-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/06/2019] [Indexed: 11/30/2022] Open
Abstract
Cervical cancer is associated with the human papilloma virus (HPV) and nowadays is the fourth most frequent cancer among women. One of the treatments for this disease is based on the application of imiquimod. In this study, we postulated that the use of imiquimod in nanoemulsion results in a better antitumoral effect than the drug administered in its nonencapsulated form for the treatment of cervical cancer. Permeability studies using vaginal mucosa, as membrane, and in vitro studies involving cervical cancer cells (viability, clonogenic assay, and cell death analysis) were performed. We showed that low amount of encapsulated imiquimod permeated the vaginal mucosa. However, a higher percentage of cells died after the treatment with low amount (3.0 μmol L-1) of the formulation compared to the free drug. In addition, the innovative formulation presented a combinatory mechanism of cell death involving autophagy and apoptosis. Our results demonstrate that the imiquimod-loaded nanoemulsioncan be an alternative product for the treatment of cervical cancer validating the hypothesis.
Collapse
|
26
|
Yoon TW, Kim YI, Cho H, Brand DD, Rosloniec EF, Myers LK, Postlethwaite AE, Hasty KA, Stuart JM, Yi AK. Ameliorating effects of Gö6976, a pharmacological agent that inhibits protein kinase D, on collagen-induced arthritis. PLoS One 2019; 14:e0226145. [PMID: 31809526 PMCID: PMC6897462 DOI: 10.1371/journal.pone.0226145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/20/2019] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptor (TLR) signaling can contribute to the pathogenesis of arthritis. Disruption of TLR signaling at early stages of arthritis might thereby provide an opportunity to halt the disease progression and ameliorate outcomes. We previously found that Gö6976 inhibits TLR-mediated cytokine production in human and mouse macrophages by inhibiting TLR-dependent activation of protein kinase D1 (PKD1), and that PKD1 is essential for proinflammatory responses mediated by MyD88-dependent TLRs. In this study, we investigated whether PKD1 contributes to TLR-mediated proinflammatory responses in human synovial cells, and whether Gö6976 treatment can suppress the development and progression of type II collagen (CII)-induced arthritis (CIA) in mouse. We found that TLR/IL-1R ligands induced activation of PKD1 in human fibroblast-like synoviocytes (HFLS). TLR/IL-1R-induced expression of cytokines/chemokines was substantially inhibited in Gö6976-treated HFLS and PKD1-knockdown HFLS. In addition, serum levels of anti-CII IgG antibodies, and the incidence and severity of arthritis after CII immunization were significantly reduced in mice treated daily with Gö6976. Synergistic effects of T-cell receptor and TLR, as well as TLR alone, on spleen cell proliferation and cytokine production were significantly inhibited in the presence of Gö6976. Our results suggest a possibility that ameliorating effects of Gö6976 on CIA may be due to its ability to inhibit TLR/IL-1R-activated PKD1, which might play an important role in proinflammatory responses in arthritis, and that PKD1 could be a therapeutic target for inflammatory arthritis.
Collapse
Affiliation(s)
- Tae Won Yoon
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Young-In Kim
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Hongsik Cho
- Department of Orthopedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - David D. Brand
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Edward F. Rosloniec
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Linda K. Myers
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Arnold E. Postlethwaite
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Karen A. Hasty
- Department of Orthopedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - John M. Stuart
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Ae-Kyung Yi
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
27
|
Imiquimod-loaded nanocapsules improve cytotoxicity in cervical cancer cell line. Eur J Pharm Biopharm 2019; 136:9-17. [DOI: 10.1016/j.ejpb.2019.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/10/2018] [Accepted: 01/03/2019] [Indexed: 12/26/2022]
|
28
|
Alculumbre S, Raieli S, Hoffmann C, Chelbi R, Danlos FX, Soumelis V. Plasmacytoid pre-dendritic cells (pDC): from molecular pathways to function and disease association. Semin Cell Dev Biol 2019; 86:24-35. [DOI: 10.1016/j.semcdb.2018.02.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 12/28/2017] [Accepted: 02/10/2018] [Indexed: 12/14/2022]
|
29
|
Abstract
Radiation enteritis is an old but emerging question induced by the application of radiation. However, no effective drugs for radiation enteritis in clinic. In this study, we found that thymoquinone (TQ) could mitigate intestinal damages induced by irradiation. After exposure to irradiation, TQ-treated improved the irradiated mice survival rate, ameliorated intestinal injury and increased the numbers of intestinal crypts. Furthermore, Lgr5+ ISCs and their daughter cells, including Vil1+ enterocytes, Ki67+ cells and lysozyme+ Paneth cells, were all significantly increased with TQ treatment. In addition, P53, γH2AX, caspase8, caspase9 and caspase3 expression were all reduced by TQ. Our data showed that TQ modulated DNA damages and decreased the apoptosis in the small intestine. TQ might be used for radiation enteritis treatment.
Collapse
|
30
|
Cen X, Liu S, Cheng K. The Role of Toll-Like Receptor in Inflammation and Tumor Immunity. Front Pharmacol 2018; 9:878. [PMID: 30127747 PMCID: PMC6088210 DOI: 10.3389/fphar.2018.00878] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/19/2018] [Indexed: 12/15/2022] Open
Abstract
Toll-like receptors (TLRs) activation enables host to recognize a large number of pathogen-associated molecule patterns (PAMPs), ignite immune cells to discriminate between self and non-self, and then promote the following innate and adaptive immune responses. Accumulated clinical/preclinical evidences have proven TLRs to be critical role in the autoimmune diseases, including inflammatory and tumor-associated diseases. Activation of TLRs is becoming or has been a target for cancer treatment. It is shown that TLRs can induce preferable anti-tumor effect by eliciting inflammatory cytokines expression and cytotoxic T lymphocytes (CTLs) response. As adjuvant, TLRs agonists can launch a strong immune response to assist cancer radiotherapy and bio-chemotherapy. On the other hand, tumor-associated antigens acting as PAMPs, can also activate TLRs and induce tumor gene-related programmed cell death, including apoptosis, autophagy and programmed necrosis. While there are also arguments that the excessive TLRs expression will promote tumor deterioration in various organisms, as the TLR-induced inflammation will accelerate the cancer cells boost in the tumor microenvironment (TME). However, the effect of TLRs acting on cancers is still not quite clear today. In this review, we will summarize the recent researches of TLRs in cancer treatment and their role in TME, giving a brief overview on future expectation.
Collapse
Affiliation(s)
- Xiaohong Cen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Kui Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
31
|
Ohara K, Ohkuri T, Kumai T, Nagato T, Nozaki Y, Ishibashi K, Kosaka A, Nagata M, Harabuchi S, Ohara M, Oikawa K, Aoki N, Harabuchi Y, Celis E, Kobayashi H. Targeting phosphorylated p53 to elicit tumor-reactive T helper responses against head and neck squamous cell carcinoma. Oncoimmunology 2018; 7:e1466771. [PMID: 30510853 DOI: 10.1080/2162402x.2018.1466771] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 12/19/2022] Open
Abstract
The human T cell receptor is capable of distinguishing between normal and post-translationally modified peptides. Because aberrant phosphorylation of cellular proteins is a hallmark of malignant transformation, the expression of the phosphorylated epitope could be an ideal antigen to combat cancer without damaging normal tissues. p53 activates transcription factors to suppress tumors by upregulating growth arrest and apoptosis-related genes. In response to DNA damage, p53 is phosphorylated at multiple sites including Ser33 and Ser37. Here, we identified phosphorylated peptide epitopes from p53 that could elicit effective T helper responses. These epitope peptides, p5322-41/Phospho-S33 and p5322-41/Phospho-S37, induced T helper responses against tumor cells expressing the phosphorylated p53 protein. Moreover, chemotherapeutic agents augmented the responses of such CD4 T cells via upregulation of phosphorylated p53. The upregulation of phosphorylated p53 expression by chemotherapy was confirmed in in vitro and xenograft models. We evaluated phosphorylated p53 expression in the clinical samples of oropharyngeal squamous cell carcinoma and revealed that 13/24 cases (54%) were positive for phosphorylated p53. Importantly, the lymphocytes specific for the phosphorylated p53 peptide epitopes were observed in the head and neck squamous cell cancer (HNSCC) patients. These results reveal that a combination of phosphorylated p53 peptides and chemotherapy could be a novel immunologic approach to treat HNSCC patients.
Collapse
Affiliation(s)
- Kenzo Ohara
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Takayuki Ohkuri
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Takumi Kumai
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan.,Department of Innovative Head & Neck Cancer Research and Treatment (IHNCRT), Asahikawa Medical University, Asahikawa, Japan
| | - Toshihiro Nagato
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Yui Nozaki
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Kei Ishibashi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Akemi Kosaka
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Marino Nagata
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Shohei Harabuchi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Mizuho Ohara
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan.,Department of surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Kensuke Oikawa
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Naoko Aoki
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Yasuaki Harabuchi
- Department of Otolaryngology-Head and Neck surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Esteban Celis
- Cancer Immunology, Inflammation and Tolerance Program, Augusta University, Georgia Cancer Center, Augusta, GA
| | - Hiroya Kobayashi
- Department of Pathology, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
32
|
Gastroprotective and Antioxidant Activity of Kalanchoe brasiliensis and Kalanchoe pinnata Leaf Juices against Indomethacin and Ethanol-Induced Gastric Lesions in Rats. Int J Mol Sci 2018; 19:ijms19051265. [PMID: 29695040 PMCID: PMC5983682 DOI: 10.3390/ijms19051265] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/26/2018] [Accepted: 03/29/2018] [Indexed: 12/31/2022] Open
Abstract
Kalanchoe brasiliensis and Kalanchoe pinnata are used interchangeably in traditional medicine for treating peptic ulcers and inflammatory problems. In this context, this study aims to characterize the chemical constituents and evaluate the gastroprotective activity of the leaf juices of the two species in acute gastric lesions models. Thin Layer Chromatography (TLC) and Ultra High Performance Liquid Chromatography coupled to Mass Spectrometer (UHPLC-MS) were performed for chemical characterization. Wistar rats were pre-treated orally with leaf juices (125, 250 and 500 mg/kg) or ranitidine (50 mg/kg). The peaks observed in the chromatogram of K. brasiliensis showed similar mass spectra to flavonoid glycosides derived from patuletin and eupafolin, while K. pinnata showed mass spectra similar to compounds derived from quercetin, patuletin, eupafolin and kaempferol. K. brasiliensis at all doses and K. pinnata at doses of 250 mg/kg and 500 mg/kg significantly reduced the lesions in the ethanol induction model. In the indomethacin induction model, both species showed significant results at doses of 250 and 500 mg/kg. Also, the pre-treatment with leaf juices increased the antioxidant defense system, glutathione (GSH), whereas malondialdehyde (MDA), myeloperoxidase (MPO), interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) levels were significantly decreased. Treatment with leaf juices led to the upregulation of zone occludes-1 (ZO-1) and the downregulation of inducible nitric oxide synthase (iNOS) and factor nuclear-κβ transcription (NF-κB-p65), while also showing a cytoprotective effect and maintaining mucus production. These findings show that the leaf juices of the two species showed gastroprotective effects on ethanol and gastric indomethacin injury which were a consequence of gastric inflammation suppression, antioxidant activity and the maintenance of cytoprotective defenses and mucosal structure architecture.
Collapse
|
33
|
Yuan J, Ni G, Wang T, Mounsey K, Cavezza S, Pan X, Liu X. Genital warts treatment: Beyond imiquimod. Hum Vaccin Immunother 2018; 14:1815-1819. [PMID: 29505317 DOI: 10.1080/21645515.2018.1445947] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Genital warts are one of the most common sexually transmitted diseases worldwide. The disease is a result of infection with low-risk types of human papillomaviruses, mostly type 6 and 11. Current therapies for genital warts are mainly ablative, or alternatively topical application of imiquimod cream and sinecatechin (polyphenon E) ointment to the warts. However, low patient compliance and high recurrence rate are significant problems for the treatment of genital warts by imiquimod and ablative therapies. We summarise recent literature in this area and propose combining imiquimod with other therapies to increase the efficacy of imiquimod.
Collapse
Affiliation(s)
- Jianwei Yuan
- a Institute of Molecular Diagnosis and Target Therapy, First Affiliated Hospital , Guangdong Pharmaceutical University , Guangzhou , Guangdong , China
| | - Guoying Ni
- c Inflammation and Healing research cluster, Faculty of Science, Health , Education and Engineering, University of the Sunshine Coast , Maroochydore DC , Australia
| | - Tianfang Wang
- b Genecology Research Centre, Faculty of Science, Health, Education and Engineering , University of the Sunshine Coast , Maroochydore DC , Australia
| | - Kate Mounsey
- c Inflammation and Healing research cluster, Faculty of Science, Health , Education and Engineering, University of the Sunshine Coast , Maroochydore DC , Australia
| | - Shelley Cavezza
- c Inflammation and Healing research cluster, Faculty of Science, Health , Education and Engineering, University of the Sunshine Coast , Maroochydore DC , Australia
| | - Xuan Pan
- a Institute of Molecular Diagnosis and Target Therapy, First Affiliated Hospital , Guangdong Pharmaceutical University , Guangzhou , Guangdong , China
| | - Xiaosong Liu
- a Institute of Molecular Diagnosis and Target Therapy, First Affiliated Hospital , Guangdong Pharmaceutical University , Guangzhou , Guangdong , China.,c Inflammation and Healing research cluster, Faculty of Science, Health , Education and Engineering, University of the Sunshine Coast , Maroochydore DC , Australia
| |
Collapse
|
34
|
Patchett AL, Wilson R, Charlesworth JC, Corcoran LM, Papenfuss AT, Lyons BA, Woods GM, Tovar C. Transcriptome and proteome profiling reveals stress-induced expression signatures of imiquimod-treated Tasmanian devil facial tumor disease (DFTD) cells. Oncotarget 2018; 9:15895-15914. [PMID: 29662615 PMCID: PMC5882306 DOI: 10.18632/oncotarget.24634] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/26/2018] [Indexed: 02/06/2023] Open
Abstract
As a topical cancer immunotherapy, the toll-like receptor 7 ligand imiquimod activates tumor regression via stimulation of immune cell infiltration and cytotoxic responses. Imiquimod also exerts direct pro-apoptotic effects on tumor cells in vitro, but a role for these effects in imiquimod-induced tumor regression remains undefined. We previously demonstrated that cell lines derived from devil facial tumor disease (DFTD), a transmissible cancer threatening the survival of the Tasmanian devil (Sarcophilus harrisii), are sensitive to imiquimod-induced apoptosis. In this study, the pro-apoptotic effects of imiquimod in DFTD have been investigated using RNA-sequencing and label-free quantitative proteomics. This analysis revealed that changes to gene and protein expression in imiquimod treated DFTD cells are consistent with the onset of oxidative and endoplasmic reticulum stress responses, and subsequent activation of the unfolded protein response, autophagy, cell cycle arrest and apoptosis. Imiquimod also regulates the expression of oncogenic pathways, providing a direct mechanism by which this drug may increase tumor susceptibility to immune cytotoxicity in vivo. Our study has provided the first global analysis of imiquimod-induced effects in any tumor cell line. These findings have highlighted the potential of cell stress pathways as therapeutic targets in DFTD, and will allow for improved mechanistic use of imiquimod as a therapy in both the Tasmanian devil and human cancers.
Collapse
Affiliation(s)
- Amanda L Patchett
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Richard Wilson
- Central Science Laboratory, University of Tasmania, Hobart, Tasmania 7001, Australia
| | - Jac C Charlesworth
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Lynn M Corcoran
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anthony T Papenfuss
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia.,Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria 3000, Australia
| | - Bruce A Lyons
- School of Medicine, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Gregory M Woods
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia.,School of Medicine, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Cesar Tovar
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| |
Collapse
|
35
|
Villamón E, González-Fernández J, Such E, Cervera JV, Gozalbo D, Luisa Gil M. Imiquimod inhibits growth and induces differentiation of myeloid leukemia cell lines. Cancer Cell Int 2018; 18:15. [PMID: 29422777 PMCID: PMC5791367 DOI: 10.1186/s12935-018-0515-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/24/2018] [Indexed: 12/13/2022] Open
Abstract
Background The antitumoral effects of different Toll-like receptor (TLRs) agonists is mediated by activating immune responses to suppress tumors growth, although TLR ligands may also have a direct effect on tumoral cells. Given that TLR signaling induces hematopoietic cell differentiations this may serve as a novel differentiation therapeutic approach for AML. Methods We investigated the effects of agonists for the ten human TLRs on the proliferation, apoptosis, cell cycle and differentiation of ten different types of myeloid leukemia cell lines (HL-60, U-937, KG-1, KG-1a, K-562, Kasumi-1, EOL-1, NB4, MOLM-13 and HEL). Proliferation was measured using the CellTiter 96® Aqueous One Solution Cell Proliferation Assay (Promega). Staining and analysis with a flow cytometer was used to identify cell cycle progression and apoptosis. Differentiation was measured by staining cells with the EuroFlow™ antibody panel for AML and analyzed by flow cytometry. FlowJo software was used to analyze the cytometric data. In all experiments, statistical significance was determined by a two-tailed t test. Results The activation of particular TLRs on some cell lines can induce growth inhibition and Imiquimod (a TLR 7 agonist) was the most effective agonist in all leukemic cell lines examined. Imiquimod was able to induce apoptosis, as well as to induce cell cycle alteration and upregulation of myeloid differentiation markers on some of the cell lines tested. Conclusions Our results, together with the known efficacy of Imiquimod against many tumor entities, suggest that Imiquimod can be a potential alternative therapy to AML. This drug has a direct cytotoxic effect on leukemic cells, has the potential to induce differentiation, and can also stimulate the activation of cellular immune responses anti-AML.
Collapse
Affiliation(s)
- Eva Villamón
- 1Department of Hematology, University Hospital La Fe, Valencia, Spain
| | | | - Esperanza Such
- 1Department of Hematology, University Hospital La Fe, Valencia, Spain
| | | | - Daniel Gozalbo
- 2Departamento de Microbiología y Ecología, Universitat de València, Burjasot, Spain.,3Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Burjasot, Spain
| | - M Luisa Gil
- 2Departamento de Microbiología y Ecología, Universitat de València, Burjasot, Spain.,3Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universitat de València, Burjasot, Spain
| |
Collapse
|
36
|
Abstract
The use of complementary and alternative medicines in Australia has grown significantly. Much of this growth is due to their ease of accessibility from online vendors, often marketed with claims that are not scientifically tested. Black salve is a topical escharotic compound containing the active component sanguinarine, derived from the bloodroot plant. It has been advertised as a natural treatment for skin cancer. This article reviews the current state of black salve as an alternative skin cancer treatment, discussing its distribution and regulation, and provides a summary of clinical and laboratory studies. Clinical trials in this area are lacking, with most clinical data in the form of case reports demonstrating suboptimal therapeutic and cosmetic outcomes associated with its use. However, in vitro studies of sanguinarine suggest it causes indiscriminate destruction of healthy and cancerous tissue at doses higher than 5 µM, limiting its practical utility. It is vital that members of the public are aware of the potential effects and toxicity of commercial salve products.
Collapse
Affiliation(s)
- Alvin Lim
- a Faculty of Medicine, Dentistry and Health Sciences , University of Melbourne , Parkville , Australia
| |
Collapse
|
37
|
Bakshi A, Chaudhary SC, Rana M, Elmets CA, Athar M. Basal cell carcinoma pathogenesis and therapy involving hedgehog signaling and beyond. Mol Carcinog 2017; 56:2543-2557. [PMID: 28574612 DOI: 10.1002/mc.22690] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/23/2017] [Accepted: 06/01/2017] [Indexed: 02/06/2023]
Abstract
Basal cell carcinoma (BCC) of the skin is driven by aberrant hedgehog signaling. Thus blocking this signaling pathway by small molecules such as vismodegib inhibits tumor growth. Primary cilium in the epidermal cells plays an integral role in the processing of hedgehog signaling-related proteins. Recent genomic studies point to the involvement of additional genetic mutations that might be associated with the development of BCCs, suggesting significance of other signaling pathways, such as WNT, NOTCH, mTOR, and Hippo, aside from hedgehog in the pathogenesis of this human neoplasm. Some of these pathways could be regulated by noncoding microRNA. Altered microRNA expression profile is recognized with the progression of these lesions. Stopping treatment with Smoothened (SMO) inhibitors often leads to tumor reoccurrence in the patients with basal cell nevus syndrome, who develop 10-100 of BCCs. In addition, the initial effectiveness of these SMO inhibitors is impaired due to the onset of mutations in the drug-binding domain of SMO. These data point to a need to develop strategies to overcome tumor recurrence and resistance and to enhance efficacy by developing novel single agent-based or multiple agents-based combinatorial approaches. Immunotherapy and photodynamic therapy could be additional successful approaches particularly if developed in combination with chemotherapy for inoperable and metastatic BCCs.
Collapse
Affiliation(s)
- Anshika Bakshi
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama.,Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Sandeep C Chaudhary
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mehtab Rana
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Craig A Elmets
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mohammad Athar
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
38
|
Chi H, Li C, Zhao FS, Zhang L, Ng TB, Jin G, Sha O. Anti-tumor Activity of Toll-Like Receptor 7 Agonists. Front Pharmacol 2017; 8:304. [PMID: 28620298 PMCID: PMC5450331 DOI: 10.3389/fphar.2017.00304] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 05/10/2017] [Indexed: 01/04/2023] Open
Abstract
Toll-like receptors (TLRs) are a class of pattern recognition receptors that play a bridging role in innate immunity and adaptive immunity. The activated TLRs not only induce inflammatory responses, but also elicit the development of antigen specific immunity. TLR7, a member of TLR family, is an intracellular receptor expressed on the membrane of endosomes. TLR7 can be triggered not only by ssRNA during viral infections, but also by immune modifiers that share a similar structure to nucleosides. Its powerful immune stimulatory action can be potentially used in the anti-tumor therapy. This article reviewed the anti-tumor activity and mechanism of TLR7 agonists that are frequently applied in preclinical and clinical investigations, and mainly focused on small synthetic molecules, including imiquimod, resiquimod, gardiquimod, and 852A, etc.
Collapse
Affiliation(s)
- Huju Chi
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science CentreShenzhen, China
| | - Chunman Li
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science CentreShenzhen, China
| | - Flora Sha Zhao
- School of Life Sciences, Faculty of Science, The Chinese University of Hong KongHong Kong, Hong Kong
| | - Li Zhang
- Department of Physiology and Neurology, University of ConnecticutStorrs, CT, United States
| | - Tzi Bun Ng
- Departmet of Biochemistry, Faculty of Science, The Chinese University of Hong KongHong Kong, Hong Kong
| | - Guangyi Jin
- Department of Pharmacy, Shenzhen University Health Science CentreShenzhen, China
| | - Ou Sha
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science CentreShenzhen, China
| |
Collapse
|
39
|
Guo FZ, Xu Y, Ren LH, Zhang J, Zhang F, Duan J, Zhou XQ, Sun ZW. Endosulfan induces apoptosis by activating the negative regulation pathway of cell cycle and death receptor pathway in spermatogenic cells. Toxicol Res (Camb) 2017; 6:223-231. [PMID: 30090493 DOI: 10.1039/c6tx00315j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/05/2017] [Indexed: 01/17/2023] Open
Abstract
The male reproductive toxicity of endosulfan has been proved. Nevertheless, the underlying molecular mechanisms of the apoptosis caused by endosulfan in spermatogenic cells remains poorly understood. In order to investigate the reproductive toxicity mechanism caused by endosulfan, there were four groups, which had eight Wistar male rats randomly assigned to them, and the rats in different groups received different doses of endosulfan for a period of 21 days. GC-1 spermatogenic cell lines were divided into four groups, and each group was exposed to different doses of endosulfan for 24 hours. The results of this research showed that endosulfan decreased the cell viability, damaged cell membranes and induced apoptosis in spermatogenic cells. Endosulfan had obviously activated the protein expression of PKC-δ, p53, p21cip1, p27kip1, Fas, FasL, Caspase-8, Caspase-3, and inhibited the expression of E2F-1. Endosulfan also induced oxidative stress and DNA damage in spermatogenic cells. The results of this research suggested that endosulfan could lead to E2F-1-induced apoptosis of spermatogenic cells by activating the negative regulation factors of the cell cycle, and endosulfan might cause apoptosis by death receptor pathway, causing oxidative stress.
Collapse
Affiliation(s)
- Fang-Zi Guo
- Department of Toxicology and Hygienic Chemistry , School of Public Health , Capital Medical University , Beijing , 100069 , China . ; ; Tel: +8610-83911775.,Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Ying Xu
- Department of Toxicology and Hygienic Chemistry , School of Public Health , Capital Medical University , Beijing , 100069 , China . ; ; Tel: +8610-83911775.,Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Li-Hua Ren
- Department of Toxicology and Hygienic Chemistry , School of Public Health , Capital Medical University , Beijing , 100069 , China . ; ; Tel: +8610-83911775.,Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Jin Zhang
- Department of Toxicology and Hygienic Chemistry , School of Public Health , Capital Medical University , Beijing , 100069 , China . ; ; Tel: +8610-83911775.,Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Feng Zhang
- College of Life Science , Qilu Normal University , Jinan 250013 , China
| | - Junchao Duan
- Department of Toxicology and Hygienic Chemistry , School of Public Health , Capital Medical University , Beijing , 100069 , China . ; ; Tel: +8610-83911775.,Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Xian-Qing Zhou
- Department of Toxicology and Hygienic Chemistry , School of Public Health , Capital Medical University , Beijing , 100069 , China . ; ; Tel: +8610-83911775.,Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| | - Zhi-Wei Sun
- Department of Toxicology and Hygienic Chemistry , School of Public Health , Capital Medical University , Beijing , 100069 , China . ; ; Tel: +8610-83911775.,Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing 100069 , China
| |
Collapse
|
40
|
The Immunomodulatory Small Molecule Imiquimod Induces Apoptosis in Devil Facial Tumour Cell Lines. PLoS One 2016; 11:e0168068. [PMID: 27936237 PMCID: PMC5148113 DOI: 10.1371/journal.pone.0168068] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/28/2016] [Indexed: 12/13/2022] Open
Abstract
The survival of the Tasmanian devil (Sarcophilus harrisii) is threatened by devil facial tumour disease (DFTD). This transmissible cancer is usually fatal, and no successful treatments have been developed. In human studies, the small immunomodulatory molecule imiquimod is a successful immunotherapy, activating anti-tumour immunity via stimulation of toll-like receptor-7 (TLR7) signaling pathways. In addition, imiquimod is a potent inducer of apoptosis in human tumour cell lines via TLR7 independent mechanisms. Here we investigate the potential of imiquimod as a DFTD therapy through analysis of treated DFTD cell lines and Tasmanian devil fibroblasts. WST-8 proliferation assays and annexin V apoptosis assays were performed to monitor apoptosis, and changes to the expression of pro- and anti-apoptotic genes were analysed using qRT-PCR. Our results show that DFTD cell lines, but not Tasmanian devil fibroblasts, are sensitive to imiquimod-induced apoptosis in a time and concentration dependent manner. Induction of apoptosis was accompanied by down-regulation of the anti-apoptotic BCL2 and BCLXL genes, and up-regulation of the pro-apoptotic BIM gene. Continuous imiquimod treatment was required for these effects to occur. These results demonstrate that imiquimod can deregulate DFTD cell growth and survival in direct and targeted manner. In vivo, this may increase DFTD vulnerability to imiquimod-induced TLR7-mediated immune responses. Our findings have improved the current knowledge of imiquimod action in tumour cells for application to both DFTD and human cancer therapy.
Collapse
|
41
|
Saeed AL-Wajeeh N, Halabi MF, Hajrezaie M, M. Dhiyaaldeen S, Abdulaziz Bardi D, M. Salama S, Rouhollahi E, Karimian H, Abdolmalaki R, Azizan AHS, Mohd Ali H, Mohd Noor S, Abdulla MA. The Gastroprotective Effect of Vitex pubescens Leaf Extract against Ethanol-Provoked Gastric Mucosal Damage in Sprague-Dawley Rats. PLoS One 2016; 11:e0157431. [PMID: 27689880 PMCID: PMC5045201 DOI: 10.1371/journal.pone.0157431] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 05/30/2016] [Indexed: 12/26/2022] Open
Abstract
Vitex pubescens is a Malaysian therapeutic plant employed in traditional drug to remedy a variety of disorders. The purpose of this research is to assess the gastroprotective efficiency of V. pubescens leaves against ethanol-induced gastric hemorrhagic laceration in rats. Animals were randomly allocated into seven groups and pre-treated, separately, with 10% Tween 20 (normal and ulcer control groups), 20 mg/kg omeprazole (reference group), and 62.5, 125, 250, and 500 mg/kg of V. pubescens extract (experimental groups). All animals were sacrificed after another hour. Histological evaluation of the ulcer control group revealed significant injury to the gastric mucosa with edema and leucocyte infiltration of the submucosal layer. PAS staining, showed remarkably intense magenta color, remarkable increase of HSP70 and decrease of Bax proteins in rats pre-treated with plant extracts compared to the ulcer control group. Gastric homogenates revealed a remarkable increase in endogenous antioxidant enzyme activities (CAT, SOD, GSH) and a decrease in the lipid peroxidation level (MDA) in animals pre-treated with V. pubescens extract compared with the ulcer control group. The gastroprotective activity of this plant might be related to increased antioxidant enzymes and decrease lipid peroxidation upsurge of HSP70 and reduced expression of Bax proteins.
Collapse
Affiliation(s)
- Nahla Saeed AL-Wajeeh
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mohammed Farouq Halabi
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Maryam Hajrezaie
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Summaya M. Dhiyaaldeen
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Daleya Abdulaziz Bardi
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Suzy M. Salama
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Elham Rouhollahi
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Hamed Karimian
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Rojin Abdolmalaki
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | | | - Hapipah Mohd Ali
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Suzita Mohd Noor
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mahmood Ameen Abdulla
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| |
Collapse
|