1
|
Kim G, Kim EY, Lee H, Shin SH, Lee SH, Sohn KY, Kim JW, Lee JS. 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol treatment inhibits abnormal tumor growth by regulating neutrophil infiltration in a non-small cell lung carcinoma mouse model. Biomed Pharmacother 2024; 178:117269. [PMID: 39137654 DOI: 10.1016/j.biopha.2024.117269] [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: 05/07/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Excessive neutrophil infiltration into the tumor microenvironment (TME) is an important factor that contributes to tumor overgrowth and limited immunotherapy efficacy. Neutrophils activate various receptors involved in tumor progression, while suppressing the infiltration and activity of cytotoxic T cells and creating optimal conditions for tumor growth. Therefore, the appropriate control of neutrophil infiltration is an effective strategy for tumor treatment. In the present study, 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) inhibited tumor overgrowth by suppressing excessive neutrophil infiltration, resulting in >74.97 % reduction in tumor size in a Lewis lung carcinoma (LLC-1) mouse model. All subjects in the positive control group died during the 90-day survival period, whereas only four subjects in the PLAG treatment group survived. PLAG had a significantly higher tumor growth inhibitory effect and survival rate than other neutrophil infiltration-targeting inhibitors (e.g., Navarixin, lymphocyte antigen 6 complex locus G6D antibody [aLy6G]). The ability of PLAG to regulate neutrophil infiltration and inhibit tumor growth depends on thioredoxin-interacting protein (TXNIP). In tumors lacking TXNIP expression, PLAG failed to control neutrophil infiltration and infiltration-related factor release, and the inhibitory effect of PLAG on tumor growth was reduced. PLAG-mediated inhibition of neutrophil infiltration enhances the efficacy of immune checkpoint inhibitors (ICIs), increasing the antitumor efficacy and survival rate by 30 %. In conclusion, PLAG could be a novel alternative to anti-tumor drugs that effectively targets excessive neutrophil infiltration into cancer tissues.
Collapse
Affiliation(s)
- Guentae Kim
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea; Biotoxtech, 53 Yeongudanji-ro, Ochang-eup, Cheongju-si, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea
| | - Jae Sam Lee
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea.
| |
Collapse
|
2
|
Luisa Valerio de Mello Braga L, Simão G, Silva Schiebel C, Caroline Dos Santos Maia A, Mulinari Turin de Oliveira N, Barbosa da Luz B, Rita Corso C, Soares Fernandes E, Maria Ferreira D. Rodent models for anticancer toxicity studies: contributions to drug development and future perspectives. Drug Discov Today 2023:103626. [PMID: 37224998 DOI: 10.1016/j.drudis.2023.103626] [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: 08/12/2022] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023]
Abstract
Antineoplastic treatment induces a type of gastrointestinal toxicity known as mucositis. Findings in animal models are usually easily reproducible, and standardized treatment regimens are often used, thus supporting translational science. Essential characteristics of mucositis, including intestinal permeability, inflammation, immune and oxidative responses, and tissue repair mechanisms, can be easily investigated in these models. Given the effects of mucositis on the quality of life of patients with cancer, and the importance of experimental models in the development of more effective new therapeutic alternatives, this review discusses progress and current challenges in using experimental models of mucositis in translational pharmacology research. Teaser Experimental models for studying gastrointestinal mucositis have provided a wealth of information improving the understanding of antineoplastic toxicity.
Collapse
Affiliation(s)
- Lara Luisa Valerio de Mello Braga
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Gisele Simão
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Carolina Silva Schiebel
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Andressa Caroline Dos Santos Maia
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Bruna Barbosa da Luz
- Departamento de Farmacologia, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Claudia Rita Corso
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Elizabeth Soares Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| | - Daniele Maria Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, PR, Brazil.
| |
Collapse
|
3
|
Villa A, Sonis ST. Radiotherapy-induced severe oral mucositis: pharmacotherapies in recent and current clinical trials. Expert Opin Investig Drugs 2023; 32:301-310. [PMID: 36932830 DOI: 10.1080/13543784.2023.2193324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
INTRODUCTION Oral mucositis (OM) is among the most common, damaging side effects of head and neck radiation therapy and may interfere with patients' ability to comply with optimal treatment. AREAS COVERED The increasing unmet clinical need, recent clinical trial successes, and the commercial potential have catalyzed interest in the development of effective intervention for OM. A range of small molecules are under development - some still in the preclinical stage, but others close to NDA submission. This review will focus on those drugs which have recently been assessed in a clinical trial and those which are still under clinical study as a prevention or treatment for radiation-associated OM. EXPERT OPINION In response to the unmet clinical need, both the biotechnology and pharmacological industries have been actively pursuing an agent to prevent/treat radiation-associated OM. This effort has been catalyzed by the identification of multiple drug targets which contribute to OM's pathogenesis. The lessons learned from the many trials which have previously stumbled have led to standardization of clinical trial design, endpoint efficacy definitions, rater assessment, and data interpretation over the past decade. Consequently, results of recently completed clinical trials provide optimism that effective treatment options should be available in the not-too-distant future.
Collapse
Affiliation(s)
- Alessandro Villa
- Oral Medicine, Oral Oncology and Dentistry. Miami Cancer Institute, Baptist Health South Florida, Miami, United States
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco
| | - Stephen T Sonis
- Divisions of Oral Medicine and Dentistry, Brigham and Women's Hospital and the Dana-Farber Cancer Institute, Boston
- Biomodels, LLC and Primary Endpoint Solutions, LLC, Waltham
| |
Collapse
|
4
|
Kim GT, Kim EY, Shin SH, Lee H, Lee SH, Sohn KY, Kim JW. Improving anticancer effect of aPD-L1 through lowering neutrophil infiltration by PLAG in tumor implanted with MB49 mouse urothelial carcinoma. BMC Cancer 2022; 22:727. [PMID: 35787261 PMCID: PMC9251917 DOI: 10.1186/s12885-022-09815-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The PD-L1 antibody is an immune checkpoint inhibitor (ICI) attracting attention. The third-generation anticancer drug has been proven to be very effective due to fewer side effects and higher tumor-specific reactions than conventional anticancer drugs. However, as tumors produce additional resistance in the host immune system, the effectiveness of ICI is gradually weakening. Therefore, it is very important to develop a combination therapy that increases the anticancer effect of ICI by removing anticancer resistance factors present around the tumor. METHODS The syngeneic model was used (n = 6) to investigate the enhanced anti-tumor effect of PD-L1 antibody with the addition of PLAG. MB49 murine urothelial cancer cells were implanted into the C57BL/6 mice subcutaneously. PLAG at different dosages (50/100 mpk) was daily administered orally for another 4 weeks with or without 5 mpk PD-L1 antibody (10F.9G2). PD-L1 antibody was delivered via IP injection once a week. RESULTS The aPD-L1 monotherapy group inhibited tumor growth of 56% compared to the positive group, while the PLAG and aPD-L1 co-treatment inhibited by 89%. PLAG treatment effectively reduced neutrophils infiltrating localized in tumor and converted to a tumor microenvironment with anti-tumor effective T-cells. PLAG increased tumor infiltration of CD8 positive cytotoxic T-cell populations while effectively inhibiting the infiltration of neoplastic T-cells such as CD4/FoxP3. Eventually, neutrophil-induced tumor ICI resistance was resolved by restoring the neutrophil-to-lymphocyte ratio to the normal range. In addition, regulation of cytokine and chemokine factors that inhibit neutrophil infiltration and increase the killing activity of cytotoxic T cells was observed in the tumors of mice treated with PLAG + aPD-L1. CONCLUSIONS PLAG effectively turned the tumor-promoting microenvironment into a tumor-suppressing microenvironment. As a molecule that increases the anti-tumor effectiveness of aPD-L1, PLAG has the potential to be an essential and effective ICI co-therapeutic agent.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- grid.249967.70000 0004 0636 3099Division of Systems Biology and Bioengineering, Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea
| |
Collapse
|
5
|
Kim GT, Shin SH, Kim EY, Lee H, Lee SH, Sohn KY, Kim JW. PLAG co-treatment increases the anticancer effect of Adriamycin and cyclophosphamide in a triple-negative breast cancer xenograft mouse model. Biochem Biophys Res Commun 2022; 619:110-116. [PMID: 35753218 DOI: 10.1016/j.bbrc.2022.06.051] [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: 05/24/2022] [Revised: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 11/02/2022]
Abstract
Chemotherapy induces tumor cell death and inhibits tumor progression, but the accompanying immune responses in the surrounding dying tissue cause significant inflammation. These responses, such as excessive neutrophil infiltration into tumor tissue, are the main causes of resistance to anticancer treatment. The development of drugs that reduce neutrophil infiltration into tumors is necessary to increase the anticancer effect of chemotherapy. Here, we show that the antitumor effect of the chemotherapy AC regimen (Adriamycin and cyclophosphamide) was increased by 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) cotreatment in the MDA-MB-231 triple-negative breast cancer xenograft mouse model. Tumor growth was inhibited up to 56% in mice treated with AC and inhibited up to 94% in mice cotreated with AC and PLAG. Side effects of chemotherapy, such as a reduction in body weight, were alleviated in mice cotreated with AC and PLAG. Excessive neutrophil infiltration caused by the AC regimen was successfully cleared in mice cotreated with AC and PLAG. We conclude that PLAG inhibits excessive neutrophil infiltration that aids tumor growth. Reduced neutrophils and increased lymphocytes in PLAG-treated mice can maximize the antitumor effect of the AC regimen and inhibit tumor growth.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea.
| |
Collapse
|
6
|
Kim GT, Kim EY, Shin SH, Lee H, Lee SH, Sohn KY, Kim JW. Suppression of tumor progression by thioredoxin-interacting protein-dependent adenosine 2B receptor degradation in a PLAG-treated Lewis lung carcinoma-1 model of non-small cell lung cancer. Neoplasia 2022; 31:100815. [PMID: 35728512 PMCID: PMC9209866 DOI: 10.1016/j.neo.2022.100815] [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: 05/17/2022] [Revised: 05/23/2022] [Accepted: 06/08/2022] [Indexed: 11/03/2022]
Abstract
PLAG effectively inhibited excessive growth of LLC1 cells in an NSCLC model. PLAG inhibited tumor growth by inducing adenosine 2B receptor (A2BR) degradation. Unlike antagonists, PLAG terminates rather than suppresses signaling pathways. A2BR degradation by PLAG occurs through expression and re-localization of TXNIP.
Extracellular adenosine in the tumor microenvironment plays a vital role in cancer development. Specifically, activation of adenosine receptors affects tumor cell growth and adenosine release. We examined the anti-tumor efficacy of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) in animal models, revealing the role of PLAG in inhibiting tumor progression by promoting the degradation of adenosine 2B receptors (A2BRs) in tumors. PLAG induced the expression of thioredoxin-interacting protein (TXNIP), a type of α-arrestin that accelerates A2BR internalization by interacting with A2BR complexes containing β-arrestin. Engulfed receptors bound to TXNIP were rapidly degraded after E3 ligase recruitment and ubiquitination, resulting in early termination of intracellular signals that promote tumor overgrowth. However, in control cancer cells, A2BRs bound to protein phosphatase 2A and were returned to the cell membrane instead of being degraded, resulting in continuous receptor-mediated signaling by pathways including the Raf-Erk axis, which promotes tumor proliferation. A TXNIP-silenced cell-implanted mouse model and TXNIP knockout (KO) mice were used to verify that PLAG-mediated suppression of tumor progression is dependent on TXNIP expression. Increased tumor growth was observed in TXNIP-silenced cell-implanted mice, and the anti-tumor effects of PLAG, including delayed tumor overgrowth, were greatly reduced. However, the anti-tumor effects of PLAG were observed in cancer cell-implanted TXNIP-KO mice, which indicates that PLAG produces anti-tumor effects by enhancing TXNIP expression in tumor cells. These essential functions of PLAG, including delaying tumor growth via A2BR degradation, suggest innovative directions for anticancer drug development.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea.
| |
Collapse
|
7
|
Kim GT, Kim EY, Shin SH, Lee H, Lee SH, Park K, Sohn KY, Yoon SY, Kim JW. PLAG alleviates cisplatin-induced cachexia in lung cancer implanted mice. Transl Oncol 2022; 20:101398. [PMID: 35339890 PMCID: PMC8957043 DOI: 10.1016/j.tranon.2022.101398] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/23/2022] Open
Abstract
PLAG effectively relieves cachexia symptoms caused by chemotherapy. PLAG restores leg muscles atrophy by cisplatin to normal levels. PLAG returns cachexia-related elements modulated by cisplatin to normal levels. PLAG did not downregulate the anti-tumor effect of cisplatin.
Chemotherapy-induced cachexia has been a significant challenge to the successful treatment of cancer patients. Chemotherapy leads to loss of muscle, loss of appetite, and excessive weight loss, which makes these necessary treatments intolerable for most patients. Therefore, it is necessary to alleviate cachexia to successfully treat cancer patients. In this study, tumor-implanted mouse models administered cisplatin showed rapid weight loss and reduced feeding rate by the second week of treatment, and 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) effectively alleviated cisplatin-induced cachexia. In mice treated with cisplatin on a sacrificial day after 6 weeks, the weight of the two major leg muscles (quadriceps femoris and gastrocnemius) were reduced by up to 70%, but this muscle reduction was successfully prevented in the PLAG co-treatment group. The distribution and size of muscle fibers that appear in small units in cisplatin-treated mice were restored to normal levels by PLAG co-treatment. Furthermore, myostatin expression levels were upregulated by cisplatin, whereas myostatin decreased to normal levels with muscle recovery in the PLAG co-treated group. Tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), which are commonly expressed in cachexia, were significantly increased in cisplatin-treated mice but were reduced to normal levels in PLAG co-treated mice. Glucose absorption, an indicator of muscle tissue activity, decreased with cisplatin treatment and recovered to normal levels with PLAG co-treatment. Overall, PLAG effectively alleviated cisplatin-induced cachexia symptoms and reduced tumor growth in tumor-implanted mice. These findings suggest PLAG may be a promising drug to alleviate cachexia in cancer patients receiving chemotherapy.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Kaapjoo Park
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Sun Young Yoon
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea.
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea.
| |
Collapse
|
8
|
Emerging approaches of wound healing in experimental models of high-grade oral mucositis induced by anticancer therapy. Oncotarget 2021; 12:2283-2299. [PMID: 34733419 PMCID: PMC8555685 DOI: 10.18632/oncotarget.28091] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/24/2021] [Indexed: 01/22/2023] Open
Abstract
Clinical guidelines for oral mucositis (OM) still consist in palliative care. Herein, we summarize cellular and molecular mechanisms of OM ulceration in response to chemical therapies in animal models. We discuss evidenced anti-inflammatory and anti-oxidant drugs which have not been ever used for OM, such as synthetic peptides as well as cell therapy with mesenchymal stem cells; amniotic membranes, mucoadhesive polymers loaded with anti-inflammatory agents and natural or synthetic electrospun. These approaches have been promising to allow the production of drug-loaded membranes, scaffolds for cells encapsulation or guided tissue regeneration.
Collapse
|
9
|
Saul-McBeth J, Dillon J, Lee A, Launder D, Kratch JM, Abutaha E, Williamson AA, Schroering AG, Michalski G, Biswas P, Conti SR, Shetty AC, McCracken C, Bruno VM, Parsai EI, Conti HR. Tissue Damage in Radiation-Induced Oral Mucositis Is Mitigated by IL-17 Receptor Signaling. Front Immunol 2021; 12:687627. [PMID: 34220843 PMCID: PMC8248500 DOI: 10.3389/fimmu.2021.687627] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/31/2021] [Indexed: 01/13/2023] Open
Abstract
Oral mucositis (OM) is a treatment-limiting adverse side effect of radiation and chemotherapy. Approximately 80% of patients undergoing radiotherapy (RT) for head and neck cancers (HNC) develop OM, representing a major unmet medical condition. Our understanding of the immunopathogenesis of OM is limited, due in part to the surprising paucity of information regarding healing mechanisms in the oral mucosa. RNAseq of oral tissue in a murine model that closely mimics human OM, showed elevated expression of IL-17 and related immune pathways in response to head and neck irradiation (HNI). Strikingly, mice lacking the IL-17 receptor (IL-17RA) exhibited markedly more severe OM. Restoration of the oral mucosa was compromised in Il17ra-/- mice and components associated with healing, including matrix metalloproteinase 3, 10 and IL-24 were diminished. IL-17 is typically associated with recruitment of neutrophils to mucosal sites following oral infections. Unexpectedly, in OM the absence of IL-17RA resulted in excessive neutrophil recruitment and immunopathology. Instead, neutrophil activation was IL-1R-driven in Il17ra-/- mice. Blockade of IL-1R and depletion of neutrophils lessened the severity of damage in these mice. Overall, we show IL-17 is protective in OM through multiple mechanisms including restoration of the damaged epithelia and control of the neutrophil response. We also present a clinically relevant murine model of human OM to improve mechanistic understanding and develop rational translational therapeutics.
Collapse
Affiliation(s)
- Jessica Saul-McBeth
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - John Dillon
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Aaron Lee
- Department of Radiation Oncology, Division of Medical Physics, The University of Toledo, Toledo, OH, United States
| | - Dylan Launder
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Jacqueline M. Kratch
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Eanas Abutaha
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | | | | | - Grace Michalski
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Priosmita Biswas
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Samuel R. Conti
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Amol C. Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Carrie McCracken
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Vincent M. Bruno
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - E. Ishmael Parsai
- Department of Radiation Oncology, Division of Medical Physics, The University of Toledo, Toledo, OH, United States
| | - Heather R. Conti
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| |
Collapse
|
10
|
Chantree P, Na-Bangchang K, Martviset P. Anticancer Activity of Fucoidan via Apoptosis and Cell Cycle Arrest on Cholangiocarcinoma Cell. Asian Pac J Cancer Prev 2021; 22:209-217. [PMID: 33507701 PMCID: PMC8184191 DOI: 10.31557/apjcp.2021.22.1.209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/15/2021] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Many previous studies reported that fucoidan has antitumor activities. The objective of the present study was to determine the cytotoxic effects and related mechanisms of cell death induced by fucoidan extracted from Fucus vesiculosus on CL-6 cholangiocarcinoma cell. METHODS CL-6 and OUMS cells were treated with 0, 100, 200, and 300 μg/mL of fucoidan. MTT assay was used to determine cytotoxicity. Flow cytometry-based assay was used to examine the distribution of apoptosis and cell cycle. The changes in nuclear morphology were determined using Hoechst 33,342 staining. Mitochondrial membrane potential (ΔΨm) was evaluated using the JC-1 kit. The apoptotic, anti-apoptotic, and cell cycle-related proteins study were examined by Western blot analysis. RESULTS The relative viable cell number of treated CL-6 cells was decreased but no effect was observed in OUMS normal cells. Furthermore, treated cells were arrested in the G0/G1 phase with down-regulation of cyclin D1 and CDK4. Annexin V/PI staining with flow cytometry analysis suggested that fucoidan could induce apoptosis in CL-6 cells. Western blot study revealed the up-regulation of apoptotic markers including Bax, cleaved PARP, cleaved caspase-3, but down-regulation of anti-apoptotic markers, cl-2. Moreover, fucoidan could induce nuclear fragmentation and chromatin condensation with alteration of ΔΨm. Conclusion: Fucoidan exerts antitumor properties against CL-6 cholangiocarcinoma cells illustrated by the induction of apoptosis and cell cycle arrest. .
Collapse
Affiliation(s)
- Pathanin Chantree
- Division of Anatomy, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
- Research Unit in Nutraceuticals and Food Safety, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
| | - Kesara Na-Bangchang
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
- Center of Excellence in Molecular Biology and Pharmacology of Malaria and Cholangiocarcinoma, Thammasat University, Pathumthani, 12120, Thailand.
| | - Pongsakorn Martviset
- Center of Excellence in Molecular Biology and Pharmacology of Malaria and Cholangiocarcinoma, Thammasat University, Pathumthani, 12120, Thailand.
- Division of Parasitology, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand.
| |
Collapse
|
11
|
Chen C, Zhang Q, Yu W, Chang B, Le AD. Oral Mucositis: An Update on Innate Immunity and New Interventional Targets. J Dent Res 2020; 99:1122-1130. [PMID: 32479139 DOI: 10.1177/0022034520925421] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oral mucositis (OM), a common debilitating toxicity associated with chemo- and radiation therapies, is a significant unmet clinical need for head and neck cancer patients. The biological complexities of chemoradiotherapy-induced OM involve interactions among disrupted tissue structures, inflammatory infiltrations, and oral microbiome, whereby several master inflammatory pathways constitute the complicated regulatory networks. Oral mucosal damages triggered by chemoradiotherapy-induced cell apoptosis were further exacerbated by the amplified inflammatory cascades dominantly governed by the innate immune responses. The coexistence of microbiome and innate immune components in oral mucosal barriers indicates that a signaling hub coordinates the interaction between environmental cues and host cells during tissue and immune homeostasis. Dysbiotic shifts in oral microbiota caused by cytotoxic cancer therapies may also contribute to the progression and severity of chemoradiotherapy-induced OM. In this review, we have updated the mechanisms involving innate immunity-governed inflammatory cascades in the pathobiology of chemoradiotherapy-induced OM and the development of new interventional targets for the management of this severe morbidity in head and neck cancer patients.
Collapse
Affiliation(s)
- C Chen
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center of Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Q Zhang
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - W Yu
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - B Chang
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - A D Le
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center of Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.,Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
12
|
Yang KH, Kim GT, Choi S, Yoon SY, Kim JW. 1‑Palmitoyl‑2‑linoleoyl‑3‑acetyl‑rac‑glycerol ameliorates EGF‑induced MMP‑9 expression by promoting receptor desensitization in MDA‑MB‑231 cells. Oncol Rep 2020; 44:241-251. [PMID: 32377695 PMCID: PMC7254954 DOI: 10.3892/or.2020.7599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
Activated epidermal growth factor receptors (EGFRs) are crucial for inducing metastasis in cancer cells by promoting matrix metalloproteinase (MMP) expression. The present study was designed to investigate the effects of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) on MMP expression in epidermal growth factor (EGF)-stimulated breast cancer cells in vitro. EGF stimulation induced internalization of its cognate receptor, EGFR, for stimulus-desensitization. These internalized receptors, complexed with the ubiquitin ligase c-Cbl and EGFR pathway substrate 15 (EPS15) (for degradation), were evaluated by confocal microscopy at 5–90 min time intervals. During intracellular trafficking of EGFRs, EGF-induced signaling cascades were analyzed by examining EGFR and SHC phosphorylation. Modulation of MMP expression was assessed by evaluating the activity of transcription factor AP-1 using a luciferase assay. PLAG accelerated the assembly of EGFRs with c-Cbl and EPS15 and promoted receptor degradation. This faster intracellular EGFR degradation reduced AP-1-mediated MMP expression. PLAG stimulation upregulated thioredoxin-interacting protein (TXNIP) expression, and this mediated the accelerated receptor internalization. This PLAG-induced increase in EGFR trafficking was blocked in TXNIP-silenced cells. By downregulating MMP expression, PLAG effectively attenuated EGF-induced mobility and invasiveness in these cancer cells. These data suggest that PLAG may be a potential therapeutic agent for blocking metastasis.
Collapse
Affiliation(s)
- Kwang Hoon Yang
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Guen Tae Kim
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Solji Choi
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Sun Young Yoon
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, Chungcheongbukdo 27159, Republic of Korea
| | - Jae Wha Kim
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
13
|
Shin SH, Jeong J, Kim JH, Sohn KY, Yoon SY, Kim JW. 1-Palmitoyl-2-Linoleoyl-3-Acetyl-rac-Glycerol (PLAG) Mitigates Monosodium Urate (MSU)-Induced Acute Gouty Inflammation in BALB/c Mice. Front Immunol 2020; 11:710. [PMID: 32395118 PMCID: PMC7196669 DOI: 10.3389/fimmu.2020.00710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/30/2020] [Indexed: 12/25/2022] Open
Abstract
Acute gouty arthritis is an auto-inflammatory disease caused by the deposition of monosodium urate (MSU) crystals in joints or tissues. Excessive neutrophil recruitment into gouty lesions is a general clinical sign and induces a pain phenotype. Attenuation of successive periods of neutrophil infiltration might be a beneficial approach to achieve therapeutic efficacy. In this study, the activity of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) in attenuation of excess neutrophil infiltration was assessed in gout-induced lesions of BALB/c mice. Neutrophil infiltration in MSU-induced gouty lesions was analyzed using immunohistochemical staining. ELISA and RT-PCR were used to measure attenuation of expression of the major neutrophil chemoattractant, CXC motif chemokine ligand 8 (CXCL8), in a PLAG-treated animal model and in cells in vitro. The animal model revealed massive increased neutrophil infiltration in the MSU-induced gouty lesions, but the PLAG-treated mice had significantly reduced neutrophil numbers in these lesions. The results also indicated that the MSU crystals stimulated a damage-associated molecular pattern that was recognized by the P2Y6 purinergic receptor. This MSU-stimulated P2Y6 receptor was destined to intracellular trafficking. During intracellular endosomal trafficking of the receptor, endosome-dependent signaling provided expression of CXCL8 chemokines for neutrophil recruitment. PLAG accelerated initiation of the intracellular trafficking of the P2Y6 receptor and returning the receptor to the membrane. This process shortened the intracellular retention time of the receptor anchoring endosome and subsequently attenuated endosome-dependent signaling for CXCL8 expression. These study results suggested that PLAG could be used for resolution of acute inflammation induced in gout lesions.
Collapse
Affiliation(s)
- Su-Hyun Shin
- Division of Systems Biology and Bioengineering, Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea
| | - Jinseon Jeong
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Seoul, South Korea
| | - Joo Heon Kim
- Department of Pathology, EulJi University School of Medicine, Daejeon, South Korea
| | - Ki-Young Sohn
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Seoul, South Korea
| | - Sun Young Yoon
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Seoul, South Korea
| | - Jae Wha Kim
- Division of Systems Biology and Bioengineering, Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
14
|
PLAG Exerts Anti-Metastatic Effects by Interfering with Neutrophil Elastase/PAR2/EGFR Signaling in A549 Lung Cancer Orthotopic Model. Cancers (Basel) 2020; 12:cancers12030560. [PMID: 32121107 PMCID: PMC7139301 DOI: 10.3390/cancers12030560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022] Open
Abstract
The effectiveness of chemotherapy and radiotherapy to treat lung cancer is limited because of highly metastatic nature. Novel strategies and drugs to attenuate metastatic activity are urgently required. In this study, red fluorescence proteins (RFP)-labeled A549 human lung cancer cells were orthotopically implantation, where they developed primary tumors. Metastasis in brain and intestines were reduced by up to 80% by treatment with 100 mpk 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) compared with that in control mice. PLAG treatment also reduced the migration of the primary tumors. Interestingly, substantial neutrophil infiltration was observed in the tumors in control mice. The neutrophil contribution to A549 cell metastatic activity was examined in in vitro co-culture system. Metastatic activity could be achieved in the A549 cells through epidermal growth factor receptor (EGFR) transactivation mediated by protease activating receptor 2 (PAR2) receptor. Neutrophil elastase secreted from tumor-infiltrating neutrophils stimulated PAR2 and induced EGFR transactivation. However, this transactivation was inhibited by inducing PAR2 degradation following PLAG treatment and metastatic activity was effectively inhibited. PLAG attenuated cancer metastatic activity via modulated PAR2/EGFR transactivation by accelerating PAR2 degradation. These results suggest PLAG as potential therapeutic agent to combat tumor metastasis via regulating the activation signal pathway of PAR2 by tumor infiltrate-neutrophils.
Collapse
|
15
|
Villa A, Sonis ST. An update on pharmacotherapies in active development for the management of cancer regimen-associated oral mucositis. Expert Opin Pharmacother 2020; 21:541-548. [DOI: 10.1080/14656566.2020.1718652] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Alessandro Villa
- Divisions of Oral Medicine and Dentistry, Brigham and Women’s Hospital and the Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Oral Medicine, Infection and Immunity. Harvard School of Dental Medicine, Boston, MA, USA
- Primary Endpoint Solutions, Watertown, MA, USA
| | - Stephen T. Sonis
- Divisions of Oral Medicine and Dentistry, Brigham and Women’s Hospital and the Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Oral Medicine, Infection and Immunity. Harvard School of Dental Medicine, Boston, MA, USA
- Primary Endpoint Solutions, Watertown, MA, USA
| |
Collapse
|
16
|
Mitigating Effect of 1-Palmitoyl-2-Linoleoyl-3-Acetyl-Rac-Glycerol (PLAG) on a Murine Model of 5-Fluorouracil-Induced Hematological Toxicity. Cancers (Basel) 2019; 11:cancers11111811. [PMID: 31752148 PMCID: PMC6896120 DOI: 10.3390/cancers11111811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 12/19/2022] Open
Abstract
5-Fluorouracil (5-FU) is an antimetabolite chemotherapy widely used for the treatment of various cancers. However, many cancer patients experience hematological side effects following 5-FU treatment. Here, we investigated the protective effects of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) as a mitigator against 5-FU-induced hematologic toxicity, including neutropenia, monocytopenia, thrombocytopenia, and thrombocytosis, in Balb/c mice injected with 5-FU (100 mg/kg, i.p.). Administration of PLAG significantly and dose-dependently reduced the duration of neutropenia and improved the nadirs of absolute neutrophil counts (ANCs). Moreover, while the ANCs of all mice in the control fell to the severely neutropenic range, none of the mice in the PLAG 200 and 400 mg/kg-treated groups experienced severe neutropenia. Administration of PLAG significantly delayed the mean first day of monocytopenia and reduced the duration of monocytopenia. PLAG also effectively reduced extreme changes in platelet counts induced by 5-FU treatment, thus preventing 5-FU-induced thrombocytopenia and thrombocytosis. PLAG significantly decreased plasma levels of the chemokine (C–X–C motif) ligand 1 (CXCL1), CXCL2, interleukin (IL)-6, and C-reactive protein (CRP), which were elevated consistently with the occurrence time of neutropenia, monocytopenia, and thrombocytopenia. When compared with olive oil and palmitic linoleic hydroxyl glycerol (PLH), only PLAG effectively mitigated 5-FU-induced hematological toxicity, indicating that it has a distinctive mechanism of action. In conclusion, PLAG may have therapeutic potential as a mitigator for 5-FU-induced neutropenia and other hematological disorders.
Collapse
|
17
|
1-Palmitoyl-2-Linoleoyl-3-Acetyl- rac-Glycerol Attenuates Streptozotocin-Induced Pancreatic Beta Cell Damage by Promoting Glucose Transporter 2 Endocytosis. Mol Cell Biol 2019; 39:MCB.00157-19. [PMID: 31481450 PMCID: PMC6791655 DOI: 10.1128/mcb.00157-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/14/2019] [Indexed: 01/09/2023] Open
Abstract
Streptozotocin (STZ) is widely used to induce diabetic rodent models. It is specifically toxic to pancreatic beta cells and causes severe destruction and dysfunction. We investigated the effect of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) on an STZ-induced diabetic mouse model. PLAG attenuated the glucose increase and maintained serum insulin at levels similar to those seen with control mice. Streptozotocin (STZ) is widely used to induce diabetic rodent models. It is specifically toxic to pancreatic beta cells and causes severe destruction and dysfunction. We investigated the effect of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) on an STZ-induced diabetic mouse model. PLAG attenuated the glucose increase and maintained serum insulin at levels similar to those seen with control mice. In pancreatic beta cell line INS-1, STZ-induced cell apoptosis and intracellular reactive oxygen species (ROS) generation were significantly reduced to nearly normal levels after PLAG treatment. Glucose transporter 2 (GLUT2) localization analyses and glucose uptake assays showed that PLAG accelerated GLUT2 internalization, which ameliorated excessive entry of glucose, as well as STZ. STZ-induced cytotoxic effects were significantly reduced in PLAG-treated groups. The biological activity of PLAG was further confirmed in GLUT2-silenced cells, and the specificity of PLAG was verified using its derivative 1-palmitoyl-2-linoleoyl-3-hydroxyl-rac-glycerol (PLH). Our results suggest that PLAG may be a useful agent for protecting beta cells in the setting of excessive glucose influx.
Collapse
|
18
|
Kim YJ, Jeong J, Shin SH, Lee DY, Sohn KY, Yoon SY, Kim JW. Mitigating Effects of 1-Palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) on Hematopoietic Acute Radiation Syndrome after Total-Body Ionizing Irradiation in Mice. Radiat Res 2019; 192:602-611. [DOI: 10.1667/rr15440.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Yong-Jae Kim
- Division of Global New Drug Development, Enzychem Lifesciences, Jecheon 27159, Republic of Korea
| | - Jinseon Jeong
- Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Su-Hyun Shin
- Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Do Young Lee
- Division of Global New Drug Development, Enzychem Lifesciences, Jecheon 27159, Republic of Korea
| | - Ki-Young Sohn
- Division of Global New Drug Development, Enzychem Lifesciences, Jecheon 27159, Republic of Korea
| | - Sun Young Yoon
- Division of Global New Drug Development, Enzychem Lifesciences, Jecheon 27159, Republic of Korea
| | - Jae Wha Kim
- Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| |
Collapse
|
19
|
Lee HR, Shin SH, Kim JH, Sohn KY, Yoon SY, Kim JW. 1-Palmitoyl-2-Linoleoyl-3-Acetyl-rac-Glycerol (PLAG) Rapidly Resolves LPS-Induced Acute Lung Injury Through the Effective Control of Neutrophil Recruitment. Front Immunol 2019; 10:2177. [PMID: 31620122 PMCID: PMC6759802 DOI: 10.3389/fimmu.2019.02177] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 08/28/2019] [Indexed: 12/30/2022] Open
Abstract
Acute lung injury (ALI) is an acute respiratory failure that is associated with excessive neutrophil recruitment and high mortality. To assess the efficacy of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) as a therapeutic agent for ALI, this compound was administered orally to mice challenged with an intranasal dose of lipopolysaccharide (LPS). Using this model, we found that PLAG promotes resolution of ALI through effective control of LPS-induced neutrophil infiltration, endothelial permeability, and inflammatory chemokine production. In addition, the Toll like Receptor 4 (TLR4) endocytosis/exocytosis cycle was significantly accelerated in Raw 264.7 cells co-treated with PLAG/LPS, as compared to cells treated only with LPS. During this cycle, a PLAG-induced exotoxin clearance pathway was observed to occur through the prompt assembly of nicotinamide adenine dinucleotide phosphate (NADPH) units and production of reactive oxygen species (ROS), which ultimately lead to earlier LPS clearance. We further detected reduced expression, as well as faster return to homeostatic levels, of macrophage inflammatory protein (MIP)-2, in PLAG/LPS- vs. LPS-treated cells. MIP-2 is a main inducer of neutrophil migration that is mainly controlled by interferon regulatory factor 3 (IRF3) activation and is involved in the TLR4 endosomal-signaling pathway. PLAG induced TLR4-mediated TRIF-related adaptor molecules/Toll-interleukin receptor (TIR) domain-containing adaptor protein including interferon (IFN)-β/IRF3 endosomal signaling, leading to rapid association of TRAM/TRIF and TLR4 and earlier IRF3 phosphorylation in PLAG/LPS-treated vs. LPS-treated cells. PLAG specificity was further verified with PLAG analogs and metabolites known to control excessive neutrophil infiltration, suggesting that this acetylated diacylglycerol has a unique biological role in neutrophil motility. Thus, our data indicate that PLAG may represent a potential therapeutic agent for resolution of LPS-induced lung inflammation through effective MIP-2 modulation.
Collapse
Affiliation(s)
| | - Su-Hyun Shin
- Division of Systems Biology and Bioengineering, Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea
| | - Joo Heon Kim
- Department of Pathology, EulJi University School of Medicine, Daejeon, South Korea
| | | | | | - Jae Wha Kim
- Division of Systems Biology and Bioengineering, Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
20
|
Jeong J, Kim YJ, Lee DY, Moon BG, Sohn KY, Yoon SY, Kim JW. 1-Palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) attenuates gemcitabine-induced neutrophil extravasation. Cell Biosci 2019; 9:4. [PMID: 30622698 PMCID: PMC6317242 DOI: 10.1186/s13578-018-0266-7] [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: 10/04/2018] [Accepted: 12/24/2018] [Indexed: 11/10/2022] Open
Abstract
Cancer patients treated with chemotherapy often experience a rapid decline of blood neutrophils, a dose-limiting side effect called chemotherapy-induced neutropenia. This complication brings about dose reductions or cessation of chemotherapy during treatment of cancer patients because a rapid decline of neutrophil counts increases susceptibility to infection. Here, we found that 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) attenuates gemcitabine-induced neutrophil extravasation via the inhibition of neutrophil-attracting chemokine production in macrophages using in vivo and in vitro approaches. A single intraperitoneal administration of gemcitabine induced the migration of circulating neutrophils into the peritoneal cavity in normal mice, and PLAG effectively decreased neutrophil migration by inhibiting the expression of adhesion molecules, L-selectin and LFA-1. Inhibition of CXCR2 by its antagonist, reparixin, abrogated gemcitabine-induced neutrophil migration, indicating that chemokines produced by gemcitabine mainly support neutrophil activation. In vitro experiments demonstrated that PLAG inhibited NADPH oxidase 2 (NOX2)-mediated reactive oxygen species production induced by gemcitabine, which is the upstream of MIP-2 and/or CXCL8. Importantly, PLAG down-regulated gemcitabine-induced membrane translocation of the cytosolic NOX subunit, Rac1, and phosphorylation of p47phox. The activation of upstream signaling molecules of p47phox phosphorylation, phospholipase C β3 and protein kinase C, were effectively regulated by PLAG. We also demonstrated that 1-palmitoyl-2-linoleic-3-hydroxyl-rac-glycerol (PLH), the natural form of diacylglycerol, has no effects on gemcitabine-induced CXCL8 production and dHL-60 migration, suggesting that an acetyl group at the third position of the glycerol backbone may have a key role in the regulation of neutrophil activation. Altogether, this study suggests the potential of PLAG as a therapeutic strategy to modulate chemotherapy-induced neutrophil activation for cancer patients undergoing chemotherapeutic treatment.
Collapse
Affiliation(s)
- Jinseon Jeong
- 1Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 305-333 Republic of Korea.,2Department of Functional Genomics, University of Science & Technology, Daejeon, Republic of Korea.,Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Yong-Jae Kim
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Do Young Lee
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Byoung-Gon Moon
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Ki-Young Sohn
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Sun Young Yoon
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Jae Wha Kim
- 1Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 305-333 Republic of Korea.,2Department of Functional Genomics, University of Science & Technology, Daejeon, Republic of Korea
| |
Collapse
|
21
|
Park JW, Oh J, Ko SJ, Chang MS, Kim J. Effects of Onchung-eum, an Herbal Prescription, on 5-Fluorouracil-Induced Oral Mucositis. Integr Cancer Ther 2018; 17:1285-1296. [PMID: 30296855 PMCID: PMC6247538 DOI: 10.1177/1534735418805560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
In most cancer patients, chemotherapy-induced oral mucositis (OM) is a frequent
side effect, leading to low quality of life and delay in therapy. The aim of
this study was to evaluate the effects of Onchung-eum, a
well-known herbal prescription in traditional medicine comprising 8 herbs that
has long been used for skin diseases, on 5-fluorouracil (5-FU)–induced OM in
human pharyngeal cells and golden Syrian hamsters. DPPH
(2,2-diphenyl-1-picrylhydrazyl) radical scavenging activity, MTT
(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, and
reactive oxygen species production were measured in vitro. The effects of
Onchung-eum on OM of hamster cheek pouches induced by 5-FU
were evaluated histologically and using TUNEL assay. In addition, the expression
of nuclear factor-κB, caspase-3, and pro-inflammatory cytokines were measured by
immunoblotting and immunohistochemistry. Significantly increased cell viability
was observed in the Onchung-eum–treated groups compared with
the 5-FU–treated control group. In 500 and 1000 mg/kg
Onchung-eum–treated groups, the damaged epithelial layers
in the cheek pouches of hamsters were significantly recovered. Moreover, at all
concentrations, cell death in the cheek pouches of hamsters in the
Onchung-eum–treated groups significantly decreased. The
expression of pro-inflammatory cytokines, nuclear factor-κB, and caspase-3 also
significantly decreased in Onchung-eum–treated groups at 500
and 1000 mg/kg. In conclusion, this study revealed that
Onchung-eum can be used to treat chemotherapy-induced OM.
However, further studies are required to understand the underlying
mechanisms.
Collapse
Affiliation(s)
- Jae-Woo Park
- 1 Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Jayoung Oh
- 2 Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Seok-Jae Ko
- 1 Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Mun Seog Chang
- 3 Department of Prescriptionology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Jinsung Kim
- 1 Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
22
|
Sobue T, Bertolini M, Thompson A, Peterson DE, Diaz PI, Dongari-Bagtzoglou A. Chemotherapy-induced oral mucositis and associated infections in a novel organotypic model. Mol Oral Microbiol 2018; 33:212-223. [PMID: 29314782 PMCID: PMC5945319 DOI: 10.1111/omi.12214] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2017] [Indexed: 12/31/2022]
Abstract
Oral mucositis is a common side effect of cancer chemotherapy, with significant adverse impact on the delivery of anti-neoplastic treatment. There is a lack of consensus regarding the role of oral commensal microorganisms in the initiation or progression of mucositis because relevant experimental models are non-existent. The goal of this study was to develop an in vitro mucosal injury model that mimics chemotherapy-induced mucositis, where the effect of oral commensals can be studied. A novel organotypic model of chemotherapy-induced mucositis was developed based on a human oral epithelial cell line and a fibroblast-embedded collagen matrix. Treatment of organotypic constructs with 5-fluorouracil (5-FU) reproduced major histopathologic characteristics of oral mucositis, such as DNA synthesis inhibition, apoptosis and cytoplasmic vacuolation, without compromising the three-dimensional structure of the multilayer organotypic mucosa. Although structural integrity of the model was preserved, 5-FU treatment resulted in a widening of epithelial intercellular spaces, characterized by E-cadherin dissolution from adherens junctions. In a neutrophil transmigration assay we discovered that this treatment facilitated transport of neutrophils through epithelial layers. Moreover, 5-FU treatment stimulated key proinflammatory cytokines that are associated with the pathogenesis of oral mucositis. 5-FU treatment of mucosal constructs did not significantly affect fungal or bacterial biofilm growth under the conditions tested in this study; however, it exacerbated the inflammatory response to certain bacterial and fungal commensals. These findings suggest that commensals may play a role in the pathogenesis of oral mucositis by amplifying the proinflammatory signals to mucosa that is injured by cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Takanori Sobue
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
| | - Martinna Bertolini
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
| | - Angela Thompson
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
| | - Douglas E. Peterson
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
| | - Patricia I. Diaz
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
| | - Anna Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, 263 Farmington Avenue, Farmington, CT 06030 USA
| |
Collapse
|
23
|
Marcussen M, Skrubbeltrang C, Bødker JS, Christiansen I, Bøgsted M, Dybkær K, Bergmann OJ, Johnsen HE. A systematic review of molecular responses to cancer therapy in normal human mucosa. Oral Surg Oral Med Oral Pathol Oral Radiol 2017; 124:355-366. [PMID: 29042035 DOI: 10.1016/j.oooo.2017.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 07/24/2017] [Accepted: 08/01/2017] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Cancer therapy-induced inflammation of oral and gastrointestinal mucosae affects patients nonuniformly. Preventive strategies are limited; no biomarker exists for pretreatment identification of patients likely to be severely affected. Animal models are preferred for studying molecular responses in mucosae during chemotherapy, but translation into clinical practice is difficult. We performed a systematic review to retrieve articles that described molecular changes in human mucosae during cancer therapy. STUDY DESIGN We searched MEDLINE and Ovid Embase searches for studies reported in the English language literature from January 1990 to November 2016 and studies referenced in selected articles, which analyzed mucosae from patients at risk of developing mucositis during cancer therapy. Two authors extracted data according to predefined data fields, including study quality indicators. RESULTS We identified 17 human studies on chemotherapy (n = 9) and radiotherapy (n = 8), but no studies on targeted therapy. Studies were heterogeneous with regard to patient cohorts, analysis methods, cancer treatments, biopsy timings, and correlations to clinical mucositis. Consequently, a meta-analysis was not feasible. CONCLUSIONS Few human studies described the molecular responses of the normal mucosa to cancer therapy. Studies were heterogeneous and had sparse correlations to clinical mucositis. We proposed a model for acquiring data on treatment- and disease-specific phenotypes and transcriptomes for predictive or preventive initiatives.
Collapse
Affiliation(s)
- Mette Marcussen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | | | - Julie Støve Bødker
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Ilse Christiansen
- Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| | - Martin Bøgsted
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| | - Karen Dybkær
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| | - Olav Jonas Bergmann
- School of Dentistry, Faculty of Health Science, Aarhus University; Aarhus, Denmark
| | - Hans Erik Johnsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
24
|
Kim YJ, Shin JM, Shin SH, Kim JH, Sohn KY, Kim HJ, Kang JK, Yoon SY, Kim JW. 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol ameliorates arthritic joints through reducing neutrophil infiltration mediated by IL-6/STAT3 and MIP-2 activation. Oncotarget 2017; 8:96636-96648. [PMID: 29228558 PMCID: PMC5722510 DOI: 10.18632/oncotarget.19384] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 07/06/2017] [Indexed: 12/24/2022] Open
Abstract
The pathogenesis of rheumatoid arthritis (RA) has been implicated neutrophil extracellular traps (NETs) formation which could generate autoantigen. Neutrophil contributes to initiate and maintain the inflammatory process in the joint. In this study, we show that 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) decreases neutrophil migration by regulating the activity of STAT3, a regulator of IL-6 and MIP-2 expression. PLAG caused a decrease in IL-6 production in the RAW264.7 macrophage cell line and in rheumatoid arthritis–fibroblast-like synoviocytes via the regulation of STAT3 signaling without affecting NF-κB signaling. In a mouse model of collagen-induced arthritis (CIA), arthritic symptoms were recapitulated, with increased IL-6 level in the synovium, and PLAG treatment restored IL-6 to a level comparable to that achieved with commercial therapeutics (such as Remicade or methotrexate). Staining of joint tissue with neutrophil-specific antibody showed that PLAG significantly reduced the infiltration of neutrophils into the joint synovium of CIA mice. The inhibitory effect of PLAG on IL-6/STAT3 or MIP-2 signaling also reduced the migration of differentiated neutrophils in vitro. Therefore, PLAG inhibits the infiltration of destructive neutrophils into inflammatory sites, and can be utilized as a potent therapeutic agent for the treatment of sustained inflammation and joint destruction.
Collapse
Affiliation(s)
- Young-Jun Kim
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jae Min Shin
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Su-Hyun Shin
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Joo Heon Kim
- Department of Pathology, EulJi University School of Medicine, Daejeon, Republic of Korea
| | | | | | - Jong-Koo Kang
- Department of Laboratory Animal medicine, College of Veterinary medicine, Chungbuk National University, Cheongju, Republic of Korea
| | | | - Jae Wha Kim
- Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| |
Collapse
|