1
|
Li B, Zhang T, Tan G, Pu Z, Shen Y. Neuroprotective Effects of Astragalus Polysaccharide on Retina Cells and Ganglion Cell Projection in NMDA-Induced Retinal Injury. Curr Eye Res 2024:1-13. [PMID: 39373214 DOI: 10.1080/02713683.2024.2412304] [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: 02/22/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
PURPOSE Astragalus polysaccharide (APS), a water-soluble heteropolysaccharide, possesses immunomodulatory, anti-inflammatory, and cardioprotective properties. This study investigates the neuroprotective potential of APS in a model of N-Methyl-d-aspartic acid (NMDA)-induced retinal neurodegeneration, aiming to explore its potential as a treatment for retinal degenerative diseases. METHODS Retinal function was evaluated using electroretinography (ERG), optomotor reflex (OMR), and flash visual evoked potentials (FVEP). Retinal inflammatory responses were examined through immunohistochemistry, western blotting (WB), and quantitative reverse transcription PCR (qRT-PCR). To assess the integrity of visual projections, an intravitreal injection of adeno-associated virus (AAV) was employed to trace the projections of retinal ganglion cells (RGCs) to the visual centers. RESULTS APS treatment conferred protection to retinal cells, as indicated by ERG and OMR assessments. And APS intervention mitigated NMDA-induced apoptosis, evidenced by a decrease in TUNEL-positive cells. Furthermore, APS treatment attenuated the NMDA-induced reduction in RGC projections to the visual centers, including the superior colliculus and lateral geniculate nucleus, as demonstrated by AAV tracing. CONCLUSIONS Our findings reveal that APS shields the retina from NMDA-induced damage by inhibiting the NF-κB signaling pathway and reduces the detrimental effects of NMDA on RGC projections to the visual centers. These findings propose APS as a potential novel therapeutic agent for the treatment of retinal diseases.
Collapse
Affiliation(s)
- Baige Li
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Tianlu Zhang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Gao Tan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Zeyuan Pu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, P. R. China
- Frontier Science Center for lmmunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, P. R. China
| |
Collapse
|
2
|
Choi SJ, Lee SJ, Lee D, Im GJ, Jung HH, Lee SU, Park E. Protective Effect of Memantine on Cisplatin-Induced Ototoxicity: An In Vitro Study. Otol Neurotol 2024; 45:998-1005. [PMID: 39186064 DOI: 10.1097/mao.0000000000004317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
HYPOTHESIS Memantine, an N -methyl- d -aspartate receptor antagonist, is widely used to treat Alzheimer's disease and has been found to have potential neuroprotective effects. In this study, we evaluated the protective effects of memantine against cisplatin-induced ototoxicity. BACKGROUND Cisplatin is a widely used anticancer drug for various cancers; however, its use is limited by its side effects, including ototoxicity. Several drugs have been developed to reduce cisplatin toxicity. In this study, we treated cisplatin-damaged cochlear hair cells with memantine and evaluated its protective effects. METHOD House Ear Institute Organ of Corti 1 (HEI-OC1) cells and cochlear explants were treated with cisplatin or memantine. Cell viability, apoptotic patterns, reactive oxygen species (ROS) production, Bcl-2/caspase-3 activity, and cell numbers were measured to evaluate the anti-apoptotic and antioxidative effects of memantine. RESULT Memantine treatment significantly improved cell viability and reduced cisplatin-induced apoptosis in auditory cells. Bcl-2/caspase-3 activity was also significantly increased, suggesting anti-apoptotic effects against cisplatin-induced ototoxicity. CONCLUSION Our results suggest that memantine protects against cisplatin-induced ototoxicity in vitro, providing a potential new strategy for preventing hearing loss in patients undergoing cisplatin chemotherapy.
Collapse
Affiliation(s)
- Soo Jeong Choi
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Soo Jin Lee
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Dabin Lee
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Gi Jung Im
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hak Hyun Jung
- Department of Otolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | | | | |
Collapse
|
3
|
Basak M, Kulkarni M, Narisepalli S, Chitkara D, Mittal A. Exosomal fragment enclosed polyamine-salt nano-complex for co-delivery of docetaxel and mir-34a exhibits higher cytotoxicity and apoptosis in breast cancer cells. Sci Rep 2024; 14:21669. [PMID: 39289425 PMCID: PMC11408524 DOI: 10.1038/s41598-024-72226-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
A novel core-shell nanocarrier system has been designed for co-delivery of a small anticancer drug, docetaxel (DTX) and tumor suppressor (TS) miR-34a named as Exo(PAN34a+DTX). The core is formed by pH dependent polyamine salt aggregates (PSA) containing both the payloads and the shell is formed by RAW 264.7 cell derived exosomal fragments. Herein, phosphate driven polyallylamine hydrochloride (PAH, MW:17,500 Da) PSA was formed in presence of miR-34a and DTX to form PAN34a+DTX. The formulation exhibited pH dependent DTX release with only 33.55 ± 2.12% DTX release at pH 7.2 and 75.21 ± 1.8% DTX release till 144 h at pH 5.5. At 1.21 molar ratio of phosphate to the amine (known as R value), efficient complexation of miR-34a (3.6 μM) in the PAN particles was obtained. PAN34a+DTX demonstrated particle size (163.86 ± 12.89 nm) and zeta-potential value of 17.53 ± 5.10 mV which upon exosomal fragment layering changed to - 7.23 ± 2.75 mV which is similar to the zeta-potential of the exosomal fragments, i.e., - 8.40 ± 1.79 mV. The final formulation Exo(PAN34a+DTX), loaded with 40 ng/mL DTX and 50 nM miR-34a exhibited 48.20 ± 4.59% cytotoxicity in triple negative breast cancer (TNBC) cells, 4T1. Co-localization of CM-DiI (red fluorescence) stained exosomal fragments and FAM-siRNA (green fluorescence) in the cytoplasm of 4T1 cells after 6 h of Exo(PANFAM) treatment confirmed the efficiency of the designed system to co-deliver two actives. Exo(PAN34a+DTX) also reduced BCL-2 expression (target gene for miR-34a) by 8.98 folds in comparison to free DTX confirming promising co-delivery and apoptosis inducing effect of Exo(PAN34a+DTX) in 4T1.
Collapse
Affiliation(s)
- Moumita Basak
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Mrunal Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Saibhargav Narisepalli
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science (BITS PILANI), Pilani Campus, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
4
|
Jin JJ, Ko IG, Hwang L, Kim SH, Jee YS, Jeon H, Park SB, Jeon JW. Simultaneous Treatment of 5-Aminosalicylic Acid and Treadmill Exercise More Effectively Improves Ulcerative Colitis in Mice. Int J Mol Sci 2024; 25:5076. [PMID: 38791116 PMCID: PMC11120947 DOI: 10.3390/ijms25105076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Ulcerative colitis (UC) is characterized by continuous mucosal ulceration of the colon, starting in the rectum. 5-Aminosalicylic acid (5-ASA) is the main therapy for ulcerative colitis; however, it has side effects. Physical exercise effectively increases the number of anti-inflammatory and anti-immune cells in the body. In the current study, the effects of simultaneous treatment of treadmill exercise and 5-ASA were compared with monotherapy with physical exercise or 5-ASA in UC mice. To induce the UC animal model, the mice consumed 2% dextran sulfate sodium dissolved in drinking water for 7 days. The mice in the exercise groups exercised on a treadmill for 1 h once a day for 14 days after UC induction. The 5-ASA-treated groups received 5-ASA by enema injection using a 200 μL polyethylene catheter once a day for 14 days. Simultaneous treatment improved histological damage and increased body weight, colon weight, and colon length, whereas the disease activity index score and collagen deposition were decreased. Simultaneous treatment with treadmill exercise and 5-ASA suppressed pro-inflammatory cytokines and apoptosis following UC. The benefits of this simultaneous treatment may be due to inhibition on nuclear factor-κB/mitogen-activated protein kinase signaling activation. Based on this study, simultaneous treatment of treadmill exercise and 5-ASA can be considered as a new therapy of UC.
Collapse
Affiliation(s)
- Jun-Jang Jin
- Team of Efficacy Evaluation, Orient Genia Inc., Seongnam 13201, Republic of Korea; (J.-J.J.); (L.H.)
| | - Il-Gyu Ko
- Research Support Center, School of Medicine, Keimyung University, Deagu 42601, Republic of Korea;
| | - Lakkyong Hwang
- Team of Efficacy Evaluation, Orient Genia Inc., Seongnam 13201, Republic of Korea; (J.-J.J.); (L.H.)
| | - Sang-Hoon Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The Stat University of New Jersey, Piscataway, NJ 08854, USA;
| | - Yong-Seok Jee
- Research Institute of Sports and Industry Science, Hanseo University, Seosan 31962, Republic of Korea;
| | - Hyeon Jeon
- Department of Computer Science and Engineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea;
| | - Su Bee Park
- Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul 05278, Republic of Korea;
| | - Jung Won Jeon
- Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul 05278, Republic of Korea;
| |
Collapse
|
5
|
Kim EJ, Park H, Kim EY, Kim DK, Jung HS, Sohn Y. Ursodeoxycholic acid alleviates atopic dermatitis-associated inflammatory responses in HaCaT and RBL-2H3 cells and DNCB/DFE-treated mice. Life Sci 2024; 344:122560. [PMID: 38490296 DOI: 10.1016/j.lfs.2024.122560] [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: 05/14/2023] [Revised: 08/18/2023] [Accepted: 03/09/2024] [Indexed: 03/17/2024]
Abstract
AIMS Ursodeoxycholic acid (UDCA) is a hydrophilic dihydroxy bile acid used for cholestatic liver disease and exhibits antioxidant, antitumor, and anti-inflammatory effects. However, its potential effects on atopic dermatitis (AD) have not been elucidated. This study aimed to evaluate the efficacy of UDCA in inhibiting the inflammatory response and alleviating lesions in AD-like mice. MAIN METHODS To investigate the efficacy of UDCA in AD-like inflammatory responses, tumor necrosis factor-alpha (TNF-α)- and interferon-gamma (IFN-γ)-stimulated HaCaT cells and anti-dinitrophenyl immunoglobulin E (DNP-IgE)- and human serum albumin (HSA)-stimulated RBL-2H3 cells were used to investigate the levels of inflammatory factors and their mechanisms. AD-like lesions were induced by applying DNCB/DFE to mice. The effect of UDCA administration in AD-like mice was analyzed by assessing organ weight, serum IgE and inflammatory cytokine levels, and histopathological changes using immunohistochemical and immunofluorescent staining. KEY FINDINGS In HaCaT cells, UDCA significantly diminished TARC, MDC, MCP-1, and IL-6 expression by inhibiting the phosphorylation of nuclear NF-κB and cytoplasmic IκB, and also increased the levels of skin barrier protein. In RBL-2H3 cells, UDCA reduced β-hexosaminidase and IL-4 levels. In AD-like mice, UDCA suppressed organ hypertrophy, ear edema, SCORAD index, DFE-specific IgE levels, inflammatory cytokine levels, skin hypertrophy, mast cell invasion, skin barrier loss, and thymic stromal lymphopoietin-positive areas. SIGNIFICANCE UDCA suppressed the expression of pro-inflammatory cytokines by keratinocytes and mast cells. It also alleviated atopy by suppressing symptoms without organ toxicity in AD-like mice. UDCA may be an effective and safe treatment for AD.
Collapse
Affiliation(s)
- Eom Ji Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Hoyeon Park
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Eun-Young Kim
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Do Kyung Kim
- Department of Anatomy, Konyang University College of Medicine, Daejeon 35365, Republic of Korea
| | - Hyuk-Sang Jung
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Youngjoo Sohn
- Department of Anatomy, College of Korean Medicine, Kyung Hee University, 26, Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
6
|
Li WH, Wang F, Song GY, Yu QH, Du RP, Xu P. PARP-1: a critical regulator in radioprotection and radiotherapy-mechanisms, challenges, and therapeutic opportunities. Front Pharmacol 2023; 14:1198948. [PMID: 37351512 PMCID: PMC10283042 DOI: 10.3389/fphar.2023.1198948] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/22/2023] [Indexed: 06/24/2023] Open
Abstract
Background: Since its discovery, poly (ADP-ribose) polymerase 1 (PARP-1) has been extensively studied due to its regulatory role in numerous biologically crucial pathways. PARP inhibitors have opened new therapeutic avenues for cancer patients and have gained approval as standalone treatments for certain types of cancer. With continued advancements in the research of PARP inhibitors, we can fully realize their potential as therapeutic targets for various diseases. Purpose: To assess the current understanding of PARP-1 mechanisms in radioprotection and radiotherapy based on the literature. Methods: We searched the PubMed database and summarized information on PARP inhibitors, the interaction of PARP-1 with DNA, and the relationships between PARP-1 and p53/ROS, NF-κB/DNA-PK, and caspase3/AIF, respectively. Results: The enzyme PARP-1 plays a crucial role in repairing DNA damage and modifying proteins. Cells exposed to radiation can experience DNA damage, such as single-, intra-, or inter-strand damage. This damage, associated with replication fork stagnation, triggers DNA repair mechanisms, including those involving PARP-1. The activity of PARP-1 increases 500-fold on DNA binding. Studies on PARP-1-knockdown mice have shown that the protein regulates the response to radiation. A lack of PARP-1 also increases the organism's sensitivity to radiation injury. PARP-1 has been found positively or negatively regulate the expression of specific genes through its modulation of key transcription factors and other molecules, including NF-κB, p53, Caspase 3, reactive oxygen species (ROS), and apoptosis-inducing factor (AIF). Conclusion: This review provides a comprehensive analysis of the physiological and pathological roles of PARP-1 and examines the impact of PARP-1 inhibitors under conditions of ionizing radiation exposure. The review also emphasizes the challenges and opportunities for developing PARP-1 inhibitors to improve the clinical outcomes of ionizing radiation damage.
Collapse
Affiliation(s)
- Wen-Hao Li
- School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Fei Wang
- School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Gui-Yuan Song
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Qing-Hua Yu
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Rui-Peng Du
- School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
| | - Ping Xu
- School of Food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong, China
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
7
|
MA Y, WANG Q. Structural characterization of a polysaccharide from Lyophyllum decastes with MAPK-mediated immune regulation ability in mice. FOOD SCIENCE AND TECHNOLOGY 2023. [DOI: 10.1590/fst.003523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Affiliation(s)
- Yisi MA
- Jilin Agricultural University, China; Changchun Second Middle School, China
| | - Qi WANG
- Jilin Agricultural University, China
| |
Collapse
|
8
|
Zhu H, Wang H, Cheng Y, Liu D, Zhang A, Wen Z, Gao J. Hadh deficiency induced oligoasthenoteratozoospermia through the TNF-α/Bcl-2 pathway in male mice. FASEB J 2022; 36:e22661. [PMID: 36398584 DOI: 10.1096/fj.202201144r] [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: 07/18/2022] [Revised: 10/11/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
The process of spermatogenesis is a complex and delicate process that is still not fully understood. In this study, we examined the role of fatty acid oxidase 3-hydroxy acyl CoA dehydrogenase (HADH) in maintaining normal spermatogenesis in mice. In male mice, ablation of the Hadh gene using CRISPR/Cas9 technology arrested spermatocyte meiosis, increased multinucleated giant germ cells and vacuoles in seminiferous tubules, and accompanied with acrosomal dysplasia. Hadh-/- male mice showed the typical features of oligoasthenoteratozoospermia (OAT), including decreased sperm concentration and motility and increased sperm abnormalities. Next, we explored the molecular events in the testes of the mutant mice. We found fatty acids accumulated in the testis of Hadh-/- mice. And also, inflammatory factors TNF-α, IL-1β, and IL-6 were significantly increased, apoptosis-related protein Bcl-2 was decreased, and Bax and cleaved-Caspase3 were increased in Hadh-/- male mice testis. After using etanercept, a specific inhibitor of TNF-α, testis injury caused by Hadh knockout was significantly alleviated, the sperm quality and motility were improved, and germ cell apoptosis was reduced. So our study demonstrated that Hadh deletion caused an increase in fatty acids. The accumulated fatty acids further induced testicular inflammation and germ cell apoptosis through the TNF-α/Bcl-2 signaling pathway, finally resulting in OAT in the Hadh-/- mice. Inhibiting TNF-α may be used as a new treatment approach for testicular inflammation and OAT.
Collapse
Affiliation(s)
- Haixia Zhu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Hongxiang Wang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Yin Cheng
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Dongyue Liu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Aizhen Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Zongzhuang Wen
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China.,Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| |
Collapse
|
9
|
Modena DAO, Soares CD, Martignago CCS, Almeida S, Cazzo E, Chaim EA. Effects of LED photobiomodulation therapy on the subcutaneous fatty tissue of obese individuals - histological and immunohistochemical analysis. J COSMET LASER THER 2022; 24:84-90. [PMID: 36074934 DOI: 10.1080/14764172.2022.2109677] [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: 10/14/2022]
Abstract
Photobiomodulation therapy (PBMT) has become an adjuvant therapeutic possibility in body remodeling procedures. Given this scenario, this study was proposed with the aim of evaluating the effects of PBMT to Light Emitting Diode (LED) associating the red (630 nm) and infrared (850 nm) wavelengths in the subcutaneous fatty tissue. This controlled study of comparative intervention that evaluated a sample of subcutaneous fatty tissue from women with grade II obesity. The participants received the LED PBMT treatment with associated red and infrared wavelengths sequentially on the left side of the abdomen and the right side was considered as control, with the collection of biological material performed at the time of bariatric surgery. For histological and immunohistochemical evaluation, Caspase 3, Cleaved Caspase 3, CD68+, HSL and adipophilin markers were used. The participants showed positivity in the expression of Caspase 3 and Cleaved Caspase (p < .0001), CD68+ macrophages (p < .0001), HSL (p < .0001) and adipophilin (p < .0013) in the intervention sample when compared to the control. PBMT and LED associating red and infrared wavelengths were able to promote autophagic lipolysis induced by adipocyte cell apoptosis in the subcutaneous tissue of obese individuals.
Collapse
Affiliation(s)
| | - Ciro Dantas Soares
- Oral Pathology Section, Department of Oral Diagnosis, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | | | - Stephani Almeida
- Department of Surgery, Medical Sciences Institute, Campinas University (Unicamp), São Paulo, Brazil
| | - Everton Cazzo
- Department of Surgery, Medical Sciences Institute, Campinas University (Unicamp), São Paulo, Brazil
| | - Elinton Adami Chaim
- Department of Surgery, Medical Sciences Institute, Campinas University (Unicamp), São Paulo, Brazil
| |
Collapse
|
10
|
Wang TY, Hu YJ, Wang X, Li YF, Zhang F, Yan YD, Dou WT, Cheng CY, Xu P. Targeting p65 to inhibit Cas3 transcription by Onjisaponin B for radiation damage therapy in p65+/- mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154317. [PMID: 35816993 DOI: 10.1016/j.phymed.2022.154317] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/25/2022] [Accepted: 07/03/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND In response to radiation injury, p65 becomes activated. The formation of p65 is one target of Onjisaponin B (OB), but it has not been studied in radioprotection. In addition, there is a binding site for p65 in the promoter region of Cas3. This study evaluates the use of OB as an intervention to modulate p65/Cas3 following radiation exposure. PURPOSE This study aimed to confirm that OB regulated the transcription of Cas3 via p65 to overcome radiation-induced damage. STUDY DESIGN AND METHODS Cells and mice were exposed to X-rays at a dose of 6 Gy. Immunofluorescence was used to locate intracellular p65. For the protein and mRNA analyses, Western blotting and RT-qPCR-based assays were conducted accordingly. HE staining was used to observe pathological changes in tissues. DNA damage was detected by the comet assay and DNA ladder assay. Next, apoptosis was detected by flow cytometry and Hoechst staining. RESULTS Compared with the radiation group, the expression levels of p-p65 and c-Cas3 in the drug group were significantly down-regulated by OB 20 µg/ml. When the expression of p65 was suppressed in V79 and TC cells, OB did not significantly inhibit the activation of p65 or Cas3 in response to irradiation, nor did it significantly inhibit the phosphorylation of p65 and subsequent nuclear translocation. Overexpression of p65 in V79 and MTEC-1 cells resulted in OB significantly inhibiting the activation of p65 and Cas3, and the phosphorylation and translocation of p65 into the nucleus. At 3 d for V79 cells and 24 h for MTEC-1 cells after radiation, compared with the Cas3 over plasmid transfection group, the drug transfection group had no significant effect on reducing apoptosis. In p65+/- mice, expression of the p65 gene was knocked down, leading to increased tissue apoptosis and inflammation, and serious tissue pathological changes. The inhibition of p65 activation by OB after radiation exposure was not apparent in the thymus, although it was observed in the lung. CONCLUSIONS OB interfered with radiation injury by targeting and regulating p65/Cas3. Therefore, it has been concluded that p65 is an important target molecule for the treatment of radiation injury.
Collapse
Affiliation(s)
- Tao-Yang Wang
- School of food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong 277160, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Yong-Jian Hu
- School of food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong 277160, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Xia Wang
- College of Medical Laboratory, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Yu-Feng Li
- Radiology Laboratory, Central laboratory, Rizhao people's Hospital, Rizhao, Shandong 276800, China
| | - Fan Zhang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Yi-di Yan
- Basic Medical school, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Wen-Tao Dou
- Basic Medical school, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Chen-Yi Cheng
- Basic Medical school, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Ping Xu
- School of food and Biomedicine, Zaozhuang University, Zaozhuang, Shandong 277160, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, China.
| |
Collapse
|
11
|
Zhang S, He W, Li A, Zhao C, Chen Y, Xu C, Zhang Q, Zheng D, Chen M, Miao H, Huang Y. Involvement of the TNF-α/SATB2 axis in the induced apoptosis and inhibited autophagy of osteoblasts by the antipsychotic Risperidone. Mol Med 2022; 28:46. [PMID: 35505281 PMCID: PMC9066868 DOI: 10.1186/s10020-022-00466-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Risperidone, an atypical antipsychotic, impedes serotonin and dopamine receptor systems. Meanwhile, tumor necrosis factor-α (TNF-α) is known to participate in regulating osteoblast functions. Consequently, the current study aimed to investigate whether the influences of Risperidone on osteoblast functions are associated with TNF-α and special AT-rich sequence-binding protein (SATB2). METHODS Firstly, we searched the DGIdb, MEM and GeneCards databases to identify the critical factors involved in the effects of Risperidone on osteoblasts, as well as their interactions. Afterwards, osteoblast cell line MC3T3-E1 was transduced with lentivirus carrying si-TNF-α, si-SATB2 or both and subsequently treated with Risperidone. Various abilities including differentiation, autophagy and apoptosis of osteoblasts were examined after different treatments. Finally, animal experiments were performed with Risperidone alone or together with lentivirus to verify the function of Risperidone in vivo and the mechanism. RESULTS It was found that Risperidone might promote TNF-α expression, thereby inhibiting the expression of SATB2 to affect the autophagy and apoptosis in osteoblasts. Furthermore, as shown by our experimental findings, Risperidone treatment inhibited the differentiation and autophagy, and promoted the apoptosis of osteoblasts, as evidenced by elevated levels of OPG, p62, cleaved PARP1, cleaved caspase-3, cleaved caspase-8, and cleaved caspase-9, and reduced levels of LC3 II/I, Beclin1, collagen I, and RANKL. In addition, Risperidone was also found to elevate the expression of TNF-α to down-regulate SATB2, thereby inhibiting the differentiation and autophagy and enhancing the apoptosis of osteoblasts in vitro and in vivo. CONCLUSIONS Collectively, our findings indicated that Risperidone affects the differentiation of osteoblasts by inhibiting autophagy and enhancing apoptosis via TNF-α-mediated down-regulation of SATB2.
Collapse
Affiliation(s)
- Shuyao Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Wei He
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Aiguo Li
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Chengkuan Zhao
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Yun Chen
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Chengcheng Xu
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Qiuzhen Zhang
- Department of Pharmacy, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Danling Zheng
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Meini Chen
- Department of Pharmacology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
| | - Haixiong Miao
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, 510220, China
- Department of Pediatrics, Guangzhou Red Cross Hospital, Jinan University, No. 396, Tongfuzhong Road, Haizhu District, Guangzhou, 510220, Guangdong, China
| | - Yihui Huang
- Department of Pediatrics, Shantou University Medical College, Shantou, 515041, China
- Department of Pediatrics, Guangzhou Red Cross Hospital, Jinan University, No. 396, Tongfuzhong Road, Haizhu District, Guangzhou, 510220, Guangdong, China
| |
Collapse
|
12
|
Sun Z, Deng G, Peng X, Xu X, Liu L, Peng J, Ma Y, Zhang P, Wen A, Wang Y, Yang Z, Gong P, Jiang W, Cai L. Intelligent photothermal dendritic cells restart the cancer immunity cycle through enhanced immunogenic cell death. Biomaterials 2021; 279:121228. [PMID: 34717198 DOI: 10.1016/j.biomaterials.2021.121228] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/17/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) play a pivotal role in initiating antigen-specific tumor immunity. However, the abnormal function of DCs owing to the immunosuppressive tumor microenvironment (TME) and the insufficient number of tumor infiltrating DCs could promote immune tolerance and tumor immune escape. Thus, there is great potential to employ DCs to induce efficient antitumor immunity. In this paper, we developed intelligent DCs (iDCs), which consist of nanoparticles loaded with photothermal agents (IR-797) and coated with a mature DC membrane. The DC cell membrane on the surface of iDCs preserves the ability to present antigens and prime T cells. The iDCs can also enter the lymph node and stimulate T cells. The activated T cells reduced the expression of heat shock proteins (HSPs) in tumor cells, rendering them more sensitive to heat stress. Subsequently, we used mild photothermal therapy (42-45 °C) to induce immunogenic cell death and contribute to a synergistic antitumor effect. iDCs as a refined and precise system in combination with DC-based immunotherapy and thermal therapy can be stored long-term and on a large scale, so they can be applied in many patients.
Collapse
Affiliation(s)
- Zhihong Sun
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Qindao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, 264000, PR China
| | - Guanjun Deng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xinghua Peng
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiuli Xu
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jiaofeng Peng
- Instrumental Analysis Center of Shenzhen University, Shenzhen University, Shenzhen, 518055, China
| | - Yifan Ma
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; HRYZ Biotech Co., Shenzhen, 518057, PR China
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Austin Wen
- Pomona College, 333 N College Way, Claremont, CA, 91711, USA
| | - Yifan Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhaogang Yang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ping Gong
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations, CAS-HK Joint Lab for Biomaterials, CAS Key Laboratory of Health Informatics, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
13
|
Sano E, Kazaana A, Tadakuma H, Takei T, Yoshimura S, Hanashima Y, Ozawa Y, Yoshino A, Suzuki Y, Ueda T. Interleukin-6 sensitizes TNF-α and TRAIL/Apo2L dependent cell death through upregulation of death receptors in human cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119037. [PMID: 33839168 DOI: 10.1016/j.bbamcr.2021.119037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
Interleukin-6 (IL-6) enhanced TNF-α and TRAIL/Apo2L induced cell death in various human cancer cells derived from malignant glioma, melanoma, breast cancer and leukemia, although the effect was not detected with IL-6 alone. The effects of IL-6 using SKBR3 cells were associated with the generation of apoptotic cells as analyzed by fluorescence microscopy and flow cytometry. IL-6 activated p53 and upregulated TRAIL death receptors (DR-4 and DR-5) and stimulated the TNF-α and TRAIL dependent extrinsic apoptotic pathway without activation of the p53 mediated intrinsic apoptotic pathway. TNF-α and TRAIL induced cleavage of caspase-8 and caspase-3 was more enhanced by IL-6, although these caspases were not cleaved by IL-6 alone. The dead cell generation elicited by the combination with IL-6 was blocked by anti-human TRAIL R2/TNFRSF10B Fc chimera antibody which can neutralize the DR-5 mediated death signal. These findings indicate that IL-6 could contribute to the enhancement of TNF-α or TRAIL induced apoptosis through p53 dependent upregulation of DR-4 and DR-5. The data suggest that a favorable therapeutic interaction could occur between TNF-α or TRAIL and IL-6, and provide an experimental basis for rational clinical treatments in various cancers.
Collapse
Affiliation(s)
- Emiko Sano
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan.
| | - Akira Kazaana
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Hisashi Tadakuma
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Toshiaki Takei
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Sodai Yoshimura
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yuya Hanashima
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yoshinari Ozawa
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Atsuo Yoshino
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan
| | - Takuya Ueda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8562, Japan; Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Science and Engineering, Waseda University, Tokyo 162-8480, Japan
| |
Collapse
|
14
|
Tan L, Liu L, Yao J, Piao C. miR-145-5p attenuates inflammatory response and apoptosis in myocardial ischemia-reperfusion injury by inhibiting (NADPH) oxidase homolog 1. Exp Anim 2021; 70:311-321. [PMID: 33658472 PMCID: PMC8390312 DOI: 10.1538/expanim.20-0160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a common complication following reperfusion therapy that involves a series of immune or apoptotic reactions. Studies have revealed the potential roles of miRNAs in I/R injury. Herein, we established a myocardial I/R model in rats and a hypoxia/reoxygenation (H/R) model in H9c2 cells and investigated the effect of miR-145-5p on myocardial I/R injury. After 3 h or 24 h of reperfusion, left ventricular end-systolic pressure (LVESP), ejection fraction (EF), and fractional shortening (FS) were obviously decreased, and left ventricular end-diastolic pressure (LVEDP) was increased. Meanwhile, I/R induced an increase in myocardial infarction area. Moreover, a decrease in miR-145-5p and increase in (NADPH) oxidase homolog 1 (NOH-1) were observed following I/R injury. With this in mind, we performed a luciferase reporter assay and demonstrated that miR-145-5p directly bound to NOH-1 3' untranslated region (UTR). Furthermore, miR-145-5p mimics decreased the levels of tumor necrosis factor (TNF)-α, IL-1β, and IL-6 via oxygen and glucose deprivation/reperfusion (OGD/R) stimulation. Upregulation of miR-145-5p increased cell viability and reduced apoptosis accompanied by downregulation of Bax, cleaved caspase-3, cleaved poly(ADP-ribose) polymerase (PARP) and upregulation of Bcl2. In addition, miR-145-5p overexpression increased superoxide dismutase (SOD) activity and reduced reactive oxygen species (ROS) and malondialdehyde (MDA) content under OGD/R stress. Notably, NOH-1 could significantly abrogate the above effects, suggesting that it is involved in miR-145-5p-regulated I/R injury. In summary, our findings indicated that miR-145-5p/NOH-1 has a protective effect on myocardial I/R injury by inhibiting the inflammatory response and apoptosis.
Collapse
Affiliation(s)
- Lili Tan
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Limin Liu
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Jian Yao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Chenghao Piao
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| |
Collapse
|
15
|
Damame HH, Rooge SB, Patil RS, Arvindekar AU. In vitro model using cytokine cocktail to evaluate apoptosis in Min6 pancreatic beta cells. J Pharmacol Toxicol Methods 2020; 106:106914. [PMID: 32828949 DOI: 10.1016/j.vascn.2020.106914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 08/08/2020] [Accepted: 08/13/2020] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Development of therapy options for treatment of type 1 diabetes mellitus is hampered by non-availability of appropriate experimental models that can exactly mimic the in vivo situation. Apoptosis of beta cells by T cells and cytokine action leads to loss of beta cells. We propose a simple and elegant model using cytokine cocktail of TNF-α, IFN-γ and IL-1β, the major cytokines responsible for apoptosis in Min6 beta cell line. METHODS A cocktail of TNF-α, IFN-γ and IL-1β was used to induce apoptosis in Min6 beta cell line. Apoptosis was assessed by flow cytometry using CytoFLEX (Beckman Coulter). The destruction of beta cells is through production of nitric oxide (NO), oxidative stress and change in mitochondrial membrane permeability. NO was measured using Griess reagent. Oxidative stress was assessed using 2',7'-dichlorofluorescein diacetate, a cell-permeable fluorogenic dye and mitochondrial membrane potential was determined on the basis of retention of rhodamine 123 using flow cytometer. RESULTS AND DISCUSSION Very low concentration of the cocktail viz. TNF-α 25 ng/ml, IFN-γ 25 ng/ml and IL-1β 50 ng/ml has demonstrated effective early and late apoptosis in as short a time period as 6 h. The experimental model used demonstrated 1.5 fold higher production of NO, 1.2 fold increased oxidative stress and lower mitochondrial membrane potential as compared to the positive control used. Hence the above model can be easily used for assessment and screening of drugs that can prevent apoptosis of beta cells and stop progression of type 1 diabetes.
Collapse
Affiliation(s)
- Hemangee H Damame
- Department of Biochemistry, Shivaji University, Kolhapur 416 004, Maharashtra, India
| | - Sheetalnath B Rooge
- Department of Biochemistry, Shivaji University, Kolhapur 416 004, Maharashtra, India
| | - Rahul S Patil
- Department of Biochemistry, Shivaji University, Kolhapur 416 004, Maharashtra, India
| | - Akalpita U Arvindekar
- Department of Biochemistry, Shivaji University, Kolhapur 416 004, Maharashtra, India.
| |
Collapse
|
16
|
Hao Y, Wang W, Wu D, Liu K, Sun Y. Retracted: Bilobalide alleviates tumor necrosis factor‐alpha‐induced pancreatic beta‐cell MIN6 apoptosis and dysfunction through upregulation of miR‐153. Phytother Res 2019; 34:409-417. [PMID: 31667906 DOI: 10.1002/ptr.6533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/22/2019] [Accepted: 10/09/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Yan Hao
- Department of EndocrinologyJining No.1 People's Hospital Jining China
| | - Weiwei Wang
- Department of EndocrinologyJining No.1 People's Hospital Jining China
| | - Dong Wu
- Emergency DepartmentJining No.1 People's Hospital Jining China
| | - Kai Liu
- Emergency DepartmentJinxiang People's Hospital Jining China
| | - Yihan Sun
- Department of EndocrinologyJining No.1 People's Hospital Jining China
| |
Collapse
|
17
|
Bal T, Inceoglu Y, Karaoz E, Kizilel S. Sensitivity Study for the Key Parameters in Heterospheroid Preparation with Insulin-Secreting β-Cells and Mesenchymal Stem Cells. ACS Biomater Sci Eng 2019; 5:5229-5239. [DOI: 10.1021/acsbiomaterials.9b00570] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Tuğba Bal
- Chemical and Biological Engineering, Koc University, 34450 Sariyer, Istanbul, Turkey
| | - Yasemin Inceoglu
- Chemical and Biological Engineering, Koc University, 34450 Sariyer, Istanbul, Turkey
| | - Erdal Karaoz
- Center for Regenerative Medicine and Stem Cell Research, Liv Hospital, 34340 Besiktas, Istanbul, Turkey
- School of Medicine, Istinye University, 34010 Zeytinburnu, Istanbul, Turkey
| | - Seda Kizilel
- Chemical and Biological Engineering, Koc University, 34450 Sariyer, Istanbul, Turkey
- Biomedical Science and Engineering, Koc University, 34450 Sariyer, Istanbul, Turkey
| |
Collapse
|
18
|
Zhang YQ, Shen Y, Liao MM, Mao X, Mi GJ, You C, Guo QY, Li WJ, Wang XY, Lin N, Webster TJ. Galactosylated chitosan triptolide nanoparticles for overcoming hepatocellular carcinoma: Enhanced therapeutic efficacy, low toxicity, and validated network regulatory mechanisms. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 15:86-97. [PMID: 30244085 DOI: 10.1016/j.nano.2018.09.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/22/2018] [Accepted: 09/03/2018] [Indexed: 12/31/2022]
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide. Current therapies present significant limitations. Triptolide (TP) is highly effective against multiple cancers including HCC. However, high toxicity, low water solubility, and unknown therapeutic targets limit its clinical application. Herein, we designed galactosylated-chitosan-TP-nanoparticles (GC-TP-NPs) with high drug loading capacities for targeted delivery to HCC. In addition to a sustained release pattern, an efficient asialoglycoprotein receptor mediated cellular uptake in vitro, and high liver tumor accumulation in vivo, GC-TP-NPs showed lower systemic and male reproductive toxicities than free TP. Importantly, GC-TP-NPs retained the anti-cancer activities of the free TP, exerting the same pro-apoptotic and anti-proliferative effects on HCC cells in vitro, and displayed higher efficacies in reducing tumor sizes in vivo. Further investigation revealed that GC-TP-NPs induced cancer cell apoptosis via blocking TNF/NF-κB/BCL2 signaling. Collectively, GC-TP-NP represents a promising candidate in halting liver cancer progression while minimizing systemic toxicity.
Collapse
Affiliation(s)
- Yan-Qiong Zhang
- Institute of Chinese Materials Medical, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan Shen
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Ming-Mei Liao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xia Mao
- Institute of Chinese Materials Medical, China Academy of Chinese Medical Sciences, Beijing, China
| | - Gu-Jie Mi
- Department of Chemical Engineering, Northeastern University, Boston, USA
| | - Chen You
- Jiangsu Provincial Xuzhou Pharmaceutical Vocational College, Xuzhou, China
| | - Qiu-Yan Guo
- Institute of Chinese Materials Medical, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wei-Jie Li
- Institute of Chinese Materials Medical, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao-Yue Wang
- Institute of Chinese Materials Medical, China Academy of Chinese Medical Sciences, Beijing, China
| | - Na Lin
- Institute of Chinese Materials Medical, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, USA.
| |
Collapse
|
19
|
You W, Wang K, Yu C, Song L. Retracted
: Baicalin prevents tumor necrosis factor‐α−induced apoptosis and dysfunction of pancreatic β‐cell line Min6 via upregulation of miR‐205. J Cell Biochem 2018; 119:8547-8554. [DOI: 10.1002/jcb.27095] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/26/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Wenjun You
- Department of Endocrinology The Affiliated Hospital of Qingdao University Qingdao Shandong China
- Department of Endocrinology Jining No.1 People’s Hospital Jining Shandong China
| | - Kun Wang
- Department of Endocrinology Jining No.1 People’s Hospital Jining Shandong China
| | - Changhong Yu
- First Department of Gastroenterology First Affiliated Hospital of Jiamusi University Jiamusi Heilongjiang China
| | - Lijuan Song
- Department of Endocrinology Jining No.1 People’s Hospital Jining Shandong China
| |
Collapse
|
20
|
Li D, Zhang P. Protective effect and molecular mechanism of liquiritin on oxybuprocaine-induced apoptosis of human corneal endothelial cells. Exp Ther Med 2018; 15:3432-3438. [PMID: 29545865 PMCID: PMC5841024 DOI: 10.3892/etm.2018.5860] [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] [Received: 08/25/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022] Open
Abstract
This study was designed to investigate the protective effect and possible molecular mechanism of liquiritin on oxybuprocaine-induced apoptosis of human corneal endothelial cells (HCECs). In this study, the effect of oxybuprocaine on the proliferation of HCEC-12 was detected using cell counting kit-8 (CCK-8). The inductive effect of oxybuprocaine on HCEC-12 apoptosis and protective effect of liquiritin against oxybuprocaine-induced HCEC-12 apoptosis were tested by Annexin V/propidium iodide (PI) staining and flow cytometry. The production of reactive oxygen species (ROS) was analyzed by 2,7-dichlorodi-hydrofluorescein diacetate (DCFH-DA) staining and fluorescent-activated cell sorting (FACS), and the expression of nuclear factor-κB (NF-κB) p65 and apoptosis-related proteins, caspase-3 and Bax, was determined by western blot analysis. Our results show that liquiritin resisted the inhibitory effect of oxybuprocaine on the proliferation of HCEC-12, and cell activity had the most significant increase in pretreatment with liquiritin group in the concentration of 8 mg/ml; compared with that in oxybuprocaine group. Apoptosis in pretreatment with liquiritin was distinctly decreased and liquiritin resisted the production of ROS in HCEC-12 induced by oxybuprocaine. Investigation of molecular mechanism revealed that the pretreatment with liquiritin and pyrrolidinedithiocarbamic acid (PDTC) obviously blocked the expression of NF-κB p65 in nuclear protein increased by oxybuprocaine and the expression levels of total proteins, caspase-3 and Bax.Moreover, tumor necrosis factor-α (TNF-α) blocked the inhibitory effect of liquiritin on the expression of NF-κB p65 in nuclear protein and total proteins, caspase-3 and Bax, thus obstructing the protective effect of liquiritin on corneal epithelial cells. The results of this study indicated that liquiritin reduces the expression of apoptosis protein and increases the expression of anti-apoptotic protein through inhibiting NF-κB signal pathway, thus resisting HCEC-12 apoptosis induced by oxybuprocaine.
Collapse
Affiliation(s)
- Dan Li
- Department of Anesthesiology, Ezhou Central Hospital of Wuhan University, Ezhou, Hubei 436000, P.R. China
| | - Peng Zhang
- Department of Ophthalmology, Ezhou Central Hospital of Wuhan University, Ezhou, Hubei 436000, P.R. China
| |
Collapse
|
21
|
Yao Q, Wang W, Jin J, Min K, Yang J, Zhong Y, Xu C, Deng J, Zhou Y. Synergistic role of Caspase-8 and Caspase-3 expressions: Prognostic and predictive biomarkers in colorectal cancer. Cancer Biomark 2018; 21:899-908. [PMID: 29355114 DOI: 10.3233/cbm-170967] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Qiang Yao
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Soochow, Jiangsu, China
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Weimin Wang
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
- Institute of Combining Chinese Traditional and Western Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Jun Jin
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Ke Min
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Jian Yang
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Yubing Zhong
- Department of General Surgery, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Chunni Xu
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Jianliang Deng
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
| | - Yan Zhou
- Department of Oncology, Yixing Hospital Affiliated to Medical College of Yangzhou University, Yixing, Jiangsu, China
- Institute of Combining Chinese Traditional and Western Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
22
|
Park JW, Lee HS, Lim Y, Paik JH, Kwon OK, Kim JH, Paryanto I, Yunianto P, Choi S, Oh SR, Ahn KS. Rhododendron album Blume extract inhibits TNF-α/IFN-γ-induced chemokine production via blockade of NF-κB and JAK/STAT activation in human epidermal keratinocytes. Int J Mol Med 2018. [PMID: 29532855 DOI: 10.3892/ijmm.2018.3556] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Rhododendron album Blume (RA) has traditionally been used as an herbal medicine and is considered to have anti‑inflammatory properties. It is a well‑known medicine for treatment of allergic or atopic diseases. In the present study, the biological effects of an RA methanol extract (RAME) on inflammation were investigated in tumor necrosis factor‑α (TNF‑α)/interferon‑γ (IFN‑γ)‑stimulated human keratinocytes. The present study aimed to investigate the potential mechanisms by which RAME inhibited TNF‑α/IFN‑γ‑induced expression of chemokines [thymus‑ and activation-regulated chemokine (TARC) and macrophage‑derived chemokine (MDC)] and cytokines [interleukin (IL)‑6 and IL‑8] through the nuclear factor‑κB (NF‑κB) pathway in human keratinocytes. The effects of RAME treatment on cell viability were investigated in TNF‑α/IFN‑γ‑stimulated HaCaT cells. The expression of TARC, MDC, IL‑6 and IL‑8 was assessed using reverse transcription‑quantitative polymerase chain reaction analysis or ELISA, and its effect on the inhibitory mitogen-activated protein kinase pathway was also studied using western blot analysis. TNF‑α/IFN‑γ induced the expression of IL‑6, IL‑8, TARC and MDC in a dose‑dependent manner through NF‑κB and Janus kinase/signal transducers and activators of transcription (JAK/STAT) activation. Notably, treatment with RAME significantly suppressed TNF-α/IFN-γ-induced expression of IL‑6, IL‑8, TARC, and MDC. In addition, RAME treatment inhibited the activation of NF‑κB and the JAK/STAT pathway in TNF‑α/IFN‑γ‑induced HaCaT cells. These results suggest that RAME decreases the production of chemokines and pro‑inflammatory cytokines by suppressing the NF‑κB and the JAK/STAT pathways. Consequently, RAME may potentially be used for treatment of atopic dermatitis.
Collapse
Affiliation(s)
- Ji-Won Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Han-Sol Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Yourim Lim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Jin-Hyub Paik
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Ok-Kyoung Kwon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Jung-Hee Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Imam Paryanto
- Center for Pharmaceutical and Medical Technology, Kawasan Puspiptek Serpong, LAPTIAB, Tangerang, Banten 15314, Indonesia
| | - Prasetyawan Yunianto
- Center for Pharmaceutical and Medical Technology, Kawasan Puspiptek Serpong, LAPTIAB, Tangerang, Banten 15314, Indonesia
| | - Sangho Choi
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Chungbuk 28116, Republic of Korea
| |
Collapse
|
23
|
Role of pterostilbene in attenuating immune mediated devastation of pancreatic beta cells via Nrf2 signaling cascade. J Nutr Biochem 2017; 44:11-21. [DOI: 10.1016/j.jnutbio.2017.02.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/01/2017] [Accepted: 02/16/2017] [Indexed: 01/12/2023]
|
24
|
Zheng QY, Liang SJ, Li GQ, Lv YB, Li Y, Tang M, Zhang K, Xu GL, Zhang KQ. Complement component 3 deficiency prolongs MHC-II disparate skin allograft survival by increasing the CD4(+) CD25(+) regulatory T cells population. Sci Rep 2016; 6:33489. [PMID: 27641978 PMCID: PMC5027598 DOI: 10.1038/srep33489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022] Open
Abstract
Recent reports suggest that complement system contributes to allograft rejection. However, its underlying mechanism is poorly understood. Herein, we investigate the role of complement component 3 (C3) in a single MHC-II molecule mismatched murine model of allograft rejection using C3 deficient mice (C3−/−) as skin graft donors or recipients. Compared with C3+/+ B6 allografts, C3−/− B6 grafts dramatically prolonged survival in MHC-II molecule mismatched H-2bm12 B6 recipients, indicating that C3 plays a critical role in allograft rejection. Compared with C3+/+ allografts, both Th17 cell infiltration and Th1/Th17 associated cytokine mRNA levels were clearly reduced in C3−/− allografts. Moreover, C3−/− allografts caused attenuated Th1/Th17 responses, but increased CD4+CD25+Foxp3+ regulatory T (Treg) cell expression markedly in local intragraft and H-2bm12 recipients. Depletion of Treg cells by anti-CD25 monoclonal antibody (mAb) negated the survival advantages conferred by C3 deficiency. Our results indicate for the first time that C3 deficiency can prolong MHC-II molecule mismatched skin allograft survival, which is further confirmed to be associated with increased CD4+ CD25+ Treg cell population expansion and attenuated Th1/Th17 response.
Collapse
Affiliation(s)
- Quan-You Zheng
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.,Department of Urology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Shen-Ju Liang
- Department of Rheumatism and Immunology, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Gui-Qing Li
- Department of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Yan-Bo Lv
- Department of Immunology, Third Military Medical University, Chongqing 400038, China
| | - You Li
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ming Tang
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Kun Zhang
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Gui-Lian Xu
- Department of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Ke-Qin Zhang
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
25
|
Baicalin attenuates lipopolysaccharide induced inflammation and apoptosis of cow mammary epithelial cells by regulating NF-κB and HSP72. Int Immunopharmacol 2016; 40:139-145. [PMID: 27588914 DOI: 10.1016/j.intimp.2016.08.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 01/06/2023]
Abstract
Baicalin is the main ingredient of traditional Chinese herbal medicine, Scutellaria baicalensis, which has been widely used clinically as an anti-inflammatory agent. However, molecular mechanism of action of this drug is not yet clear. In the present study, the protective mechanism of baicalin against lipopolysaccharide (LPS) induced inflammatory injury in cow mammary epithelial cells (CMECs) was explored. For this purpose, in vitro cultured CMECs were treated with baicalin (10μg/mL) and LPS (10μg/mL) for 24 and 12h, respectively, and the cell viability was measured by using cell counting kit-8 (CCK-8). The results revealed that LPS induced inflammatory responses, as p-p65/p65 and p-IκBα/IκBα ratios and TNF-α and IL-1β production was increased in the CMECs. Both Bcl-2/Bax ratio and cell viability were decreased and caspase-3 cleaved following LPS treatment, indicating apoptosis of CMECs. Moreover, both LPS and baicalin increased HSP72 expression of the CMECs. However, cellular inflammatory responses and apoptosis were significantly reduced in baicalin treated CMECs. In conclusion, baicalin ameliorated inflammation and apoptosis of the CMECs induced by LPS via inhibiting NF-κB activation and up regulation of HSP72.
Collapse
|
26
|
Zheng QY, Cao ZH, Hu XB, Li GQ, Dong SF, Xu GL, Zhang KQ. LIGHT/IFN-γ triggers β cells apoptosis via NF-κB/Bcl2-dependent mitochondrial pathway. J Cell Mol Med 2016; 20:1861-71. [PMID: 27241100 PMCID: PMC5020636 DOI: 10.1111/jcmm.12876] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/08/2016] [Indexed: 01/14/2023] Open
Abstract
LIGHT recruits and activates naive T cells in the islets at the onset of diabetes. IFN‐γ secreted by activated T lymphocytes is involved in beta cell apoptosis. However, whether LIGHT sensitizes IFNγ‐induced beta cells destruction remains unclear. In this study, we used the murine beta cell line MIN6 and primary islet cells as models for investigating the underlying cellular mechanisms involved in LIGHT/IFNγ – induced pancreatic beta cell destruction. LIGHT and IFN‐γ synergistically reduced MIN6 and primary islet cells viability; decreased cell viability was due to apoptosis, as demonstrated by a significant increase in Annexin V+ cell percentage, detected by flow cytometry. In addition to marked increases in cytochrome c release and NF‐κB activation, the combination of LIGHT and IFN‐γ caused an obvious decrease in expression of the anti‐apoptotic proteins Bcl‐2 and Bcl‐xL, but an increase in expression of the pro‐apoptotic proteins Bak and Bax in MIN6 cells. Accordingly, LIGHT deficiency led to a decrease in NF‐κB activation and Bak expression, and peri‐insulitis in non‐obese diabetes mice. Inhibition of NF‐κB activation with the specific NF‐κB inhibitor, PDTC (pyrrolidine dithiocarbamate), reversed Bcl‐xL down‐regulation and Bax up‐regulation, and led to a significant increase in LIGHT‐ and IFN‐γ‐treated cell viability. Moreover, cleaved caspase‐9, ‐3, and PARP (poly (ADP‐ribose) polymerase) were observed after LIGHT and IFN‐γ treatment. Pretreatment with caspase inhibitors remarkably attenuated LIGHT‐ and IFNγ‐induced cell apoptosis. Taken together, our results indicate that LIGHT signalling pathway combined with IFN‐γ induces beta cells apoptosis via an NF‐κB/Bcl2‐dependent mitochondrial pathway.
Collapse
Affiliation(s)
- Quan-You Zheng
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhao-Hui Cao
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacy and Biosciences, University of South China, Hengyang, China.,Department of Immunology, School of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Xiao-Bo Hu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmacy and Biosciences, University of South China, Hengyang, China
| | - Gui-Qing Li
- Department of Immunology, School of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Shi-Fang Dong
- Department of Immunology, School of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Gui-Lian Xu
- Department of Immunology, School of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Ke-Qin Zhang
- Department of Nephrology, Southwest Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
27
|
Lee SJ, Kang HK, Song DK, Eum WS, Park J, Choi SY, Kwon HY. Transduction of PEP-1-heme oxygenase-1 into insulin-producing INS-1 cells protects them against cytokine-induced cell death. Biochem Biophys Res Commun 2015; 461:549-54. [DOI: 10.1016/j.bbrc.2015.04.076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 04/07/2015] [Indexed: 11/27/2022]
|
28
|
Cao ZH, Zheng QY, Li GQ, Hu XB, Feng SL, Xu GL, Zhang KQ. STAT1-mediated down-regulation of Bcl-2 expression is involved in IFN-γ/TNF-α-induced apoptosis in NIT-1 cells. PLoS One 2015; 10:e0120921. [PMID: 25811609 PMCID: PMC4374929 DOI: 10.1371/journal.pone.0120921] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 02/09/2015] [Indexed: 01/30/2023] Open
Abstract
Tumor necrosis factor (TNF)-α and interferon (IFN)-γ are the major pro-inflammatory cytokines involved in beta-cell destruction. The fate of islet beta-cells in the cytokine-induced intrinsic mitochondrial apoptotic pathway is determined by the interaction between members of the Bcl-2 family. However, the mechanism through which beta-cell apoptosis is regulated remains unclear. In this study, we treated the murine beta-cell line NIT-1 with TNF-α and IFN-γ and then investigated the regulation of signal transducer and activator of transcription-1 (STAT-1) and expression of the members of the Bcl-2 family in this apoptotic pathway. Results showed that TNF-α and IFN-γ synergistically reduced NIT-1 cell viability. In addition, the decrease in cell growth was due to apoptosis as shown by apoptotic body formation, detected by confocal laser microscope, and a significant increase in Annexin-Vup+ cell percentage, detected by flow cytometry. Combination treatment with TNF-α and IFN-γ caused a remarkable increase in the release of cytochrome c, and in the activation of caspase-9 and caspase-3, as well as, an obvious enhancement in STAT-1 phosphorylation; the treatment, however, resulted in the down-regulation in Bcl-2 expression. The enhancement in STAT-1 activity and a down-regulation in Bcl-2 expression was also observed in MIN6 cells, another murine beta-cell derived line, after cells exposure to the combination of TNF-α and IFN-γ treatment. Knockdown of STAT-1 gene expression by siRNA or inhibition of STAT-1 activation with fludarabine reversed Bcl-2 down-expression and led to a significant decrease in apoptosis in TNF-α- and IFN-γ-treated NIT-1 cells. Taken together, our results suggest that STAT1-mediated down-regulation of Bcl-2 is involved in NIT-1 cell apoptosis induced by combination treatment with TNF-α and IFN-γ.
Collapse
Affiliation(s)
- Zhao-hui Cao
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Department of Biochemistry and Molecular Biology, School of Pharmacy and Biological Sciences, School of Public Health, University of South China, Hengyang 421001, China
| | - Quan-you Zheng
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Gui-qing Li
- Department of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Xiao-bo Hu
- Department of Biochemistry and Molecular Biology, School of Pharmacy and Biological Sciences, School of Public Health, University of South China, Hengyang 421001, China
| | - Shao-long Feng
- Department of Health Laboratory Technology, School of Public Health, University of South China, Hengyang 421001, China
| | - Gui-lian Xu
- Department of Immunology, Third Military Medical University, Chongqing 400038, China
- * E-mail: (GLX); (KQZ)
| | - Ke-qin Zhang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- * E-mail: (GLX); (KQZ)
| |
Collapse
|