1
|
Chen J, Fei S, Chan LWC, Gan X, Shao B, Jiang H, Li S, Kuang P, Liu X, Yang S. Inflammatory signaling pathways in pancreatic β-cell: New insights into type 2 diabetes pathogenesis. Pharmacol Res 2025; 216:107776. [PMID: 40378943 DOI: 10.1016/j.phrs.2025.107776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/27/2025] [Accepted: 05/10/2025] [Indexed: 05/19/2025]
Abstract
Type 2 diabetes (T2D) is a complex metabolic disorder with a growing global prevalence, and there is a linking between inflammation in pancreatic β-cell and impaired glucose homeostasis which has emerged as a key player in the pathogenesis of T2D. Recent advances in research have provided new insights into various inflammatory signaling cascades in β-cell among which we focus on Toll-like Receptor 4 (TLR4), Nuclear Factor kappa B (NF-κB), Janus Kinase-Signal Transducer and Activator of Transcription (JAK/STAT), Platelet-Derived Growth Factor Receptor α (PDGFR-α), Stimulator of Interferon Genes (STING), and the death receptor TMEM219. TLR4 activation by pathogen- or damage-associated molecular patterns initiates NF-κB and mitogen-activated protein kinase (MAPK) cascades, promoting pro-inflammatory cytokine release and β-cell apoptosis. NF-κB acts as a central hub, integrating metabolic stress signals (e.g., glucolipotoxicity, ER stress) and amplifying inflammatory responses through crosstalk with JAK/STAT and STING pathways. Meanwhile, JAK/STAT signaling exhibits dual roles in β-cell survival and inflammation, influenced by cytokine milieu and feedback regulation. PDGFR-α, traditionally linked to β-cell proliferation, paradoxically contributes to pathological hyperplasia in obesity, while STING activation by cytosolic DNA triggers β-cell senescence and ferroptosis via IRF3/NF-κB. In this review, we synthesize recent advancements of these inflammatory signaling pathways in β-cells, and current therapeutic strategies targeting TLR4/NF-κB inhibitors, JAK/STAT modulators, STING antagonists, and the death receptor TMEM219 are discussed, alongside challenges in pathway specificity and clinical translation. Understanding these inflammatory signaling pathways and their interactions in pancreatic β-cell is essential for the development of novel therapeutic strategies to prevent or treat T2D.
Collapse
Affiliation(s)
- Jie Chen
- Department of Respiratory Medicine, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Shinuan Fei
- Pediatrics Department, Huangshi Maternal and Child Health Care Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435003, PR China
| | - Lawrence W C Chan
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, 99077, Hong Kong
| | - Xueting Gan
- Department of Pathology, Huangshi maternal and Child Health Care Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435003, PR China
| | - Bibo Shao
- Department of Intensive Care Unit, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Hong Jiang
- Department of Laboratory Medicine, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Sheng Li
- Department of Laboratory Medicine, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Peng Kuang
- Huangshi Maternal and Child Health Hospital Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Xin Liu
- Department of Ultrasound Medicine Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China
| | - Sijun Yang
- Department of Laboratory Medicine, Huangshi Maternity and Children's Health Hospital, Affiliated Maternity and Children's Health Hospital of Hubei Polytechnic University, Huangshi Key Laboratory of Birth Defects Prevention, Huangshi, Hubei 435000, PR China.
| |
Collapse
|
2
|
Gui R, Ren Y, Wang Z, Li Y, Wu C, Li X, Li M, Li Y, Qian L, Xiong Y. Deciphering interleukin-18 in diabetes and its complications: Biological features, mechanisms, and therapeutic perspectives. Obes Rev 2024; 25:e13818. [PMID: 39191434 DOI: 10.1111/obr.13818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/16/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024]
Abstract
Interleukin-18 (IL-18), a potent and multifunctional pro-inflammatory cytokine, plays a critical role in regulating β-cell failure, β-cell death, insulin resistance, and various complications of diabetes mellitus (DM). It exerts its effects by triggering various signaling pathways, enhancing the production of pro-inflammatory cytokines and nitric oxide (NO), as well as promoting immune cells infiltration and β-cells death. Abnormal alterations in IL-18 levels have been revealed to be strongly associated with the onset and development of DM and its complications. Targeting IL-18 may present a novel and promising approach for DM therapy. An increasing number of IL-18 inhibitors, including chemical and natural inhibitors, have been developed and have been shown to protect against DM and diabetic complications. This review provides a comprehensive understanding of the production, biological functions, action mode, and activated signaling pathways of IL-18. Next, we shed light on how IL-18 contributes to the pathogenesis of DM and its associated complications with links to its roles in the modulation of β-cell failure and death, insulin resistance in various tissues, and pancreatitis. Furthermore, the therapeutic potential of targeting IL-18 for the diagnosis and treatment of DM is also highlighted. We hope that this review will help us better understand the functions of IL-18 in the pathogenesis of DM and its complications, providing novel strategies for DM diagnosis and treatment.
Collapse
Affiliation(s)
- Runlin Gui
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Zhen Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Chengsong Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Xiaofang Li
- Department of Gastroenterology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Man Li
- Department of Endocrinology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Yujia Li
- Department of Traditional Chinese Medicine, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Lu Qian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
- Scientific Research Center, Xi'an Mental Health Center, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
3
|
Ming Z, Lim SY, Stewart A, Pedersen B, Shklovskaya E, Menzies AM, Carlino MS, Kefford RF, Lee JH, Scolyer RA, Long GV, Rizos H. IFN-γ Signaling Sensitizes Melanoma Cells to BH3 Mimetics. J Invest Dermatol 2023; 143:1246-1256.e8. [PMID: 36736995 DOI: 10.1016/j.jid.2023.01.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/22/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023]
Abstract
Immunotherapy targeting PD-1 and/or CTLA4 leads to durable responses in a proportion of patients with melanoma. However, many patients will not respond to these immune checkpoint inhibitors, and up to 60% of responding patients will develop treatment resistance. We describe a vulnerability in melanoma driven by immune cell activity that provides a pathway towards additional treatment options. This study evaluated short-term melanoma cell lines (referred to as PD1 PROG cells) derived from melanoma metastases that progressed on PD-1 inhibitor-based therapy. We show that the cytokine IFN-γ primes melanoma cells for apoptosis by promoting changes in the accumulation and interactions of apoptotic regulators MCL-1, NOXA, and BAK. The addition of pro-apoptotic BH3 mimetic drugs sensitized PD1 PROG melanoma cells to apoptosis in response to IFN-γ or autologous immune cell activation. These findings provide translatable strategies for combination therapies in melanoma.
Collapse
Affiliation(s)
- Zizhen Ming
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Su Yin Lim
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Ashleigh Stewart
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Bernadette Pedersen
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Elena Shklovskaya
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Department of Medical Oncology, Mater Hospital, Sydney, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, Australia; Department of Medical Oncology, Blacktown Cancer and Haematology Centre, Blacktown Hospital, Sydney, Australia
| | - Richard F Kefford
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Jenny H Lee
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Chris O'Brien Lifehouse, Camperdown, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Department of Medical Oncology, Mater Hospital, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Helen Rizos
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, Australia.
| |
Collapse
|
4
|
Hsu CY, Huang CY, Shih CM, Lin YW, Huang PH, Lin SJ, Liu CW, Lin CY, Lin FY. Tumor Necrosis Factor Superfamily 14 (LIGHT) Restricts Neovascularization by Decreasing Circulating Endothelial Progenitor Cells and Function. Int J Mol Sci 2023; 24:ijms24086997. [PMID: 37108160 PMCID: PMC10138919 DOI: 10.3390/ijms24086997] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/29/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Tumor necrosis factor superfamily 14 (TNFSF14) is also known as the LT-related inducible ligand (LIGHT). It can bind to the herpesvirus invasion mediator and lymphotoxin-β receptor to perform its biological activity. LIGHT has multiple physiological functions, including strengthening the synthesis of nitric oxide, reactive oxygen species, and cytokines. LIGHT also stimulates angiogenesis in tumors and induces the synthesis of high endothelial venules; degrades the extracellular matrix in thoracic aortic dissection, and induces the expression of interleukin-8, cyclooxygenase-2, and cell adhesion molecules in endothelial cells. While LIGHT induces tissue inflammation, its effects on angiogenesis after tissue ischemia are unclear. Thus, we analyzed these effects in the current study. In this study, the animal model of hind limb ischemia surgery in C57BL/6 mice was performed. Doppler ultrasound, immunohistochemical staining, and Western blotting were employed to analyze the situation of angiogenesis. In addition, human endothelial progenitor cells (EPCs) were used for in vitro studies to analyze the possible mechanisms. The results in the animal study showed that LIGHT injection inhibited angiogenesis in ischemic limbs. For the in vitro studies, LIGHT inhibited the expression of integrins and E-selectin; decreased migration and tube formation capabilities, mitochondrial respiration, and succinate dehydrogenase activity; and promoted senescence in EPCs. Western blotting revealed that the impairment of EPC function by LIGHT may be due to its effects on the proper functioning of the intracellular Akt signaling pathway, endothelial nitrite oxide synthase (eNOS), and mitochondrial respiration. In conclusion, LIGHT inhibits angiogenesis after tissue ischemia. This may be related to the clamped EPC function.
Collapse
Affiliation(s)
- Chien-Yi Hsu
- Taipei Heart Institute and Division of Cardiology, Department of Internal Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Chun-Yao Huang
- Taipei Heart Institute and Division of Cardiology, Department of Internal Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City 320, Taiwan
| | - Chun-Ming Shih
- Taipei Heart Institute and Division of Cardiology, Department of Internal Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Yi-Wen Lin
- Institute of Oral Biology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Shing-Jong Lin
- Taipei Heart Institute and Division of Cardiology, Department of Internal Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Chen-Wei Liu
- Department of Basic Medical Science, College of Medicine, University of Arizona, Phoenix, AZ 85721, USA
| | - Cheng-Yen Lin
- Healthcare Information and Management Department, Ming Chuan University, Taoyuan 333, Taiwan
| | - Feng-Yen Lin
- Taipei Heart Institute and Division of Cardiology, Department of Internal Medicine, Taipei Medical University, Taipei 110, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan
| |
Collapse
|
5
|
Zheng QY, Li Y, Liang SJ, Chen XM, Tang M, Rao ZS, Li GQ, Feng JL, Zhong Y, Chen J, Xu GL, Zhang KQ. LIGHT deficiency attenuates acute kidney disease development in an in vivo experimental renal ischemia and reperfusion injury model. Cell Death Dis 2022; 8:399. [PMID: 36163116 PMCID: PMC9512920 DOI: 10.1038/s41420-022-01188-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022]
Abstract
Ischemia-reperfusion (I/R), a leading risk factor of acute kidney injury (AKI), is associated with high mortality and risk of progression to chronic kidney disease. However, the molecular mechanism of I/R-AKI remains not fully understood, which hinders its efficient clinical treatment. In this study, we observed that LIGHT deficiency remarkably attenuated I/R-AKI, as evidenced by rescued renal function, ameliorated tubular cell apoptosis, and alleviated inflammatory responses. Consistently, blocking LIGHT signaling with its soluble receptor fusion proteins (HVEM-IgG-Fc or LTβR-IgG-Fc) improved I/R renal dysfunction. RNA-sequencing and corresponding results indicated that LIGHT promoted oxidative stress and inflammation triggered by ischemic injury. Moreover, LIGHT signaling augmented ischemic stress-induced mitochondrial dysfunction characterized by an imbalance in mitochondrial fission and fusion, decreased mtDNA copies, impaired mitophagy, and increased mitochondrial membrane potential (ΔΨm). Mechanistically, LIGHT promoted mitochondrial fission by enhancing Drp1 phosphorylation (Ser616) and its translocation to the mitochondria. In conclusion, these results suggest that LIGHT-HVEM/LTβR signaling is critical for the I/R-AKI pathogenesis and it is further confirmed to be related to the increase in I/R-induced oxidative stress and mitochondria dysfunction, which may be the underlying mechanism of LIGHT signaling-mediated I/R-AKI.
Collapse
Affiliation(s)
- Quan-You Zheng
- Department of Urology, The 958th Hospital, The First Affiliated Hospital, Army Medical University, Chongqing, 400020, China.,Department of Immunology, Army Medical University, Chongqing, 400038, China
| | - You Li
- Department of Immunology, Army Medical University, Chongqing, 400038, China.,Department of Nephrology, The First Affiliated Hospital, Army Medical University, Chongqing, 400038, China.,Department of ICU, The Third Affiliated Hospital, Army Medical University, Chongqing, 400042, China
| | - Shen-Ju Liang
- Department of Rheumatism and Immunology, The Third Affiliated Hospital, Army Medical University, Chongqing, 400042, China
| | - Xi-Ming Chen
- Urinary Nephropathy Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400065, China
| | - Ming Tang
- Urinary Nephropathy Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400065, China
| | - Zheng-Sheng Rao
- Urinary Nephropathy Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400065, China
| | - Gui-Qing Li
- Department of Immunology, Army Medical University, Chongqing, 400038, China
| | - Jian-Li Feng
- Department of Urology, The 958th Hospital, The First Affiliated Hospital, Army Medical University, Chongqing, 400020, China
| | - Yu Zhong
- Department of Urology, The 958th Hospital, The First Affiliated Hospital, Army Medical University, Chongqing, 400020, China.,Department of Immunology, Army Medical University, Chongqing, 400038, China
| | - Jian Chen
- Department of Immunology, Army Medical University, Chongqing, 400038, China
| | - Gui-Lian Xu
- Department of Immunology, Army Medical University, Chongqing, 400038, China.
| | - Ke-Qin Zhang
- Department of Nephrology, The First Affiliated Hospital, Army Medical University, Chongqing, 400038, China. .,Urinary Nephropathy Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400065, China.
| |
Collapse
|
6
|
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
|
7
|
Novel therapeutic strategies and perspectives for pancreatic cancer: Autophagy and apoptosis are key mechanisms to fight pancreatic cancer. Med Oncol 2021; 38:74. [PMID: 34019188 DOI: 10.1007/s12032-021-01522-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/10/2021] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer (PC) is the most lethal malignancy of the gastrointestinal tract. The poor prognosis of patients with PC is primarily due to lack of effective treatments against its progressive and metastatic behavior. Hence, figuring out the mechanisms underlying PC development and putting up with effective targeted therapies are of great significance to improve the prognosis of patients with PC. Apoptosis and autophagy serve to maintain tissue homoeostasis. Escaping from apoptosis or autophagy is one of the features of malignancy. PC is seriously resistant to autophagy and apoptosis, which explains its invasiveness and resistance to conventional treatment. Recently, several biological activities and pharmacological functions found in natural product extracts have been reported to inhibit PC progression. The current review focuses on understanding natural product extracts and their derivatives as one kind of novel treatments through affecting the apoptosis or autophagy in PC.
Collapse
|
8
|
Good Cop, Bad Cop: The Opposing Effects of Macrophage Activation State on Maintaining or Damaging Functional β-Cell Mass. Metabolites 2020; 10:metabo10120485. [PMID: 33256225 PMCID: PMC7761161 DOI: 10.3390/metabo10120485] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
Loss of functional β-cell mass is a hallmark of Type 1 and Type 2 Diabetes. Macrophages play an integral role in the maintenance or destruction of pancreatic β-cells. The effect of the macrophage β-cell interaction is dependent on the activation state of the macrophage. Macrophages can be activated across a spectrum, from pro-inflammatory to anti-inflammatory and tissue remodeling. The factors secreted by these differentially activated macrophages and their effect on β-cells define the effect on functional β-cell mass. In this review, the spectrum of macrophage activation is discussed, as are the positive and negative effects on β-cell survival, expansion, and function as well as the defined factors released from macrophages that impinge on functional β-cell mass.
Collapse
|
9
|
Yang Y, Meng L, Wu S, Li Y, Zhong Y, Xu F, Zhou XC, Li GQ, Xu GL, Peng KF. LIGHT deficiency aggravates cisplatin-induced acute kidney injury by upregulating mitochondrial apoptosis. Int Immunopharmacol 2020; 89:106999. [PMID: 33045563 DOI: 10.1016/j.intimp.2020.106999] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022]
Abstract
Cisplatin is widely used as a chemotherapeutic agent for treating patients with solid tumors. The most common side effect of cisplatin treatment is nephrotoxicity. Recent studies have shown that mitochondrial apoptotic pathways are involved in cisplatin-induced acute kidney injury (Cis-AKI). LIGHT, the 14th member of the tumor necrosis factor superfamily (TNFSF14), was found to induce apoptosis of certain types of tumor cells. So far, a link between LIGHT and Cis-AKI has not been reported. In this study, we observed that expression of LIGHT and its receptors HVEM and LTβR was increased in kidney tissues of mice after cisplatin treatment. LIGHT deficiency aggravated kidney injury, as evidenced by more severe tubular injury; remarkably increased levels of serum creatinine (Scr), blood urea nitrogen (BUN), and both kidney injury molecule-1 (KIM-1) and inflammatory cytokine mRNAs in renal tissues. Moreover, in the renal tissues of LIGHT KO mice, cisplatin-induced mitochondrion injury and the levels of the pro-apoptotic molecules Bax, Cytochrome C (Cyt C), cleaved caspase-3, and cleaved caspase-9 were dramatically increased; in contrast, the expression of anti-apoptotic molecule Bcl-2 was markedly reduced, compared to those in WT mice, suggesting that LIGHT deficiency accelerated cisplatin-induced mitochondrial apoptosis of renal tubular cells in these mice. Accordingly, treatment with recombinant human LIGHT (rLIGHT) was shown to alleviate cisplatin-induced kidney injury in vivo. Similar results were observed after the human renal tubular epithelial cell line HK-2 cells exposure to rLIGHT stimulation, evidenced by the reduction in the mitochondrion dysfunction (as confirmed by the significant reduced oxidative stress and membrane potential changes) and in the percentage of cells apoptosis. While blocking LIGHT with the soluble fusion protein LTβR-Ig or HVEM-Ig accelerated the HK-2 cells apoptosis. In conclusion, LIGHT deficiency aggravates Cis-AKI by promoting mitochondrial apoptosis pathways.
Collapse
Affiliation(s)
- Yan Yang
- Department of Nephrology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China; Department of Immunology, Army Medical University (Third Military Medical University), Chongqing 400038, China; Department of Intensive Care Medicine, Third Affiliated Hospital (Daping Hospital), Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Li Meng
- Department of Nephrology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shun Wu
- Department of Nephrology, Huaihai Hospital affiliated with Xuzhou Medical University, Xuzhou 221004, China
| | - You Li
- Department of Intensive Care Medicine, Third Affiliated Hospital (Daping Hospital), Army Medical University (Third Military Medical University), Chongqing 400042, China
| | - Yu Zhong
- Department of Nephrology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Feng Xu
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiao-Cui Zhou
- Department of Intensive Care Medicine, First Affiliated Hospital, Chongqing Medical and Pharmaceutical College, Chongqing 400006, China
| | - Gui-Qing Li
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Gui-Lian Xu
- Department of Immunology, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Kan-Fu Peng
- Department of Nephrology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
10
|
Zhong Y, Wu S, Yang Y, Li GQ, Meng L, Zheng QY, Li Y, Xu GL, Zhang KQ, Peng KF. LIGHT aggravates sepsis-associated acute kidney injury via TLR4-MyD88-NF-κB pathway. J Cell Mol Med 2020; 24:11936-11948. [PMID: 32881263 PMCID: PMC7579683 DOI: 10.1111/jcmm.15815] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 06/30/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Sepsis‐associated acute kidney injury (SA‐AKI) is a common clinical critical care syndrome. It has received increasing attention due to its high morbidity and mortality; however, its pathophysiological mechanisms remain elusive. LIGHT, the 14th member of the tumour necrosis factor (TNF) superfamily and a bidirectional immunoregulatory molecule that regulates inflammation, plays a pivotal role in disease pathogenesis. In this study, mice with an intraperitoneal injection of LPS and HK‐2 cells challenged with LPS were employed as a model of SA‐AKI in vivo and in vitro, respectively. LIGHT deficiency notably attenuated kidney injury in pathological damage and renal function and markedly mitigated the inflammatory reaction by decreasing inflammatory mediator production and inflammatory cell infiltration in vivo. The TLR4‐Myd88‐NF‐κB signalling pathway in the kidney of LIGHT knockout mice was dramatically down‐regulated compared to the controls. Recombinant human LIGHT aggravated LPS‐treated HK‐2 cell injury by up‐regulating the expression of the TLR4‐Myd88‐NF‐κB signalling pathway and inflammation levels. TAK 242 (a selective TLR4 inhibitor) reduced this trend to some extent. In addition, blocking LIGHT with soluble receptor fusion proteins HVEM‐Fc or LTβR‐Fc in mice attenuated renal dysfunction and pathological damage in SA‐AKI. Our findings indicate that LIGHT aggravates inflammation and promotes kidney damage in LPS‐induced SA‐AKI via the TLR4‐Myd88‐NF‐κB signalling pathway, which provide potential strategies for the treatment of SA‐AKI.
Collapse
Affiliation(s)
- Yu Zhong
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Shun Wu
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yan Yang
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Gui-Qing Li
- Department of Immunology, Army Medical University, Chongqing, China
| | - Li Meng
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Quan-You Zheng
- Department of Urology, 958 Hospital, Southwest Hospital, Army Medical University, Chongqing, China
| | - You Li
- Department of intense care, Daping Hospital, Army Medical University, Chongqing, China
| | - Gui-Lian Xu
- Department of Immunology, Army Medical University, Chongqing, China
| | - Ke-Qin Zhang
- Urinary Nephropathy Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kan-Fu Peng
- Department of Nephrology, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
11
|
Skeate JG, Otsmaa ME, Prins R, Fernandez DJ, Da Silva DM, Kast WM. TNFSF14: LIGHTing the Way for Effective Cancer Immunotherapy. Front Immunol 2020; 11:922. [PMID: 32499782 PMCID: PMC7243824 DOI: 10.3389/fimmu.2020.00922] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022] Open
Abstract
Tumor necrosis factor superfamily member 14 (LIGHT) has been in pre-clinical development for over a decade and shows promise as a modality of enhancing treatment approaches in the field of cancer immunotherapy. To date, LIGHT has been used to combat cancer in multiple tumor models where it can be combined with other immunotherapy modalities to clear established solid tumors as well as treat metastatic events. When LIGHT molecules are delivered to or expressed within tumors they cause significant changes in the tumor microenvironment that are primarily driven through vascular normalization and generation of tertiary lymphoid structures. These changes can synergize with methods that induce or support anti-tumor immune responses, such as checkpoint inhibitors and/or tumor vaccines, to greatly improve immunotherapeutic strategies against cancer. While investigators have utilized multiple vectors to LIGHT-up tumor tissues, there are still improvements needed and components to be found within a human tumor microenvironment that may impede translational efforts. This review addresses the current state of this field.
Collapse
Affiliation(s)
- Joseph G Skeate
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Mikk E Otsmaa
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ruben Prins
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Daniel J Fernandez
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Diane M Da Silva
- Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - W Martin Kast
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
12
|
Cao ZH, Wu Z, Hu C, Zhang M, Wang WZ, Hu XB. Endoplasmic reticulum stress and destruction of pancreatic β cells in type 1 diabetes. Chin Med J (Engl) 2020; 133:68-73. [PMID: 31923106 PMCID: PMC7028193 DOI: 10.1097/cm9.0000000000000583] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes (T1D) results from dysfunction of pancreatic islets β cells. Recent studies supported that endoplasmic reticulum (ER) stress takes an important role in pancreatic β cell excessive loss, resulting in T1D. Here, we aimed to review the relationship between ER stress and T1D. Additionally, we also reviewed the potential mechanisms underlying ER stress mediated T1D. Studies have shown that severe ER stress is directly involved in the pancreatic β cells destruction and pathogenesis of T1D. ER stress plays a key part in pancreatic β cells and T1D, which will help in developing new effective therapeutics for T1D.
Collapse
Affiliation(s)
- Zhao-Hui Cao
- Department of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | | | | | | | | | | |
Collapse
|
13
|
Lee H, Lim Y. Gamma-tocopherol ameliorates hyperglycemia-induced hepatic inflammation associated with NLRP3 inflammasome in alloxan-induced diabetic mice. Nutr Res Pract 2019; 13:377-383. [PMID: 31583056 PMCID: PMC6760984 DOI: 10.4162/nrp.2019.13.5.377] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/27/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/OBJECTIVES Hyperglycemia-induced hepatic damage has been recognized as one of the major cause of complications in diabetes. Hepatic complications are associated with inflammation and oxidative stress in diabetes. In this study, we investigated the hypothesis that gamma-tocopherol (GT) supplementation ameliorates NLRP3 inflammasome associated hepatic inflammation in diabetes. MATERIALS/METHODS Diabetes was induced by the intraperitoneal injection of alloxan (150 mg/kg. BW) in ICR mice. All mice were fed with a control diet (AIN-76A). After diabetes was induced (fasting glucose level ≥ 250 mg/dL), the mice were treated with tocopherol-stripped corn oil or GT-supplemented (35 mg/kg) corn oil, respectively, by gavage for 2 weeks. RESULTS GT supplementation reduced fasting blood glucose levels in diabetic mice relative to non-treated diabetic mice. Moreover, GT supplementation ameliorated hyperglycemia-induced hepatic damage by regulation of NOD-like receptor protein 3 (NLRP3)-inflammasome associated inflammation represented by NLRP3, apoptosis-associated speck-like protein containing a caspase-recruitment domain, caspase-1, nuclear factor-κB pathway as well as oxidative stress demonstrated by nuclear factor erythroid 2-related factor 2, NAD(P)H dehydrogenase quinone 1, catalase and glutathione-dependent peroxidase in diabetic mice. CONCLUSION The findings suggested that GT supplementation ameliorated hepatic damage by attenuating inflammation and oxidative stress in alloxan-induced diabetic mice. Taken together, GT could be a beneficial nutrient that can ameliorate inflammatory responses associated with NLRP3 inflammasome in hyperglycemia-induced hepatic damage.
Collapse
Affiliation(s)
- Heaji Lee
- Department of Food and Nutrition, Kyung Hee University, 26 Kyunghee-Daero, Dongdaemun-Gu, Seoul 02447, Republic of Korea
| | - Yunsook Lim
- Department of Food and Nutrition, Kyung Hee University, 26 Kyunghee-Daero, Dongdaemun-Gu, Seoul 02447, Republic of Korea
| |
Collapse
|
14
|
Ran MX, Li Y, Zhang Y, Liang K, Ren YN, Zhang M, Zhou GB, Zhou YM, Wu K, Wang CD, Huang Y, Luo B, Qazi IH, Zhang HM, Zeng CJ. Transcriptome Sequencing Reveals the Differentially Expressed lncRNAs and mRNAs Involved in Cryoinjuries in Frozen-Thawed Giant Panda ( Ailuropoda melanoleuca) Sperm. Int J Mol Sci 2018; 19:ijms19103066. [PMID: 30297640 PMCID: PMC6212861 DOI: 10.3390/ijms19103066] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 02/06/2023] Open
Abstract
Sperm cryopreservation and artificial insemination are important methods for giant panda breeding and preservation of extant genetic diversity. Lower conception rates limit the use of artificial insemination with frozen-thawed giant panda sperm, due to the lack of understanding of the cryodamaging or cryoinjuring mechanisms in cryopreservation. Long non-coding RNAs (lncRNAs) are involved in regulating spermatogenesis. However, their roles during cryopreservation remain largely unexplored. Therefore, this study aimed to identify differentially expressed lncRNAs and mRNAs associated with cryodamage or freeze tolerance in frozen-thawed sperm through high throughput sequencing. A total of 61.05 Gb clean reads and 22,774 lncRNA transcripts were obtained. From the sequencing results, 1477 significantly up-regulated and 1,396 significantly down-regulated lncRNA transcripts from fresh and frozen-thawed sperm of giant panda were identified. GO and KEGG showed that the significantly dysregulated lncRNAs and mRNAs were mainly involved in regulating responses to cold stress and apoptosis, such as the integral component of membrane, calcium transport, and various signaling pathways including PI3K-Akt, p53 and cAMP. Our work is the first systematic profiling of lncRNA and mRNA in fresh and frozen-thawed giant panda sperm, and provides valuableinsights into the potential mechanism of cryodamage in sperm.
Collapse
Affiliation(s)
- Ming-Xia Ran
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Yuan Li
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Yan Zhang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Kai Liang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Ying-Nan Ren
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Ming Zhang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Guang-Bin Zhou
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Ying-Min Zhou
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Kai Wu
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Cheng-Dong Wang
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Yan Huang
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Bo Luo
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Izhar Hyder Qazi
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Department of Veterinary Anatomy & Histology, Faculty of Bio-Sciences, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand 67210, Pakistan.
| | - He-Min Zhang
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Chang-Jun Zeng
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| |
Collapse
|
15
|
Kim KH, Choi H, Kim HJ, Lee TR. TNFSF14 inhibits melanogenesis via NF-kB signaling in melanocytes. Cytokine 2018; 110:126-130. [PMID: 29730385 DOI: 10.1016/j.cyto.2018.04.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/23/2018] [Accepted: 04/28/2018] [Indexed: 01/07/2023]
Abstract
Melanin synthesis in melanocytes is affected by various cytokines. Here, we reported for the first time that tumor necrosis factor superfamily member 14 (TNFSF14) inhibits melanogenesis in the primary culture of human epidermal melanocytes. TNFSF14 is known to bind to its receptors herpes virus entry mediator (HVEM) and lymphotoxin β receptor (LTβR) for signal transduction, but TNFSF14-induced hypopigmentation was independent of HVEM and LTβR in melanocytes. To explore signaling in melanocytes treated with TNFSF14, we performed RNA-seq and found that nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) signaling is activated by TNFSF14. Further, we observed that inhibition of NF-kB effectively blocks the hypopigmentation induced by TNFSF14. We conclude that TNFSF14 inhibits melanogenesis in melanocytes via NF-κB signaling and could be applied in the treatment of cutaneous pigment disorders.
Collapse
Affiliation(s)
- Kyu-Han Kim
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do 446-729, Republic of Korea.
| | - Hyunjung Choi
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do 446-729, Republic of Korea
| | - Hyoung-June Kim
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do 446-729, Republic of Korea
| | - Tae Ryong Lee
- Basic Research & Innovation Division, R&D Unit, AmorePacific Corporation, Yongin-si, Gyeonggi-do 446-729, Republic of Korea.
| |
Collapse
|
16
|
Zhang Y, Yuan Y, Wu H, Xie Z, Wu Y, Song X, Wang J, Shu W, Xu J, Liu B, Wan L, Yan Y, Ding X, Shi X, Pan Y, Li X, Yang J, Zhao X, Wang L. Effect of verbascoside on apoptosis and metastasis in human oral squamous cell carcinoma. Int J Cancer 2018. [DOI: 10.1002/ijc.31378] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yaqin Zhang
- Department of Biochemical Molecular, School of Basic Medical Sciences; Nanjing Medical University; Nanjing China
| | - Yi Yuan
- Jiangsu Key Laboratory of Oral Diseases; Nanjing Medical University; Nanjing China
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Heming Wu
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Zhuoying Xie
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering; Southeast University; Nanjing China
| | - Yunong Wu
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Xiaomeng Song
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Jingjing Wang
- Department of Biochemical Molecular, School of Basic Medical Sciences; Nanjing Medical University; Nanjing China
| | - Wei Shu
- Stomatology Department; Jiangsu Provincial Hospital of Traditional Chinese Medicine
| | - Junyong Xu
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Bin Liu
- Department of Biomedical Engineering, School of Basic Medical Sciences; Nanjing Medical University; Nanjing China
| | - Linzhong Wan
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Yanan Yan
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Xu Ding
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Xinghui Shi
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Yongchu Pan
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Xiaokang Li
- Jiangsu Key Laboratory of Oral Diseases; Nanjing Medical University; Nanjing China
- Reproductive Medicine Center; The University of Hong Kong-Shenzhen Hospital; China
| | - Jianrong Yang
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| | - Xiaohui Zhao
- Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resources; Northwest Institute of Plateau Biology, Chinese Academy of Sciences; Xining China
| | - Lin Wang
- Jiangsu Key Laboratory of Oral Diseases; Nanjing Medical University; Nanjing China
- Institute of Stomatology; Nanjing Medical University; Nanjing China
| |
Collapse
|
17
|
Qian C, Chen X, Qi Y, Zhong S, Gao X, Zheng W, Mao Z, Yao J. Sporamin induces apoptosis and inhibits NF-κB activation in human pancreatic cancer cells. Tumour Biol 2017; 39:1010428317706917. [PMID: 28714369 DOI: 10.1177/1010428317706917] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Sporamin, a Kunitz-type trypsin inhibitor (TI) from sweet potato tuberous roots, has demonstrated anti-tumor activity through poorly defined mechanisms. Furthermore, the effects of sporamin on pancreatic cancer have not been explored. Herein, we studied the effects of sporamin on two human pancreatic cancer cell lines, PANC-1 and BxPC-3. Sporamin significantly inhibited the cell viability and proliferation activity and induced apoptosis in PANC-1 and BxPC-3 cells. Consistently, in sporamin-treated PANC-1 and BxPC-3 cells, the anti-apoptotic proteins Bcl-2 and Bcl-XL were downregulated and the pro-apoptotic protein Bax was upregulated. Moreover, nuclear factor kappa B activation and IκBα phosphorylation were inhibited, and total IκBα expression was increased in sporamin-treated PANC-1 and BxPC-3 cells. Thus, our results suggest that the anti-tumor effects of sporamin in pancreatic cancer cells might result partly from induction of apoptosis by downregulating nuclear factor kappa B pathway.
Collapse
Affiliation(s)
- Cuijuan Qian
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang, P.R. China
| | - Xiaoying Chen
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang, P.R. China
| | - Yongxiao Qi
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang, P.R. China
| | - Sheng Zhong
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang, P.R. China
| | - Xinyan Gao
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang, P.R. China
| | - Wenjun Zheng
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang, P.R. China
| | - Zhixiang Mao
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang, P.R. China
| | - Jun Yao
- Institute of Tumor, School of Medicine, Taizhou University, Taizhou, Zhejiang, P.R. China
| |
Collapse
|
18
|
Pyrrolidine dithiocarbamate ameliorates endothelial dysfunction in thoracic aorta of diabetic rats by preserving vascular DDAH activity. PLoS One 2017; 12:e0179908. [PMID: 28715444 PMCID: PMC5513417 DOI: 10.1371/journal.pone.0179908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/07/2017] [Indexed: 01/27/2023] Open
Abstract
Objective Endothelial dysfunction plays a pivotal role in the development of diabetic cardiovascular complications. Accumulation of endogenous nitric oxide synthase (NOS) inhibitor asymmetric dimethylarginine (ADMA) and inhibition of dimethylarginine dimethylaminohydrolase (DDAH) activity have been involved in diabetic endothelial dysfunction. This study was to investigate the effect of pyrrolidine dithiocarbamate (PDTC) on impairment of endothelium-dependent vasodilatation in diabetic rats and its potential mechanism. Methods Diabetic rats were induced by a single intraperitoneal injection of streptozotocin (60mg/kg), and PDTC (10mg/kg) was given in drinking water for 8 weeks. Blood glucose and serum ADMA concentrations were measured in experimental rats. Recombinant adenovirus encoding human DDAH2 gene were constructed and ex vivo transferred to isolated rat aortas. The maximal relaxation (Emax) and half maximal effective concentration (EC50) of aortic rings response to accumulative concentrations of acetylcholine and vascular DDAH activity were examined before and after gene transfection. Results Diabetic rats displayed significant elevations of blood glucose and serum ADMA levels compared to control group (P<0.01). Vascular DDAH activity and endothelium-dependent relaxation of aortas were inhibited, as expressed by the decreased Emax and increased EC50 in diabetic rats compared to control rats (P<0.01). Treatment with PDTC not only decreased blood glucose and serum ADMA concentration (P<0.01) but also restored vascular DDAH activity and endothelium-dependent relaxation, evidenced by the higher Emax and lower EC50 in PDTC-treated diabetic rats compared to untreated diabetic rats (P<0.01). Similar restoration of Emax, EC50 and DDAH activity were observed in diabetic aortas after DDAH2-gene transfection. Conclusions These results indicate that PDTC could ameliorate impairment of endothelium-dependent relaxation in diabetic rats. The underlying mechanisms might be related to preservation of vascular DDAH activity and consequent reduction of endogenous ADMA in endothelium via its antioxidant action. This study highlights the therapeutic potential of PDTC in impaired vasodilation and provides a new strategy for treatment of diabetic cardiovascular complications.
Collapse
|